The Role of Bioactive Lipids in Cancer, Inflammation and Related Diseases. Advances in Experimental Medicine and Biology
The Role of Bioactive Lipids in Cancer, Inflammation and Related Diseases. Advances in Experimental Medicine and Biology
The Role of Bioactive Lipids in Cancer, Inflammation and Related Diseases. Advances in Experimental Medicine and Biology
Kenneth V. Honn
Darryl C. Zeldin Editors
Volume 1161
Editorial Board
IRUN R. COHEN, The Weizmann Institute of Science, Rehovot, Israel
ABEL LAJTHA, N.S. Kline Institute for Psychiatric Research,
Orangeburg, NY, USA
JOHN D. LAMBRIS, University of Pennsylvania, Philadelphia, PA, USA
RODOLFO PAOLETTI, University of Milan, Milan, Italy
NIMA REZAEI, Children’s Medical Center Hospital, Tehran University
of Medical Sciences, Tehran, Iran
More information about this series at http://www.springer.com/series/5584
Kenneth V. Honn • Darryl C. Zeldin
Editors
This Springer imprint is published by the registered company Springer Nature Switzerland AG.
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Contents
1 Introduction................................................................................... 1
Kenneth V. Honn and Darryl C. Zeldin
2 Understanding the Role of Pro-resolving Lipid Mediators
in Infectious Keratitis................................................................... 3
Elizabeth A. Berger
3 Immunoresolvent Resolvin D1 Maintains the Health
of the Ocular Surface.................................................................... 13
Darlene A. Dartt, Robin R. Hodges, and Charles N. Serhan
4 The Evolving Role of Specialized Pro-resolving Mediators
in Modulating Neuroinflammation in Perioperative
Neurocognitive Disorders............................................................. 27
Ting Yang and Niccolò Terrando
5 Relationship Between Specialized Pro-resolving
Mediators and Inflammatory Markers
in Chronic Cardiac Disorders...................................................... 37
M. Brianza-Padilla and R. Bojalil
6 Specialized Pro-resolving Mediators Directs Cardiac
Healing and Repair with Activation of Inflammation
and Resolution Program in Heart Failure.................................. 45
Ganesh V. Halade and Bochra Tourki
7 Novel n-3 Docosapentaneoic Acid-Derived
Pro-resolving Mediators Are Vasculoprotective
and Mediate the Actions of Statins in Controlling
Inflammation................................................................................. 65
Jesmond Dalli, Kimberly Pistorius, and Mary E. Walker
8 Aspects of Prostaglandin Glycerol Ester Biology....................... 77
Philip J. Kingsley, Carol A. Rouzer, Amanda J. Morgan,
Sachin Patel, and Lawrence J. Marnett
9 Targeting the COX/mPGES-1/PGE2 Pathway
in Neuroblastoma.......................................................................... 89
Karin Larsson, Anna Kock, Per Kogner,
and Per-Johan Jakobsson
v
vi Contents
Index....................................................................................................... 255
About the Editors
vii
viii About the Editors
In addition to this introduction, this book con- presented a lecture on “Decoding New Lipid
tains 18 outstanding chapters based on compre- Mediators and Mechanisms of Resolution of
hensive and detailed reviews of timely topics Inflammation, Infections and Tissue
presented at the 15th International Conference on Regeneration.” Drs. Bruce D. Hammock, Jorge
Bioactive Lipids in Cancer, Inflammation and H. Capdevila and Darryl C. Zeldin shared the
Related Diseases held in Puerto Vallarta, Mexico Outstanding Achievement Award for their work
on October 22–25, 2017. on the P450 epoxygenase pathway of arachidonic
Held biennially since 1989, this ongoing con- acid metabolism and delivered lectures on the
ference series provides a scientific program that role of fatty acids in neuropathic pain, cardiovas-
is comprehensive and structured to highlight cular disease and lung inflammation. There were
research at the cutting edge of science on the role three Eicosanoid Research Foundation Young
of lipid mediators in various physiological and Investigator awards presented to Drs. Karin
pathological processes in a wide range of thera- Larsson (cancer), Jesmond Dalli (inflammation)
peutic areas. Over 315 basic researchers and cli- and Scott Hansen (structure-function). Dr. Shu
nicians representing 32 countries attended this Xu from the University of Toledo was presented
most recent meeting. Dr. Jeffrey M. Drazen, with the Santosh Nigam Memorial Outstanding
Editor-in-Chief of the New England Journal of Young Scientist award for his presentation on
Medicine, received the Exceptional Contributions “The Structure of the Catalytic Domain of
to Human Physiology and Translational Medicine 12-Lipoxygenase.” There were 50 lectures orga-
Award and presented an opening lecture on “Data nized into 3 Plenary Sessions, 14 scientific ses-
Sharing in Clinical Trials.” Dr. Charles N. Serhan sions and 140 poster presentations.
received the Lifetime Achievement Award and The specialized pro-resolving lipid mediators
or SPMs (e.g., resolvins, lipoxins, maresins) have
received wide-spread attention as to their role in
K. V. Honn (*) inflammatory processes. Reviews presented in
Departments of Pathology, Oncology and Chemistry, this book detail recent advances in understanding
Wayne State University, Detroit, MI, USA
e-mail: [email protected] the role of SPMs in inflammatory conditions of
the eye, brain and cardiovascular system. Dr.
D. C. Zeldin (*)
Division of Intramural Research, National Institutes Elizabeth Berger (Chap. 2) summarized recent
of Health/National Institute of Environmental Health literature on infectious keratitis (bacterial, viral,
Sciences, Research Triangle Park, NC, USA fungal) and endophthalmitis with an emphasis on
e-mail: [email protected]
the host response and the role of SPMs in resolu- inflammation and colorectal cancer. Dr. Klaus
tion of the eye inflammation. Drs. Darlene Dartt, Van Leyen and co-workers (Chap. 12) summa-
Robin R. Hodges, and Charles N. Serhan (Chap. rized the biological relevance of lipoxygenase-
3) provided a comprehensive summary of the derived eicosanoids in ischemic stroke and
role of resolvin D1 (RvD1), its cognate receptor neuronal cell death in the brain.
(ALX/FPR2 or GPR32) and downstream signal- Sphingolipids are a unique class of bioactive
ing pathways (PLC, PLD, PLA2, EGFR, and lipids with significant chemical diversity. Recent
ERK) in preserving ocular surface homeostasis advances in the field of sphingolipids and their
and regulating conjunctival goblet cell mucin role in the eye, brain, intestine and immune sys-
secretion. Dr. Niccolo Terrando (Chap. 4) tem are discussed in this book. Dr. Yasuyuki
reported on the role of SPMs in the brain with an Kihara (Chap. 13) provided a review of eico-
emphasis on postoperative neuroinflammation sanoid and other bioactive lipid signaling involve-
and cognitive function. Dr. Rafael Bojalil ment in the neuroinflammatory mechanisms
(Chap. 5) reviewed the role of pro-resolving underlying multiple sclerosis. Drs. Koushik
mediators in cardiovascular diseases, notably in Mondal and Nawajes Mandal (Chap. 14) reported
regulating inflammation. Dr. Ganesh Halade on the role of bioactive sphingolipids including
(Chap. 6) summarized recent literature on the ceramide and sphingosine in ocular inflammation
role of SPMs in resolution of cardiac inflamma- and eye diseases such as uveitis, diabetic retinop-
tion, tissue repair and remodeling in heart failure athy and glaucoma. Drs. Sabrin Albeituni and
models. Dr. Jesmond Dalli (Chap. 7) reported on Johnny Stiban (Chap. 15) reported on the role of
the biological actions of resolvins in the cardio- ceramides and other sphingolipids in immune
vascular system and their role in mediating the cell function and inflammation in cancer, multi-
actions of statins. ple sclerosis and inflammatory bowel disease.
The book contains two chapters that overview The endocannabinoids and their receptors
recent advances in cyclooxygenase-derived eico- affect both central nervous system and peripheral
sanoids in the brain. Dr. Philip Kingsley (Chap. 8) processes. There are two chapters on recent
reported on the identification and role of prosta- advances in endocannabinoids in the brain. Dr.
glandin glycerol esters (PG-Gs) in the brain, Heather Bradshaw and colleagues (Chap. 16)
detailing their production, metabolism and bioac- reported on the role of endogenous cannabinoids
tivity. Dr. Karin Larsson (Chap. 9) summarized and their receptors in the pathogenesis of trau-
recent data on the role of prostaglandin E2 matic brain injury. Dr. Aditi Das (Chap. 17) sum-
(PGE2) in neuroblastoma, in particular its actions marized recent information on the
in regulating angiogenesis, immunosuppression anti-inflammatory and vasoactive role of omega-3
in the tumor microenvironment, and tumor endocannabinoid epoxide metabolites in the
growth. There are three chapters that provide an brain. In addition, there is a chapter by Dr. Andres
overview on the biological relevance of cyto- Trostchansky (Chap. 18) on the relationship of
chrome P450 (CYP) and lipoxygenase-derived lipid metabolism and lipid derived metabolites
eicosanoids. Dr. Pei-an Shih (Chap. 10) reported with the onset and progression of amylotrophic
that CYP-derived eicosanoids are prognostic bio- latereral sclerosis in humans. This book is capped
markers for certain neuropsychiatric disorders off with a concise, scholarly review by Drs. Carlo
such as depression. Dr. Guodong Zhang and co- Barnaba and Ilce G. Medina-Meza on the role of
workers (Chap. 11) discussed recent studies on flavonoids in the reduction of inflammation and
the roles of CYP-derived eicosanoids and soluble lipid oxidation in the pathogenesis of cardiovas-
epoxide hydrolase in the pathogenesis of colonic cular disease (Chap. 19).
Understanding the Role of
Pro-resolving Lipid Mediators 2
in Infectious Keratitis
Elizabeth A. Berger
inflammatory response, persistence and chronic PGD2) initiate lipid mediator class switching
activation of macrophages, neutrophils/PMN and [11]. Biosynthesized during the active phase of
T cells contribute to disease pathogenesis through resolution, SPMs play a pivotal role as
sustained production of inflammatory mediators, endogenous agonists stimulating the resolution
proteolytic enzymes and resultant tissue necrosis. of inflammation – a key feature of a healthy,
Similarly, cytokines and chemokines such as productive inflammatory response [12].
IL-1α/β, TNF-α, IFN-γ and MIP-2 promote anti- Proresolving actions include limiting PMN
microbial effector mechanisms, activate phago- infiltration, regulating the cytokine profile [13,
cytic killing and recruit additional inflammatory 14], reducing pain [15], and efferocytosis [16,
cells. However, the continued production of such 17]. SPMs best associated with the eye are
pro-inflammatory cytokines/chemokines contrib- derived from two major polyunsaturated fatty
utes to the destruction of the corneal microenvi- acids; arachidonic acid (AA) and docosahexaenoic
ronment. In addition to an upregulation of acid (DHA) and are targeted by lipoxygenase
cytokines, lipid mediators are also released, (LOX) enzymes, which comprise the main path-
including prostaglandins, thromboxanes and ways for SPM biosynthesis (Fig. 2.1). Studies
platelet-activating factor. It is well-known that have demonstrated LOX activity in the cornea
these arachidonic acid-derived mediators play with 15-LOX appearing to be largely protective
key roles in the initiation of inflammation, as a key enzyme for the generation of SPMs [12,
including the recruitment of leukocytes 18–20] . Alox15 is the most abundant lipoxygen-
(particularly neutrophils). Although necessary, it ase in the healthy murine cornea [21–23]. Gronert
is imperative that this biological cascade is both et al. have established the presence of functional
inhibited and dissolved. These initial pro- 5-LOX and 12/15-LOX in mouse corneal epithe-
inflammatory pathways are balanced by lium and corneas [19]. Furthermore, both healthy
proresolving lipid signals known as specialized and inflamed corneas express 12/15- LOX as
pro-resolving lipid mediators (SPMs). In fact, it detected from epithelial cells and infiltrated neu-
has been shown that prostaglandins (PGE2 and trophils [19]. AA-derived lipoxin A4 (LXA4) can
be transcellularly biosynthesized during leuko- events. Yet, other studies have indicated delayed
cyte interactions with mucosal cells, such as epithelial healing and worsened disease outcome
PMN interactions with epithelial cells [24]. [35–37]. Additionally, a comprehensive review
Studies have revealed a protective role for LXA4, evaluating the effects of corticosteroids from
whereby it has been shown to accelerate corneal 1950 to 2000 concluded that the role of
re-epithelization and limit neovascularization corticosteroids in the adjunctive treatment of
using a thermal injury model [25]. In addition to infectious keratitis remains unsubstantiated [38].
AA-derived lipoxins, DHA is targeted by phos- Further, a review by McGhee et al. states that
pholipase A2 and subsequently LOX and COX-2 corticosteroid-induced ocular morbidity includes:
produce di- and trihydroxylated DHA derivatives ocular hypertension, glaucoma, cataract
including D series resolvins, protectins and formation, tear film instability, and crystalline
maresins [26]. These SPMs are particularly rele- keratopathy [39]. To this end, this review will
vant to the eye as studies have shown that RvD1 highlight the compelling evidence for the
reduces the inflammatory response during endo- development of pro-resolving molecules as an
toxin-induced uveitis [25], allergic eye disease adjuvant therapy for the treatment of infectious
[27], and dry eye [28]. keratitis – with focus on bacterial, viral and
fungal infections.
This collective work illuminates host factors that Bacteria remain the most common cause of infec-
exacerbate the progression of keratitis, indicating tious keratitis with studies indicating that bacte-
that the pathogenicity of disease is a combination rial keratitis comprises approximately 90% of
of microbe-induced damage and an unresolved infectious keratitis cases. Epidemiological stud-
host response – the latter of which may be due to ies have revealed that between 54–94% of all cor-
ocular immune privilege. Regardless, a number neal infections are due to bacteria. Pseudomonas
of adjunctive treatments have been considered, aeruginosa and Staphylococcus aureus have
including cross-linking, anti-collagenases, anti- been reported as the more prevalent bacteria
inflammatories, and corticosteroids. Collagen associated with infectious keratitis [40–42].
cross-linking was found to improve corneal Staphylococcus aureus as the most common ocu-
re-epithelization and resistance to enzymatic lar isolate [43]; while Pseudomonas aeruginosa
degradation, reduce corneal melting, and exert is the most frequently isolated of Gram negative
bactericidal effects [29–31]. However, ocular infections [22]. Moreover, Pseudomonas
complications include endothelial cell loss and aeruginosa ulcers have been shown to result in a
reactivation of HSV infections, thus warranting more progressive disease with large infiltrate and
further investigation. Non-steroidal anti-scarring [43, 56] and not benefit from the addi-
inflammatory treatments have been investigated tion of corticosteroids [36]. Current treatment of
in an attempt to prevent excessive inflammation; microbial keratitis consists primarily of fourth
but shown to exacerbate the disease response in generation ophthalmic fluoroquinolones (moxi-
the absence of concomitant antibiotic therapy or floxcin and gatifloxacin), which are effective
exhibit toxicity issues complicated by corneal against both Gram-positive and Gram-negative
melting [32–34]. Indecision continues to exist bacteria. This class of antibiotics also exhibits
regarding the benefits and risks associated with better mutant prevention characteristics and pen-
use of corticosteroids. The Steroids for Corneal etrates well into ocular tissues. However, antibi-
Ulcers Trial (SCUT) provides evidence for otic-resistance is of major concern; in the U.S.,
improved visual acuity in bacterial keratitis roughly 80% of methicillin-resistant S. aureus
patients with no reported increase in adverse ocular isolates have been reported as resistant to
6 E. A. Berger
fluoroquinolones [44–46]. Multi-drug resistance the importance of SPM circuit activation in the
rates for Pseudomonas are on the rise, as well resolution of inflammation following corneal
[47, 48]. infection. Further, it highlights the significant
Work using a mouse model of Pseudomonas- potential of combination treatments – where
induced keratitis has revealed the importance of antibiotics alone cannot activate the resolution
SPM circuits in the resolution of inflammation arm of inflammation; yet without the antibiotic,
[23]. Lipid mediator profiles were characterized Tβ4 (and other activators of SPM pathways) may
in susceptible (corneal thinning, perforation) and not be able to overcome the bacterial burden and
resistant (resolution) phenotypes and indicated subsequently move toward active resolution.
that activation of SPM pathways are requisite for Work carried out by the Pearlman group
a balanced inflammatory response characterized investigated the role of RvE1 in the corneal
by the resolution of disease. Furthermore, this inflammatory response induced by LPS and
study indicated a critical role for 15-LOX in antibiotic-killed Pseudomonas aeruginosa and
influencing inflammatory cell function. Without Staphylococcus aureus [56]. Cytokine production
15-LOX enzymatic activity, essential innate was significantly reduced in human corneal
inflammatory functions (phagocytosis, epithelial cells and neutrophils, as well as mouse
efferocytosis) were impaired, leading to an cornea and macrophages. In addition, a decrease
exacerbated disease response. This supports in neutrophil infiltrate and reduced corneal haze
previous work that showed 15-LOX−/− mice was observed after RvE1 treatment following
resulted in impaired wound healing and amplified bacteria- and LPS-induced corneal inflammation.
inflammatory angiogenesis [19, 49, 50]. In
addition, potential experimental treatments that
have been shown to improve disease response to 2.5 Viral Keratitis
Pseudomonas-induced keratitis appear to have a
common theme – the activation of SPM circuitry. Herpes simplex virus (HSV) keratitis affects an
Vasoactive intestinal peptide or VIP has been estimated 500,000 people in the US and an
demonstrated to exert its immunoregulatory estimated 1.5 million globally per annum [57,
effects, in part, through upregulation of 58]. Though it begins as a subclinical infection of
lipoxygenase enzymes in corneas of the cornea, HSV establishes latency within the
Pseudomonas-infected mice [51, 52]. Additional trigeminal ganglia and when reactivated, it causes
work in the mouse model has revealed that VIP recurrent infections that result in decreased
increases RvD1 and modulates the ALX/FPR2 corneal sensation, neurotrophic keratitis, stromal
receptor axis toward inflammation resolution opacity, scarring, and blindness. In fact, HSV
[52]. Ligands for this receptor include both keratitis is the predominant cause of unilateral
LXA4 and RvD1 [53]; the latter of which has infectious corneal blindness in most developed
been shown to reduce corneal neovascularization nations [59]. Though rare, bilateral infection
[54]. Evidence also indicates that thymosin does occur in approximately 1.3–12% of patients
beta-4 (Tβ4), another therapeutic molecule, may [60]. Primary HSV infections are typically
carry out its profound effects through activation overlooked in the clinic as adenoviral
of SPM circuits [55]. Adjunct Tβ4 treatment with conjunctivitis. Epithelial infection is typically
antibiotic resulted in increased 12- and 12/15- caused by actively replicating virus and presents
LOX mRNA levels, while COX-2 and 5-LOX itself as the classic herpetic corneal lesion – a
were decreased after infection [55]. This is dendritic ulcer – with minimal stromal
further supported by increased RvD1 and its inflammation [61]. Once resolved, a dendritic
receptor, ALX/FPR2. In contrast, pro-scar or ghost dendrite may persist in the
inflammatory lipid mediator pathways were superficial stroma. A more progressive geographic
affected, as evidenced by lower levels of LTB4 ulcer (also caused by live replicating virus) can
and its receptor, BLT1. This work underscores occur in immunocompromised patients and
2 Understanding the Role of Pro-resolving Lipid Mediators in Infectious Keratitis 7
first choice of treatment; though amphotericin B mediators in the treatment of this disease.
is preferred for Aspergillus and Candida keratitis. Intravitreal injections of RvD1 was shown to
Limitations exist for both however; natamycin attenuate the progression of disease while
exhibits poor penetration into the corneal stroma preserving retinal function [85]. This was
and topical amphotericin B has toxicity issues indicated by reduced levels of proinflammatory
[78]. Although voriconazole, a newer-generation mediators, reduced bacterial burden, fewer
triazole, demonstrates excellent ocular penetra- infiltrated PMN, and induction of antimicrobial
tion [79] and high susceptibility among common molecules. Further, it was suggested that this
fungal isolates [80], natamycin remains superior effect may involve TLR signaling pathways as
to voriconazole in the topical treatment of fungal TLR2 deficient mice were not protected by RvD1
keratitis [36, 81]. However, there are no reports treatment. Similar to fungal keratitis though,
examining the effects of SPMs in the treatment of therapeutic potential of pro-resolving mediators
this disease either as a monotherapy or as an have yet to be examined in fungal endopthalmitis.
adjunct to available antifungals. In vitro antifun-
gal susceptibility testing and in vivo fungal kerati-
tis models could provide valuable insight into the 2.8 Conclusion
potential use of SPMs for fungal keratitis given
the diversity of fungal aetiology and emergence of Although currently available antibiotics, antivi-
drug resistance. rals, and antifungals are efficacious in success-
fully controlling most of the pathogens implicated
in infectious keratitis, clinical outcomes remain
2.7 Endophthalmitis poor. A common limitation among these thera-
pies is evident – a sustained host response that
In rare cases, infectious keratitis can extend from compromises corneal integrity. The cornea dem-
the cornea into the vitreous and/or aqueous onstrates a deficiency in switching from active
humor and is termed endophthalmitis. Though inflammation to active resolution – perhaps due
uncommon, it is among the most devastating eye to ocular immune privilege – resulting in struc-
infections that bears the worst possible outcome – tural alterations and loss of tissue. There exists a
irreversible blindness that can occur within even clear need for treatments that modify the immune
hours of symptom onset [82]. Incidence of response toward resolution and activate tissue
disease ranges from 0.03–1.3% with cataract restoration. In fact, a review on the clinical man-
surgery to as high as 30–40% following open agement of infectious keratitis suggested that the
globe injuries [83]. The most prevalent pathogens development of future adjuvant therapies with
related to endogenous endophthalmitis are the greatest potential are likely to be multidimen-
bacteria (Staphylococcus sp., Streptococcus sp.) sional that are aimed at modifying the immune
or fungi (Candida, Aspergillus). With more than response to infection [36]. The current review
50% of keratitis-related endophthalmitis cases provides evidentiary support for using pro-
due to mold, Fusarium and Aspergillus are the resolving molecules to treat infectious keratitis.
most common etiologies [84]. Moreover, fungal SPMs are being targeted as a beneficial mono-
endophthalmitis is particularly difficult to treat therapy and adjuvant for infectious keratitis. The
due to delayed diagnosis and poor ocular concept of combining SPMs with other drugs
penetration of antifungals compounded by has been demonstrated to be highly effective in
toxicity issues. Despite this, treatment typically resolving inflammation [86]. This class of pro-
consists of intravitreal antibiotics. In addition, resolving lipid mediators holds promise in the
vitrectomy is often carried out due to the rapidity treatment of drug-resistant infections while
and severity of disease progression. avoiding ocular surface toxicity issues that cur-
Initial studies in a S. aureus-induced model of rently impede proper treatment of infectious
endophthalmitis show promise for pro-resolving keratitis. As reviewed, SPMs function to dis-
2 Understanding the Role of Pro-resolving Lipid Mediators in Infectious Keratitis 9
solve inflammation through the regulation of 4. Keay L, Stapleton F, Schein O (2007) Epidemiology
of contact lens-related inflammation and microbial
leukocyte infiltration, activation and function. keratitis: a 20-year perspective. Eye Contact Lens
Further, chemical mediators (cytokines, chemo- 33:346–353. discussion 362–343
kine’s, ROS) that would otherwise contribute to 5. Thylefors B (1992) Epidemiological patterns of
chronic inflammation are influenced by SPMs – ocular trauma. Aust N Z J Ophthalmol 20:95–98
6. Sheng XL, Li HP, Liu QX, Rong WN, Du WZ, Ma
all of which enables the host response to return L, Yan GH, Ma RQ, Zhang JL, Xu HF, Zou WQ, Bi
to homeostasis and is critical to restoration of XJ (2014) Prevalence and associated factors of cor-
corneal function and visual acuity. neal blindness in Ningxia in Northwest China. Int
One aspect of SPM-induced corneal restora- J Ophthalmol 7:557–562
7. Saha S, Banerjee D, Khetan A, Sengupta J (2009)
tion that has yet to be properly examined in the Epidemiological profile of fungal keratitis in urban
infectious keratitis model is corneal nerve regen- population of West Bengal, India. Oman J Ophthalmol
eration. As the most innervated tissue in the body, 2:114–118
the cornea contains predominately sensory nerves 8. Nirmalan PK, Katz J, Tielsch JM, Robin AL,
Thulasiraj RD, Krishnadas R, Ramakrishnan R (2004)
along with sympathetic and parasympathetic. Ocular trauma in a rural south Indian population: the
Extensive work has been carried out investigating Aravind comprehensive eye survey. Ophthalmology
the role of corneal nerves in maintaining a healthy 111:1778–1781
ocular surface, during wound healing and follow- 9. Jones DB (1981) Decision-making in the management
of microbial keratitis. Ophthalmology 88:814–820
ing infection [22, 87–90]. Clinically, damaged 10. Hazlett LD (2004) Corneal response to Pseudomonas
corneal nerves lead to decreased sensitivity, blink aeruginosa infection. Prog Retin Eye Res 23:1–30
reflexes, tear secretion, dry eye, and neurotropic 11. Levy BD, Clish CB, Schmidt B, Gronert K, Serhan
keratitis. The Bazan group has revealed a combi- CN (2001) Lipid mediator class switching dur-
ing acute inflammation: signals in resolution. Nat
nation of DHA and pigment epithelial-derived Immunol 2:612
factor (PEDF) increases NPD1 levels and corneal 12. Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang
nerve regeneration using a lamellar keratectomy N, Gronert K (2000) Novel functional sets of lipid-
injury model in the rabbit [91–93]. Further, they derived mediators with antiinflammatory actions gen-
erated from omega-3 fatty acids via cyclooxygenase
found that epithelial wound healing was acceler- 2-nonsteroidal antiinflammatory drugs and transcel-
ated after nerve damage, as well. Though these lular processing. J Exp Med 192:1197–1204
results indicate that pro-resolving mediators will 13. Serhan CN, Hong S, Gronert K, Colgan SP, Devchand
also influence corneal nerves following infection, PR, Mirick G, Moussignac RL (2002) Resolvins: a
family of bioactive products of omega-3 fatty acid
a more comprehensive assessment is required. transformation circuits initiated by aspirin treatment
Overall, it is anticipated that SPMs will provide a that counter proinflammation signals. J Exp Med
promising avenue of therapy that contributes to 196:1025–1037
the significant advancement in the treatment of 14. Hong S, Gronert K, Devchand PR, Moussignac
RL, Serhan CN (2003) Novel docosatrienes and
infectious keratitis. 17S-resolvins generated from docosahexaenoic acid in
murine brain, human blood, and glial cells. Autacoids
in anti-inflammation. J Biol Chem 278:14677–14687
References 15. Xu ZZ, Zhang L, Liu T, Park JY, Berta T, Yang R,
Serhan CN, Ji RR (2010) Resolvins RvE1 and RvD1
attenuate inflammatory pain via central and peripheral
1. Rattanatam T, Heng WJ, Rapuano CJ, Laibson PR,
actions. Nat Med 16:592–597. 591p following 597
Cohen EJ (2001) Trends in contact lens-related cor-
16. Dalli J, Serhan CN (2012) Specific lipid media-
neal ulcers. Cornea 20:290–294
tor signatures of human phagocytes: microparticles
2. Stapleton F, Naduvilath T, Keay L, Radford C, Dart J,
stimulate macrophage efferocytosis and pro-resolving
Edwards K, Carnt N, Minassian D, Holden B (2017)
mediators. Blood 120:e60–e72
Risk factors and causative organisms in microbial
17. Schwab JM, Chiang N, Arita M, Serhan CN (2007)
keratitis in daily disposable contact lens wear. PLoS
Resolvin E1 and protectin D1 activate inflammation-
One 12:e0181343
resolution programmes. Nature 447:869–874
3. Green M, Apel A, Stapleton F (2008) Risk factors and
18. Wei J, Gronert K (2017) The role of pro-resolving
causative organisms in microbial keratitis. Cornea
lipid mediators in ocular diseases. Mol Asp Med
27:22–27
58:37–43
10 E. A. Berger
19. Gronert K, Maheshwari N, Khan N, Hassan IR, Dunn steroidal anti-inflammatory drugs. Ophthalmology
M, Laniado Schwartzman M (2005) A role for the 108:936–944
mouse 12/15-lipoxygenase pathway in promoting epi- 35. Gritz DC, Kwitko S, Trousdale MD, Gonzalez VH,
thelial wound healing and host defense. J Biol Chem McDonnell PJ (1992) Recurrence of microbial kerati-
280:15267–15278 tis concomitant with antiinflammatory treatment in an
20. Liclican EL, Gronert K (2010) Molecular circuits of animal model. Cornea 11:404–408
resolution in the eye. Scientific World J 10:1029–1047 36. Austin A, Lietman T, Rose-Nussbaumer J (2017)
21. Gronert K (2005) Lipoxins in the eye and their role Update on the management of infectious keratitis.
in wound healing. Prostaglandins Leukot Essent Fatty Ophthalmology 124:1678–1689
Acids 73:221–229 37. Cohen EJ (2009) The case against the use of steroids in
22. Kenchegowda S, Bazan HE (2010) Significance of the treatment of bacterial keratitis. Arch Ophthalmol
lipid mediators in corneal injury and repair. J Lipid 127:103–104
Res 51:879–891 38. Wilhelmus KR (2002) Indecision about corticoste-
23. Carion TW, Greenwood M, Ebrahim AS, Jerome
roids for bacterial keratitis: an evidence-based update.
A, Suvas S, Gronert K, Berger EA (2018) Ophthalmology 109:835–842. quiz 843
Immunoregulatory role of 15-lipoxygenase in 39. McGhee CN, Dean S, Danesh-Meyer H (2002)
the pathogenesis of bacterial keratitis. FASEB Locally administered ocular corticosteroids: benefits
J 32:5026–5038 and risks. Drug Saf 25:33–55
24. Fredman G, Oh SF, Ayilavarapu S, Hasturk H,
40. Orlans H, Hornby S, Bowler I (2011) In vitro anti-
Serhan CN, Van Dyke TE (2011) Impaired phagocy- biotic susceptibility patterns of bacterial keratitis iso-
tosis in localized aggressive periodontitis: rescue by lates in Oxford, UK: a 10-year review. Eye 25:489
Resolvin E1. PLoS One 6:e24422 41. Lichtinger A, Yeung SN, Kim P, Amiran MD, Iovieno
25. Settimio R, Clara DF, Franca F, Francesca S, Michele A, Elbaz U, Ku JY, Wolff R, Rootman DS, Slomovic
D (2012) Resolvin D1 reduces the immunoinflamma- AR (2012) Shifting trends in bacterial keratitis
tory response of the rat eye following uveitis. Mediat in Toronto: an 11-year review. Ophthalmology
Inflamm 2012:318621 119:1785–1790
26. Serhan CN (2014) Pro-resolving lipid mediators are 42. Chang VS, Dhaliwal DK, Raju L, Kowalski RP
leads for resolution physiology. Nature 510:92–101 (2015) Antibiotic resistance in the treatment of
27. Saban DR, Hodges RR, Mathew R, Reyes NJ, Yu C, Staphylococcus aureus keratitis: a 20-year review.
Kaye R, Swift W, Botten N, Serhan CN, Dartt DA Cornea 34:698–703
(2019) Resolvin D1 treatment on goblet cell mucin 43. Teweldemedhin M, Gebreyesus H, Atsbaha AH,
and immune responses in the chronic allergic eye dis- Asgedom SW, Saravanan M (2017) Bacterial pro-
ease (AED) model. Mucosal Immunol 12:145–153 file of ocular infections: a systematic review. BMC
28. Dartt DA, Hodges RR, Li D, Shatos MA, Lashkari K, Ophthalmol 17:212
Serhan CN (2011) Conjunctival goblet cell secretion 44. Asbell PA, Colby KA, Deng S, McDonnell P, Meisler
stimulated by leukotrienes is reduced by resolvins DM, Raizman MB, Sheppard JD Jr, Sahm DF (2008)
D1 and E1 to promote resolution of inflammation. Ocular TRUST: nationwide antimicrobial suscepti-
J Immunol 186:4455–4466 bility patterns in ocular isolates. Am J Ophthalmol
29. Makdoumi K, Mortensen J, Crafoord S (2010)
145:951–958.e951
Infectious keratitis treated with corneal crosslinking. 45. Haas W, Pillar CM, Torres M, Morris TW, Sahm DF
Cornea 29:1353–1358 (2011) Monitoring antibiotic resistance in ocular
30. Papaioannou L, Miligkos M, Papathanassiou M
microorganisms: results from the ARMOR 2009
(2016) Corneal collagen cross-linking for infectious surveillance study. Am J Ophthalmol 152:567–574
keratitis: a systematic review and meta-analysis. 46. Alster Y, Herlin L, Lazar M, Loewenstein A (2000)
Cornea 35:62–71 Intraocular penetration of vancomycin eye drops after
31. Alio JL, Abbouda A, Valle DD, Del Castillo JM,
application to the medial canthus with closed lids. Br
Fernandez JA (2013) Corneal cross linking and J Ophthalmol 84:300–302
infectious keratitis: a systematic review with a meta- 47. Livermore DM (2002) Multiple mechanisms of anti-
analysis of reported cases. J Ophthalmic Inflamm microbial resistance in Pseudomonas aeruginosa: our
Infect 3:47 worst nightmare? Clin Infect Dis 34:634–640
32. Gritz DC, Lee TY, Kwitko S, McDonnell PJ (1990) 48. Su H-C, Ramkissoon K, Doolittle J, Clark M, Khatun
Topical anti-inflammatory agents in an animal J, Secrest A, Wolfgang MC, Giddings MC (2010)
model of microbial keratitis. Arch Ophthalmol The development of ciprofloxacin resistance in
108:1001–1005 Pseudomonas aeruginosa involves multiple response
33. Flach AJ (2001) Corneal melts associated with topi- stages and multiple proteins. Antimicrob Agents
cally applied nonsteroidal anti-inflammatory drugs. Chemother 54:4626–4635
Trans Am Ophthalmol Soc 99:205–210. discussion 49. Biteman B, Hassan IR, Walker E, Leedom AJ, Dunn
210–202 M, Seta F, Laniado-Schwartzman M, Gronert K
34. Guidera AC, Luchs JI, Udell IJ (2001) Keratitis, ulcer- (2007) Interdependence of lipoxin A4 and heme-
ation, and perforation associated with topical non-
2 Understanding the Role of Pro-resolving Lipid Mediators in Infectious Keratitis 11
Fusarium keratitis in soft contact lens wearers in San of fungal endophthalmitis resulting from keratitis. Am
Francisco. Arch Ophthalmol 124:1051–1053 J Ophthalmol 149:916–921
77. Gower EW, Keay LJ, Oechsler RA, Iovieno A,
85. Singh PK, Kumar A, Kumar A (2014) Resolvin D1
Alfonso EC, Jones DB, Colby K, Tuli SS, Patel SR, (RvD1) promotes the resolution of inflammation and
Lee SM, Irvine J, Stulting RD, Mauger TF, Schein protects mice from S. aureus endophthalmitis via toll-
OD (2010) Trends in fungal keratitis in the United like receptor 2 signaling. Invest Ophthalmol Vis Sci
States, 2001–2007. Ophthalmology 117:2263–2267 55:3575–3575
78. O’Day DM, Head WS, Robinson RD, Clanton JA 86. Chiang N, Fredman G, Backhed F, Oh SF, Vickery T,
(1986) Corneal penetration of topical amphotericin B Schmidt BA, Serhan CN (2012) Infection regulates
and natamycin. Curr Eye Res 5:877–882 pro-resolving mediators that lower antibiotic require-
79. Hariprasad SM, Mieler WF, Lin TK, Sponsel WE, ments. Nature 484:524–528
Graybill JR (2008) Voriconazole in the treatment of 87. Eguchi H, Hiura A, Nakagawa H, Kusaka S,
fungal eye infections: a review of current literature. Shimomura Y (2017) Corneal nerve fiber structure,
Br J Ophthalmol 92:871–878 its role in corneal function, and its changes in corneal
80. Walsh TJ, Pappas P, Winston DJ, Lazarus HM,
diseases. Biomed Res Int 2017:3242649
Petersen F, Raffalli J, Yanovich S, Stiff P, Greenberg 88. Muller RT, Abedi F, Cruzat A, Witkin D, Baniasadi
R, Donowitz G, Schuster M, Reboli A, Wingard J, N, Cavalcanti BM, Jamali A, Chodosh J, Dana R,
Arndt C, Reinhardt J, Hadley S, Finberg R, Laverdière Pavan-Langston D, Hamrah P (2015) Degeneration
M, Perfect J, Garber G, Fioritoni G, Anaissie E, Lee and regeneration of subbasal corneal nerves after
J (2002) Voriconazole compared with liposomal infectious keratitis: a longitudinal in vivo confocal
amphotericin B for empirical antifungal therapy in microscopy study. Ophthalmology 122:2200–2209
patients with neutropenia and persistent fever. N Engl 89. Cruzat A, Qazi Y, Hamrah P (2017) In vivo confocal
J Med 346:225–234 microscopy of corneal nerves in health and disease.
81. Walsh TJ, Pappas P, Winston DJ, Lazarus HM,
Ocul Surf 15:15–47
Petersen F, Raffalli J, Yanovich S, Stiff P, Greenberg 90. Mastropasqua L, Massaro-Giordano G, Nubile M,
R, Donowitz G, Schuster M, Reboli A, Wingard J, Sacchetti M (2017) Understanding the pathogenesis
Arndt C, Reinhardt J, Hadley S, Finberg R, Laverdiere of neurotrophic keratitis: the role of corneal nerves.
M, Perfect J, Garber G, Fioritoni G, Anaissie E, Lee J Cell Physiol 232:717–724
J, National Institute of, A., and Infectious Diseases 91. Cortina MS, He J, Li N, Bazan NG, Bazan HE (2010)
Mycoses Study, G (2002) Voriconazole compared Neuroprotectin D1 synthesis and corneal nerve regen-
with liposomal amphotericin B for empirical antifun- eration after experimental surgery and treatment
gal therapy in patients with neutropenia and persistent with PEDF plus DHA. Invest Ophthalmol Vis Sci
fever. N Engl J Med 346:225–234 51:804–810
82. Durand ML (2017) Bacterial and fungal endophthal- 92. Pham TL, He J, Kakazu AH, Jun B, Bazan NG, Bazan
mitis. Clin Microbiol Rev 30:597–613 HEP (2017) Defining a mechanistic link between
83. Guest JM, Singh PK, Revankar SG, Chandrasekar pigment epithelium–derived factor, docosahexaenoic
PH, Kumar A (2018) Isavuconazole for treatment acid, and corneal nerve regeneration. J Biol Chem
of experimental fungal endophthalmitis caused by 292:18486–18499
Aspergillus fumigatus. Antimicrob Agents Chemother 93. Kenchegowda S, He J, Bazan HE (2013) Involvement
62:e01537–e01518 of pigment epithelium-derived factor, docosahexae-
84. Shen YC, Wang CY, Tsai HY, Lee HN (2010)
noic acid and neuroprotectin D1 in corneal inflam-
Intracameral voriconazole injection in the treatment mation and nerve integrity after refractive surgery.
Prostaglandins Leukot Essent Fatty Acids 88:27–31
Immunoresolvent Resolvin D1
Maintains the Health of the Ocular 3
Surface
Darlene A. Dartt, Robin R. Hodges,
and Charles N. Serhan
of conjunctival goblet cell secretion. RvD1 is the film and ocular surface tissues (cornea and con-
most thoroughly studied of the specialized pro- junctiva) form the outermost protective layer that
resolution mediators in the goblet cells. This functions to maintain a transparent, healthy and
review is also focused on the role of RvD1 in impermeable cornea [1]. These components each
maintenance of ocular surface health, but not its have unique properties that contribute to the pro-
role in disease. tection of the eye from mechanical, thermal, and
chemical injury; desiccation; allergens and pol-
lutants; and pathogens from the external
3.1.2 F
unction of Tear Film environment.
and Ocular Surface At the basal aspect, the cornea consists of a
single layered endothelium, overlaid by stroma
The eye is a unique, specialized organ whose containing keratocytes, and on the apical side a
function is to provide a clear optical path for multi-layered epithelium. One of the major pro-
image presentation on the retina and its analysis tective properties of the cornea is that it is a very
by the brain (Fig. 3.1). The first optical surface in tight epithelium with limited permeability [1]. It
the eye is the tear film that overspreads the cor- is difficult to permeate the corneal epithelium
nea, a transparent avascular tissue. Together the because of the extensive tight junctions and other
tear film and cornea provide most of the refrac- types of junctions between the topmost layers of
tive power of the eye [1]. The tear film and the cells. For example, bacteria cannot penetrate the
cornea are among the first components of the corneal epithelium unless it is damaged [2].
anterior eye aided by the conjunctiva to interact Furthermore, the cornea is an immune privileged
with the external environment. The conjunctiva is site and maintains an immunosuppressive and
a vascular, optically dense tissue that surrounds anti-inflammatory environment preventing
the cornea and lines the eyelids. Together the tear immune cells from migrating into the cornea or
aqueous humor in response to an immune or The stratified squamous cells and the goblet cells
inflammatory challenge [3]. When injured, the both produce the mucous layer of the tear film but
cornea normally heals rapidly with minimal scar- release different types of mucins. Both cells
ring and no blood vessel ingrowth. The number secrete electrolytes and water.
of mechanisms that the cornea can use to respond Secretion of mucins is one of the major pro-
to the external environment is limited and often tective mechanisms of the conjunctiva. The
constrained, as this tissue needs to preserve trans- mucins produce an extensive glycocalyx and the
parency and remain avascular to ensure clear soluble mucous layer that overspreads both the
vision. cornea and conjunctiva. Stratified squamous cells
The conjunctiva, the focus of this review, sur- produce the membrane spanning mucins MUC1,
rounds the cornea and lines the lids. One of its MUC4, MUC16, and MUC20 (for a review see
major functions is to secrete electrolytes, water, [6]). These mucins help form the glycocalyx, a
and multiple types of mucins that together form thick coat of carbohydrates that emerges from the
the glycocalyx and the inner layer of the tear film apical membranes of epithelial cells and protects
[4]. The conjunctiva is a stratified squamous epi- the corneal and conjunctival surface. The glyco-
thelium that overlies a loose, disorganized stroma calyx interfaces with the tear film. The glycoca-
containing plentiful blood vessels and nerves. lyx is critical to maintaining the barrier function
This epithelium consists of two major cell types, of the cornea, preventing access of microbes to
stratified squamous cells and goblet cells the plasma membrane, and preventing apical
(Fig. 3.2) [4]. Stratified squamous cells are multi- adhesion [7]. In contrast, the goblet cells secrete
layered, have complex interdigitated lateral the large soluble, gel-forming mucin MUC5AC
membranes, and contain a plethora of small clear [8]. This mucin is the major component of the
vesicles. The second cell type is the goblet cells inner mucous layer of the tear film and is essen-
[5]. These cells occur singly or in clusters and tial for ocular surface health. MUC5AC upon
span the entire epithelium in rodents, but only secretion by the goblet cells overspreads the
occupy the mid portion of the epithelium reach- entire ocular surface including the cornea. It pro-
ing to the surface in humans [5]. The goblet cells tects the underlying epithelia as a mucus gel by
are polarized with a basal nucleus, substantial preventing attachment of bacteria and keeping
Golgi apparatus, and a plethora of secretory gran- them suspended in the gel. The water and electro-
ules that occupy most of the cell volume and lytes in the gel keep the ocular surface hydrated.
reach to the apical membrane and the tear film. The movement of the mucus gel across the ocular
surface into the lacrimal drainage ducts removes depletion of MUC5AC in the tear film lead to
particles, pollutants, and other environmental serious damaging, painful ocular surface diseases
components from the ocular surface. Goblet cells such as dry eye and vitamin A deficiency [14].
and their secretion of MUC5AC are regulated by Increase in goblet cell mucin in the tear film is
nerves. Afferent sensory nerves and efferent also characteristic of specific diseases, such as
parasympathetic and sympathetic nerves extend ocular allergy, that upset ocular surface homeo-
into the conjunctival epithelium interdigitating stasis and can be damaging to the ocular surface.
between the stratified squamous cells. Efferent The finding that both a decrease and an increase
parasympathetic and sympathetic nerves sur- in goblet cell mucin secretion leads to disease
round the goblet cells. The localization of the suggests that goblet cell mucin secretion must be
nerves in the epithelium provides the basis for tightly regulated to maintain an optimal amount
these epithelial cells to respond quickly to of mucin in the tear film [6, 8, 15].
changes in the external environment by secretion In health neural regulation of goblet cell
of MUC5AC into the tears. mucin secretion provides this regulation [16, 17].
The conjunctiva has multiple protective mech- Using a complex neural reflex, goblet cell mucin
anisms in addition to the secretion of secretion can be exquisitely regulated to respond
MUC5AC. As the conjunctival epithelium is to changes in the external environment to secrete
opaque, well vascularized, and very permeable, mucins as needed to protect the occur surface
unlike the cornea, it is not constrained in its [18]. Corneal and conjunctival afferent sensory
responses to the environment. The conjunctiva in nerves are activated by changes in temperature,
fact can respond very robustly. The conjunctiva acid or bases, or mechanical stimuli (including
contains conjunctival associated lymphoid tissue trauma and particulates that occur in pollution),
that is part of the mucosal immune system [9]. In for examples. The stimulated afferent nerves then
addition the conjunctival stroma contains numer- activate the trigeminal ganglion that by a com-
ous different cells for innate defense (macro- plex neural reflex within the brain activates effer-
phages, neutrophils, and mast cells) and for ent parasympathetic nerves In the conjunctiva
immune protection (lymphocytes, plasma cells, parasympathetic nerve endings surround the gob-
and dendritic cells). In addition for the innate let cells and stimulate them to secrete mucins into
defense system the conjunctiva has most of the the tear film [17]. The goblet cells also likely
Toll-like receptors [10], multiple NOD-like secrete electrolytes and water, but the evidence
receptors, and a constitutively assembled NLRP3 for this is indirect [19, 20].
inflammasome poised for activation [11, 12]. The Parasympathetic nerves are the major stimulus
conjunctiva also has goblet cell associated pas- of goblet cell mucin secretion in health [16, 21,
sages (GAPs) that are openings between goblet 22] (Fig. 3.3). There is no published evidence for
cells that allow the stroma to sample antigens and the role of sympathetic nerves in this secretion.
other immune activating material in the external Parasympathetic nerves release the neurotrans-
environment [13]. These GAPs are under para- mitters acetylcholine that activate muscarinic
sympathetic muscarinic control. receptors (MAchR) type 1, 2, and 3 and vasoac-
tive intestinal peptide (VIP) that uses the VPAC1
and 2 receptors [21]. Both acetylcholine (carba-
3.1.3 R
ole Goblet Cells in Ocular chol is used experimentally) and VIP stimulate
Surface Health secretion by increasing intracellular [Ca2+]
([Ca2+]i) and activating extracellular regulated
As goblet cells and their secretion of MUC5AC kinase (ERK)1/2 also known as p44/p42 mitogen
are critical for ocular surface health, both a activated protein kinase (MAPK) [13, 23]. In
decrease and an increase in goblet cell mucin addition VIP activates adenylyl cyclase to
secretion leads to ocular surface disease. Loss of increase the cellular cAMP level (For a review
goblet cells from the conjunctival epithelium and see [24]). Acetylcholine (carbachol), but not VIP,
3 Immunoresolvent Resolvin D1 Maintains the Health of the Ocular Surface 17
Fig. 3.3 Schematic representation of cholinergic path- mitogen activated kinase kinase (MEK), and ERK 1/2.
way leading to goblet cell mucin secretion. Muscarinic Vasoactive intestinal peptide (VIP) binds to its receptors
receptors activate phospholipase C (PLC) to generate the VPAC1 or 2 to activate adenylyl cyclase (AC) to generate
production of inositol trisphosphate (IP3), which releases cAMP from ATP. cAMP then activates protein kinase A
intracellular Ca2+ and diacylglycerol (DAG), which acti- (PKA) to stimulate secretion. Reprinted from Hodges
vates protein kinase C (PKC). The EGF receptor (EGFR) et al. Encyclopedia of the Eye, Dartt D, Dana R, Besharse
is transactivated leading to the activation of Ras, Raf, J eds. Elsevier, 2010
The enzymes responsible for this biosynthesis found that the normal, uninflamed conjunctiva
are the lipoxygenases (LOX) 5-LOX and 12/15 from various species of animals has the capacity
LOX. The location and specificity of the LOXs to synthesize both cyclooxygenases and lipoxy-
are cell and tissue specific. This determines the genases [27, 28]. The cycloxygenase was higher
type and amount of SPM produced. In general than the lipoxygenase activity. The major lipoxy-
15-LOX and 5-LOX are needed to produce lipox- genases produced by the conjunctiva were 12-
ins from arachidonic acid. 5-LOX is required to HETE, 5-HETE, and 5,12-diHETE suggesting
biosynthesize E-series resolvins. 15-LOX and production of hepoxilin and LTA4. Subsequently
5-LOX biosynthesize D-series resolvins and pro- conjunctiva and eyelids were demonstrated to
tectin (Fig. 3.4). 12-LOX biosynthesizes mares- possess EPA lipoxygenase products of the
ins. There is little published information on LOX 5-series suggesting the capability of producing
enzymes in the conjunctiva and especially the E-series resolvins [29].
goblet cells. Several articles from the 1980s
Docosahexaenoic Acid
15-LO
COX-2/
ASA
17S-H(p)-DHA 17R-H(p)-DHA
5-LO 5-LO
Enzymatic Enzymatic
epoxidation epoxidation
7,8-epoxy-17S-HDHA
7,8-epoxy-17R-HDHA
Enzymatic
Enzymatic
Hydrolysis
Hydrolysis
RvD1 AT-RvD1
7S,8R,17S-trihydroxy- 7S,8R,17R-trihydroxy-
4Z,9E,11E,13Z,15E,19Z-DHA 4Z,9E,11E,13Z,15E,19Z-DHA
Fig. 3.4 Synthetic Pathways of the D-series specialized pro-resolving mediators. Each is derived from DHA. 5-LO: 5
lipoxygenase Modified from Sun Y, Oh SF et al. J Biol Chem 282:9323–9234, 2007
3 Immunoresolvent Resolvin D1 Maintains the Health of the Ocular Surface 19
There is substantial work on the LOX enzymes we will review the evidence for the role of the
in the cornea and the draining lymph nodes of the D-series resolvin RvD1 in conjunctival health.
eye that contribute to ocular surface disease espe-
cially [30–32], 5- and 15-LOX are the rate limit-
ing enzymes responsible for the generating SPMs 3.3 vD1 Is Present in Tears
R
[33]. The corneal epithelium has high expression from Healthy Individuals
of 15-LOX and functions as part of a LXA4-ALX/
FPR2 circuit that controls wound healing and A number of eicosanoids as well as SPMs are
immune response in the cornea. In particular there found in human tears. Human emotional tears
is a population of tissue specific polymorphonu- were collected from six male and six female sub-
clear leukocytes (PMNs) that contain especially jects and the lipid profile analyzed using an
high amounts of 15-LOX and that in a sex-spe- LC-MS-MS based metabololipidomics along
cific manner play a role in exacerbating autoim- with deuterium-labeled SPM as internal stan-
mune dry eye disease in females. These PMNs dards for quantitation [41]. We documented the
were detected in the lacrimal gland, draining presence of pro-inflammatory prostaglandins and
lymph nodes, and limbus of the cornea. As the the leukotriene B4. For the SPMs, the D-series
cornea and its limbus is adjacent to the conjunc- resolvins (RvD1, RvD2, RvD5), protectin D1,
tiva, it is possible that conjunctival epithelial cells and lipoxin A4, but neither the maresins nor
also have elevated levels of 15-LOX. Furthermore, E-series resolvins, were identified in these sam-
conjunctival goblet cells are a target of disease in ples from healthy human subjects. The SPM bio-
autoimmune dry eye. Study of ocular surface dis- synthesis pathway markers 17-HDHA,
ease has shown the presence of high levels of the 14-HDHA, and 18-HEPE were also identified.
biosynthetic enzymes for the SPMs and the cir- These compounds could be bioactive themselves
cuits for their regulation. These enzymes could [42] or could suggest that both D- and E-series
also function in health especially as during eye resolvins may be present in higher amounts
closure in sleep, PMNs migrate into the tear film locally than measured in the present study. The
and could interact with the conjunctival epithelial presence of pathway markers would also be con-
cells to set up a transient, low level activation of a sistent with the further metabolism of these bio-
LXA4-ALX/FPR2 like circuit or other circuits active resolvins to their oxo- and dehydro-resolvin
potentially in the conjunctiva. The same enzymes products that were not profiled in the present
produce RvD1 and could be critical for regulating study and are usually less or devoid of bioactiv-
RvD1 biosynthesis in conjunctival goblet cells. ity. Thus, RvD1 was present in human tears and
In addition to the production of SPMs in dis- available to regulate the function of the conjunc-
ease, SPMs are also endogenously produced in tival goblet cells. There are two other studies on
human tissues rich in omega 3 fatty acids in the SPMs in tears that are in agreement with that of
absence of disease [34]. Thus SPMs are produced English et al. [41]. Walter et al. [43] found DHA,
in human milk [35, 36], blood [37, 38], brain [39], the ω 3 fatty acid from which RvD1 is biosynthe-
and retina [40]. SPMs are major players in the sized, in tears. RvD1 was not measured directly
regulation of ocular surface health as SPMs are in this study. Masoudi S et al. [44] found RvD1 in
detected in the tear film and stimulate conjuncti- tears, but this was the only SPM analyzed.
val goblet cell mucin secretion under physiologic Surprisingly the lipid profile of male and female
conditions. They function to maintain an opti- tears differed substantially [41]. Use of a principal
mum mucin layer of the tear film in response to component analysis demonstrated a gender differ-
the normal changes in the cornea and conjunctiva ence in the SPMs in tears (Fig. 3.5). The loading
and to the extracellular environment. Multiple plot calculated from LC-MS-MS identified RvD1,
SPM family members are present within emo- RvD2, RvD5 and protectin D1 in male tears. In
tional human tears and stimulate goblet cell secre- contrast LXA4 and aspirin-triggered LXA4 were
tion to maintain the healthy tear film [41] . Herein detected in female tears. When the ratio of SPMs,
20 D. A. Dartt et al.
AT-LXA,
LXA,
RvD1
5-HETE
a
Principal component 1
5S, 15S-diHETE 12-HETE
LTB,
Human Emotional Tears 2D Score Plot 18-HEPE 5S,12S-diHETE
PGD,
14-HDHA
RvD5
15-HETE
RvD2
Female Donors (n=6) 17-HDHA
Principal component 1
PGE,
PGF,a
PD1 20-OH-LTB,
Principal component 2
Fig. 3.5 PCA and quantitative ratio by gender for male donors & green circles are associated with female
LM-SPMs identified in human emotional tears. (a) donors. (c) Bar graph depicting the ratio of total SPMs
2-dimensional score plot of human emotional tear donors; including RvD1, RvD2, RvD5, PD1, LXA4, AT-LXA4,
blue circles (n = 6) are representative of males, while 17-HDHA, and 18-HEPE compared to PGE2 and PGF2α
green circles (n = 6) are representative of females. Gray (pg/100 μl), in males compared females (n = 6 for each
ellipse denotes 95% confidence interval. (b) 2-dimensional gender; #P < 0.05 for male donors vs. female donors;
loading plot of LM-SPMs identified in human emotional ∗P < 0.05, females vs. males). Reprinted from English
tears; blue circles are those mediators associated with et al. [41]
17-HDHA, and 18-HEPE versus the pro-inflam- was our focus. RvD1 stimulated human and rat
matory compounds PGE2 and PGF2a was com- conjunctival goblet cell high molecular weight
pared between males and females, the ratio of glycoconjugate secretion (that includes mucin)
SPMs to prostaglandins was much higher in males. [45, 46]. Secretion peaked at 1 h and then declined
over time for the next 3 h. RvD1 stimulated
secretion by increasing the [Ca2+]i and activating
3.4 xogenous RvD1 Stimulates
E ERK1/2 [45], similarly to the effect of choliner-
Conjunctival Goblet Cell gic agonists and VIP [21] (Fig. 3.6). RvD1 was
Mucin Secretion Using effective in the range of 10−10–10−8 M, a much
Multiple Intracellular lower concentration range than for carbachol
Signaling Pathways (10−6–10−4 M), but similar to VIP (10−11–10−7 M)
[21, 23]. The RvD1 stimulation of secretion was
3.4.1 R
vD1 Stimulates Goblet Cell blocked by chelating the intracellular Ca2+ with
Mucin Secretion BAPTA and by inhibiting ERK1/2 activation
with U0126 [47] substantiating the role of these
As the major function of goblet cells is to secrete signaling molecules in RvD1 stimulation of gob-
mucins, the action of RvD1 on mucin secretion let cell secretion.
3 Immunoresolvent Resolvin D1 Maintains the Health of the Ocular Surface 21
To induce goblet cell function the appropri- blocked RvD1 stimulated increase in [Ca2+]i
ate receptors must be present on conjunctival [45, 47]. Functional evidence also demon-
goblet cells. RvD1 uses the ALX/FPR2 recep- strates that RvD1 uses the GPR32/DRV1
tor in rat and the GPR32/DRV1 receptor in receptor in human goblet cells. In human cells
human goblet cells. PCR, western blot and inhibition of ALX/FPR2 with its inhibitor
immunofluorescence microscopy demon- BOC-2 does not block RvD1 stimulated
strated that the ALX/FPR2 receptor was pres- increase in [Ca2+]i [49]. Furthermore, in desen-
ent on rat conjunctival goblet cells in vivo and sitization experiments in human cells in which
in culture [48, 49]. There is as yet no published LXA4 and RvD1 were used sequentially, RvD1
report of GPR32/DRV1 on human goblet cells. does not desensitize LXA4 response [49]. This
There is also functional evidence for the pres- suggests use of separate receptors in human
ence of ALX/FPR2 receptor in rat conjunctival goblet cells. Thus RvD1 activates ALX/
goblet cells. To address whether RvD1 uses FPR2 in rat goblet cells and GPR32/DRV1 in
the ALX/FPR2 receptor in the rat, both the human goblet cells. Further experiments using
ALX/FPR2 inhibitor N-Boc−Phe-Leu-Phe- GPR32 siRNA in human goblet cells is
Leu-Phe (BOC-2) and siRNA for ALX/FPR2 warranted.
22 D. A. Dartt et al.
3.4.2 C
ellular Signaling Pathways [Ca2+]i supported the activation of Ca2+ influx by
Activated by RvD1 RvD1 [45].
The DAG arm of the PLC pathway was inves-
3.4.2.1 Phospholipase C Pathway tigated by determining the role of PKC using the
RvD1 activates a several specific intracellular PKC inhibitor Ro 31-8220. The PKC inhibitor
signaling pathways to stimulate mucin secretion blocked the RvD1-stimulated increase in [Ca2+]i,
from rat goblet cells. One pathway is activation but not in secretion. Activation of PKC may be
of phospholipase (PL) C that produces water important for the increase in [Ca2+]i, but not for
soluble 1,4,5-inositol trisphosphate (IP3) and secretion. There are multiple isoforms of PKC
lipid soluble (membrane-bound) diacylglycerol that are differentially activated by Ca2+ and diac-
(DAG) (Fig. 3.6). IP3 binds to its receptors ylglycerol and can have opposing effects on a
(IP3R) on the endoplasmic reticulum that given process [50]. Thus investigation of the
releases Ca2+ from this intracellular store to types of PKC isoforms present in goblet cells and
increase the cytosolic [Ca2+] that stimulates exo- inhibition of single isoforms would demonstrate
cytosis and mucin secretion. The DAG produced more accurately whether PKC isoforms are
activates protein kinase (PK)C that phosphory- involved in goblet cell secretion.
lates as yet unidentified substrates to stimulate We concluded that RvD1 uses a PLC pathway
secretion. We used multiple techniques and to increase IP3 that releases Ca2+ from intracel-
inhibitors to determine if a PLC-dependent path- lular stores and causes an increase in the influx of
way plays a role in RvD1 stimulated goblet cell extracellular Ca2+ (Fig. 3.6). The resultant
secretion [47]. First the active PLC inhibitor increase in the cytosolic [Ca2+]i stimulates secre-
U73122 blocked RvD1 stimulated increase in tion. The second arm of the PLC pathway pro-
[Ca2+]i and secretion as well as the increase duction of DAG and activation of PKC is used to
caused by the positive control the cholinergic increase the [Ca2+]i, but whether PKC plays a role
agonist carbachol. As expected, the inactive in secretion awaits investigation of the different
inhibitor U73343 did not block either the RvD1 PKC isoforms.
or cholinergic agonist stimulation of secretion PLCγ is activated by EGF as PLCγ is an
nor the cholinergic agonist induced increase in adapter molecule attached to the EGFR. PLCγ
[Ca2+]i. Unfortunately the inactive inhibitor can be phosphorylated and activated upon stimu-
blocked the RvD1 induced increase in [Ca2+]i but lation and dimerization of the EGFR. We recently
was not as effective as the active analog. As the found that RvD1 uses a matrix metalloproteinase
inactive inhibitor did not block three out of four ADAM17 to release EGF by ectodomain shed-
responses, we concluded that RvD1 activates ding and activate the EGFR to increase [Ca2+]i
PLC in conjunctival goblet cells. To investigate and activate ERK1/2 to stimulate secretion [51].
IP3 interaction with its receptor to release Ca2+ Thus RvD1 could also activate PLCγ in addition
from intracellular stores, an inhibitor of the IP3 to PLCβ to increase [Ca2+]i and stimulate secre-
receptor 2-aminoethyl diphenylborate (2-APB) tion. Thus two different types of PLC are used by
was used. 2-APB blocked both the RvD1 stimu- RvD1 to stimulate goblet cell mucin secretion.
lated increase in [Ca2+]i and secretion. A second
method of determining the role of intracellular 3.4.2.2 Phospholipase D
Ca2+ stores is the use of thapsigargin that blocks and A2 Pathways
the re-uptake of Ca2+ into the stores, thereby The next two pathways investigated were activa-
depleting them of Ca2+. If RvD1 used intracellu- tion of PLD and PLA2 [47] (Fig. 3.6). These path-
lar Ca2+ stores, the addition of thapsigargin ways were not investigated in as much detail as
before RvD1 should prevent the increase in PLC. The role of PLD in RVD1 stimulated increase
[Ca2+]i by RvD1. In goblet cells this did not occur in [Ca2+]i and secretion was investigated using
[45]. However, that depletion of extracellular 1-butanol the active inhibitor of PLD and tert-buta-
Ca2+ blocked the RvD1 stimulated elevation in nol, its inactive control. The RvD1- induced
3 Immunoresolvent Resolvin D1 Maintains the Health of the Ocular Surface 23
increase in [Ca2+]i and stimulation of secretion was gens, particulate matter, and pathogens in the
almost completely blocked by 1-butanol. The inac- external environment. RvD1 is present in the
tive control only partially blocked the increase in tear film where it can access its receptors on the
[Ca2+]i and did not block the stimulation of secre- basolateral membranes of the goblet cells. RvD1
tion. To study PLA2 aristolochic acid was used. uses the ALX/FPR2 receptor in rat goblet cells
Aristolochic acid blocked both the RvD1 caused and the GPR32 in human goblet cells. Activation
increase in [Ca2+]i and stimulation of secretion. of these receptors employs multiple intracellular
These results are consistent with RvD1 using both pathways including PLC, PLD, PLA2 and the
PLD and PLA2 to stimulate goblet cell secretion. EGFR to increase [Ca2+]i and activate ERK1/2 to
Additional experiments should identify the com- stimulate secretion. The main secretory product
ponents of these pathways. of the goblet cells is the large, gel-forming
mucin MU5AC, which is released into the inner-
3.4.2.3 Extracellular Regulated Kinase most layer of the tear film where it is protective
1/2 Pathway of the ocular surface. This mucin can trap bacte-
RvD1 activates ERK1/2 to increase [Ca2+]i and ria and particulate matter and remove them from
stimulate secretion as shown by inhibition of the ocular surface via the nasal lacrimal drain-
both functions by the MEK inhibitor U0126 [45] age. RvD1 thus preserves ocular surface homeo-
(Fig. 3.6). ERK1/2 could function as a compo- stasis and maintains this surface in a
nent in several of the pathways studied. ERK1/2 non-inflamed, normal physiologic state of an
could be activated by induction of the EGFR optimum amount of mucin secretion. RvD1
using the adapter proteins Ras, Raf, and forms a second important mechanism along with
MEK. ERK1/2 could also be downstream of PLD activation of a neural reflex pathway to regulate
or PLA2. goblet cell mucin secretion and protect the ocu-
lar surface in health.
7. Mauris J, Mantelli F, Woodward AM, Cao Z, Bertozzi 22. Kanno H, Horikawa Y, Hodges RR, Zoukhri D,
CR, Panjwani N et al (2013) Modulation of ocular Shatos MA, Rios JD et al (2003) Cholinergic agonists
surface glycocalyx barrier function by a galectin-3 transactivate EGFR and stimulate MAPK to induce
N-terminal deletion mutant and membrane-anchored goblet cell secretion. Am J Physiol Cell Physiol
synthetic glycopolymers. PLoS One 8(8):e72304 284(4):C988–C998
8. Hodges RR, Dartt DA (2013) Tear film mucins: front 23. Li D, Jiao J, Shatos MA, Hodges RR, Dartt DA (2013)
line defenders of the ocular surface; comparison with Effect of VIP on intracellular [Ca2+], extracellular
airway and gastrointestinal tract mucins. Exp Eye Res regulated kinase 1/2, and secretion in cultured rat
117:62–78 conjunctival goblet cells. Invest Ophthalmol Vis Sci
9. Knop E, Knop N (2005) The role of eye-associated 54(4):2872–2884
lymphoid tissue in corneal immune protection. J Anat 24. Dickson L, Finlayson K (2009) VPAC and PAC
206(3):271–285 receptors: from ligands to function. Pharmacol Ther
10. Kinoshita S, Ueta M (2010) Innate immunity of the 121(3):294–316
ocular surface. Jpn J Ophthalmol 54(3):194–198 25. Puro DG (2018) Role of ion channels in the functional
11. Li D, Hodges RR, Bispo P, Gilmore MS, Gregory- response of conjunctival goblet cells to dry eye. Am
Ksander M, Dartt DA (2017) Neither non-toxigenic J Physiol Cell Physiol 315(2):C236–CC46
Staphylococcus aureus nor commensal S. epidermidi 26.
Serhan CN (2014) Pro-resolving lipid media-
activates NLRP3 inflammasomes in human conjuncti- tors are leads for resolution physiology. Nature
val goblet cells. BMJ Open Ophthalmol 2(1):e000101 510(7503):92–101
12.
McGilligan VE, Gregory-Ksander MS, Li D, 27. Williams RN, Bhattacherjee P, Eakins KE (1983)
Moore JE, Hodges RR, Gilmore MS et al (2013) Biosynthesis of lipoxygenase products by ocular tis-
Staphylococcus aureus activates the NLRP3 inflam- sues. Exp Eye Res 36(3):397–402
masome in human and rat conjunctival goblet cells. 28. Kulkarni PS, Srinivasan BD (1989) Cyclooxygenase
PLoS One 8(9):e74010 and lipoxygenase pathways in anterior uvea and con-
13. Barbosa FL, Xiao Y, Bian F, Coursey TG, Ko BY, junctiva. Prog Clin Biol Res 312:39–52
Clevers H et al (2017) Goblet cells contribute to ocu- 29. Kulkarni PS, Kaufman PL, Srinivasan BD (1987)
lar surface immune tolerance-implications for dry eye Eicosapentaenoic acid metabolism in cynomolgus
disease. Int J Mol Sci 18(5):978 and rhesus conjunctiva and eyelid. J Ocul Pharmacol
14. Baudouin C, Rolando M, Benitez Del Castillo JM, 3(4):349–356
Messmer EM, Figueiredo FC, Irkec M et al (2018) 30. Gao Y, Su J, Zhang Y, Chan A, Sin JH, Wu D et al
Reconsidering the central role of mucins in dry eye (2018) Dietary DHA amplifies LXA4 circuits in
and ocular surface diseases. Prog Retin Eye Res. tissues and lymph node PMN and is protective in
2018, In Press. immune-driven dry eye disease. Mucosal Immunol
15. Dartt DA, Masli S (2014) Conjunctival epithelial
11(6):1674–1683
and goblet cell function in chronic inflammation and 31. Gao Y, Min K, Zhang Y, Su J, Greenwood M, Gronert
ocular allergic inflammation. Curr Opin Allergy Clin K (2015) Female-specific downregulation of tis-
Immunol 14(5):464–470 sue polymorphonuclear neutrophils drives impaired
16. Dartt DA, Kessler TL, Chung EH, Zieske JD (1996) regulatory T cell and amplified effector T cell
Vasoactive intestinal peptide-stimulated glycoconju- responses in autoimmune dry eye disease. J Immunol
gate secretion from conjunctival goblet cells. Exp Eye 195(7):3086–3099
Res 63(1):27–34 32. Wei J, Gronert K (2017) The role of pro-resolving
17. Dartt DA, McCarthy DM, Mercer HJ, Kessler TL, lipid mediators in ocular diseases. Mol Asp Med
Chung EH, Zieske JD (1995) Localization of nerves 58:37–43
adjacent to goblet cells in rat conjunctiva. Curr Eye 33. Serhan CN, Hamberg M, Samuelsson B (1984)
Res 14(11):993–1000 Lipoxins: novel series of biologically active com-
18. Kessler TL, Mercer HJ, Zieske JD, McCarthy DM, pounds formed from arachidonic acid in human
Dartt DA (1995) Stimulation of goblet cell mucous leukocytes. Proc Natl Acad Sci U S A 81(17):
secretion by activation of nerves in rat conjunctiva. 5335–5339
Curr Eye Res 14(11):985–992 34. Barden AE, Mas E, Mori TA (2016) n-3 fatty acid
19. Candia OA, Alvarez LJ (2008) Fluid transport phe- supplementation and proresolving mediators of
nomena in ocular epithelia. Prog Retin Eye Res inflammation. Curr Opin Lipidol 27(1):26–32
27(2):197–212 35. Weiss GA, Troxler H, Klinke G, Rogler D, Braegger
20. Candia OA, Kong CW, Alvarez LJ (2008) IBMX- C, Hersberger M (2013) High levels of anti-
elicited inhibition of water permeability in the iso- inflammatory and pro-resolving lipid mediators
lated rabbit conjunctival epithelium. Exp Eye Res lipoxins and resolvins and declining docosahexaenoic
86(3):480–491 acid levels in human milk during the first month of
21. Rios JD, Zoukhri D, Rawe IM, Hodges RR, Zieske lactation. Lipids Health Dis 12:89
JD, Dartt DA (1999) Immunolocalization of mus- 36. Arnardottir H, Orr SK, Dalli J, Serhan CN (2016)
carinic and VIP receptor subtypes and their role in Human milk proresolving mediators stimulate reso-
stimulating goblet cell secretion. Invest Ophthalmol lution of acute inflammation. Mucosal Immunol
Vis Sci 40(6):1102–1111 9(3):757–766
3 Immunoresolvent Resolvin D1 Maintains the Health of the Ocular Surface 25
37. Mas E, Croft KD, Zahra P, Barden A, Mori TA (2012) 45. Li D, Hodges RR, Jiao J, Carozza RB, Shatos MA,
Resolvins D1, D2, and other mediators of self-limited Chiang N et al (2013) Resolvin D1 and aspirin-
resolution of inflammation in human blood fol- triggered resolvin D1 regulate histamine-stimulated
lowing n-3 fatty acid supplementation. Clin Chem conjunctival goblet cell secretion. Mucosal Immunol
58(10):1476–1484 6(6):1119–1130
38. Colas RA, Shinohara M, Dalli J, Chiang N, Serhan CN 46. Dartt DA, Hodges RR, Li D, Shatos MA, Lashkari K,
(2014) Identification and signature profiles for pro- Serhan CN (2011) Conjunctival goblet cell secretion
resolving and inflammatory lipid mediators in human stimulated by leukotrienes is reduced by resolvins
tissue. Am J Physiol Cell Physiol 307(1):C39–C54 D1 and E1 to promote resolution of inflammation.
39. Zhu M, Wang X, Hjorth E, Colas RA, Schroeder L, J Immunol 186(7):4455–4466
Granholm AC et al (2016) Pro-resolving lipid media- 47. Lippestad M, Hodges RR, Utheim TP, Serhan CN,
tors improve neuronal survival and increase Abeta42 Dartt DA (2017) Resolvin D1 increases mucin secre-
phagocytosis. Mol Neurobiol 53(4):2733–2749 tion in cultured rat conjunctival goblet cells via mul-
40. Mukherjee PK, Marcheselli VL, Serhan CN, Bazan tiple signaling pathways. Invest Ophthalmol Vis Sci
NG (2004) Neuroprotectin D1: a docosahexaenoic 58(11):4530–4544
acid-derived docosatriene protects human retinal pig- 48. Hodges RR, Li D, Shatos MA, Bair JA, Lippestad M,
ment epithelial cells from oxidative stress. Proc Natl Serhan CN et al (2017) Lipoxin A4 activates ALX/
Acad Sci U S A 101(22):8491–8496 FPR2 receptor to regulate conjunctival goblet cell
41. English JT, Norris PC, Hodges RR, Dartt DA, Serhan secretion. Mucosal Immunol 10(1):46–57
CN (2017) Identification and profiling of specialized 49. Hodges RR, Li D, Shatos MA, Serhan CN, Dartt
pro-resolving mediators in human tears by lipid medi- DA (2016) Lipoxin A4 counter-regulates histamine-
ator metabolomics. Prostaglandins Leukot Essent Fat stimulated glycoconjugate secretion in conjunctival
Acids 117:17–27 goblet cells. Sci Rep 6:36124
42. Lima-Garcia JF, Dutra RC, da Silva K, Motta EM, 50. Zoukhri D, Hodges RR, Sergheraert C, Dartt DA
Campos MM, Calixto JB (2011) The precursor of (1998) Protein kinase C isoforms differentially con-
resolvin D series and aspirin-triggered resolvin D1 dis- trol lacrimal gland functions. Adv Exp Med Biol
play anti-hyperalgesic properties in adjuvant-induced 438:181–186
arthritis in rats. Br J Pharmacol 164(2):278–293 51. Kaye R, Botten N, Lippestad M, Li D, Hodges RR,
43. Walter SD, Gronert K, McClellan AL, Levitt RC, Utheim TP et al (2018) Resolvin D1, but not resolvin
Sarantopoulos KD, Galor A (2016) omega-3 tear film E1, transactivates the epidermal growth factor recep-
lipids correlate with clinical measures of dry eye. tor to increase intracellular calcium and glycoconju-
Invest Ophthalmol Vis Sci 57(6):2472–2478 gate secretion in rat and human conjunctival goblet
44. Masoudi S, Zhao Z, Willcox M (2017) Relation
cells. Exp Eye Res 180:53–62
between ocular comfort, arachidonic acid mediators,
and histamine. Curr Eye Res 42(6):822–826
The Evolving Role of Specialized
Pro-resolving Mediators 4
in Modulating Neuroinflammation
in Perioperative Neurocognitive
Disorders
Ting Yang and Niccolò Terrando
both PND patients and animal models, implicat- nuclear factor (NF)-κB signaling pathway [9]
ing surgery-induced neuroinflammation as a key [10]. BBB homeostasis plays fundamental role in
contributor in the pathophysiology of PNDs. The the communication between peripheral inflam-
aim of this review is to discuss the role of dys- mation and neuroinflammation [11]. Using
regulated immunity after surgery and the thera- fibrinogen as a classical marker of BBB disrup-
peutic potential for specialized pro-resolving tion, we reported a significant deposition of this
lipid mediators (SPMs) in the perioperative neu- blood-derived molecule in the hippocampal
rocognitive arena. parenchyma [12]. Similar results were reported
We and others have been interested in clarify- in other surgical models, with evident BBB open-
ing the pathogenesis of PNDs and developed pre- ing and disruption of tight junctions after anes-
clinical translational models to study the impact thesia and surgery [13]. More recently we
of anesthesia and surgery on the CNS. During described a role for neuronal processing, includ-
surgery, aside from direct effects of anesthetic ing pain signaling at the level of the spinal cord,
agents on the CNS (reviewed in [5]), acute in disrupting acute neurogenesis after orthopedic
inflammation triggered by trauma and sterile surgery [14]. These factors, together with the
injury can negatively affect brain function, con- overall activation of innate immune pathways,
tributing to sickness behavior and PND-like have been highlighted as pathogenic mechanisms
pathology [6]. If poorly controlled, dysregulated underlying cognitive decline in preclinical mod-
inflammation leads to more extensive organ dys- els and in early clinical studies. Preclinical mod-
function and tissue injury. Release of pro- els have evaluated multiple therapeutic strategies
inflammatory cytokines, alarmins, and to modulate neuroinflammation and PND-like
leukotrienes appear to associate with distant behavior. Selective targeting of key pro-
long-term effects on the brain, contributing inflammatory cytokines (such as anti-TNFα and
to neuroinflammation, neurotoxicity, and subse- IL-1 receptor agonist) prevent neuroinflamma-
quent memory impairments. The mechanisms tion and improve cognitive decline in animal
whereby systemic inflammation affects CNS models [6, 15]. Macrophage-specific deletion of
function remain partly defined and are object of IKKβ, a central coordinator of TNFα activation
active investigations. Systemic cytokines have of NF-κB, prevents BBB disruption and macro-
been shown to enter the brain through different phage infiltration in the hippocampus following
immune-to-brain signaling pathways, which surgery. Moreover, harnessing the cholinergic
include humoral, neuronal, and cellular routes reflex by stimulating the α7 subtype of nicotinic
[7]. Using a well-established PND model of acetylcholine receptors (α7 nAChR) in macro-
orthopedic surgery, which frequently leads to phages inhibited NF-κB activity and BBB dis-
cognitive dysfunctions in humans, we first ruption, thus preventing neuroinflammation and
reported a key role for systemic pro-inflammatory cognitive decline following surgery [12].
cytokines, including interleukin (IL)-1β and Although these and several other therapies may
tumor necrosis factor alpha (TNFα), in mediating successfully reduce postoperative neuroinflam-
surgery-induced neuroinflammation and cogni- mation and limit PNDs, they often contribute to
tive decline following surgery [6, 8]. These initial side effects by over suppressing the immune sys-
results as well as work from other models of tem, increasing the risk of infection and delaying
surgery-induced cognitive dysfunction, prompt wound healing, which are all crucial in the con-
for a prominent role of the systemic and humoral text of postoperative recovery. Thus, it is pivotal
response in PND pathogenesis. However, periph- to identify safer therapeutic strategies to prevent
eral cytokines in response to non-CNS surgery neuroinflammation without suppressing systemic
were also shown to disrupt the blood-brain bar- immune functions.
rier (BBB), thus facilitating the migration of Is now well appreciated that resolution of
peripheral cells, including macrophages, into the inflammation is an active process [16–18].
brain parenchyma through activation of TNFα/ Synthesis and release of anti-inflammatory
4 The Evolving Role of Specialized Pro-resolving Mediators in Modulating Neuroinflammation… 29
attenuating release of pro-inflammatory cyto- cytes actions may be determined by the danger
kines [39–41]. Importantly, other classes of signals in the local environment and regulated in
inflammatory-stop signals can modulate neuroin- time-specific manner [56]. The controversial
flammation in PND-models. Using a rat model of roles of astrocytes indicate that these cells may
cardiopulmonary bypass with deep hypothermic play key roles in cognitive function, both in
circulatory arrest treatment with annexin A1 health and diseases. The specific SPMs actions
(ANXA1) was able to improve cognitive out- on astrocytes have not been thoroughly investi-
comes and modulate microglial activation by gated. In pain models, Lipoxin A1 exerts anti-
inhibiting NF-κB p65 transcriptional activity and nociceptive effect by modulation of astrocytic
subsequent cytokine production [42]. Recently, activation [57]. Similarly, AT-RvD1 was shown
we demonstrated that prophylaxis with MaR1 to attenuate TNF-α release from spinal astro-
can rescue microglial activation after orthopedic cytes and improve mechanical hypersensitivity
surgery, as detected by morphological changes- in a rat model of carrageenan-induced peripheral
such as shifting to a more ramified homeo- inflammation [58]. In PND models we described
static morphology [43]. Thus, resolution agonists changes in astrocytes morphology at 24 h after
including SPMs may diminish neuroinflamma- orthopedic surgery [41, 43]. The astrogliosis is
tion in part by regulating microglial phenotype, associated with increased basal glutamatergic
leading to improved cognitive outcomes. To date synaptic transmission, reduced short term plas-
many of the SPMs have not been evaluated in ticity and long-term potentiation in hippocam-
models of PND and surgical recovery, which pus [41]. These pathological changes in
warrants future investigations. astrocytes can be eliminated by prophylaxis with
Aside from microglia, astrocytes also contrib- aspirin triggered resolvin-D1 (AT-RvD1) as low
ute a key role in maintaining CNS homeostasis, as ~0.1 μg/kg [41] or MaR1 at ~4 μg/kg [43].
including regulating brain blood flow [44, 45], The precise mechanisms underlying the effects
synapse function [46], extracellular ion concen- of AT-RvD1 and MaR1 on postoperative astro-
tration [47], and interacting with endothelial gliosis still require further exploration.
cells to support the BBB [48]. Notably, a role for Accumulated evidences suggest the pro-inflam-
immune-regulation has emerged from these cells matory activity of astrocytes may be regulated
[49]. As an active immune regulator in CNS, by microglia [59] via cytokines, chemokine,
astrocytes also sense stimulation and danger sig- complement activation [60], growth factors, and
nals, responding by releasing glia-transmitters other signaling molecules [61]. After surgery the
and communicating to neurons [50]. Together CCL2/CCR2 axis has been implicated in the
with microglia they also contribute to further neuroinflammatory response; Xu et al. showed
activating adaptive immune defense [51] [49]. that increased CCL2 expression in astrocytes is
Following brain injury or in neurodegenerative sufficient to activate microglia and cause learn-
diseases, astrocytes undergo pronounced trans- ing impairments [62]. Therefore, regulation of
formation called “astrogliosis”. Astrogliosis astrogliosis may indirectly regulate microglia
changes the cellular expression and morphology activity and targeting cell-specific interactions
of astrocytes and contribute to the repair and may result into effective therapies for different
scarring process in CNS [52]. However, this neurological conditions, including PNDs
reactive phenotype has also been suggested to (Figs. 4.1 and 4.2).
become detrimental by upregulating the expres- Intrinsic to the postoperative neuroinflamma-
sion of IL-17 receptor and sphingosine 1-phos- tion is the activation of the peripheral innate
phate (S1P) [53, 54]. This further triggers the immune system. Sterile injury, as during surgery,
production of pro-inflammatory cytokines and rapidly triggers the release of TNFα and alarmins
chemokines, which can lead to exacerbated neu- into the circulation. TNFα can initiate a pro-
roinflammation and neurodegeneration [55, 56]. inflammatory cytokine cascade that eventually
During neuroinflammation, the status of astro- impairs the BBB homeostasis. The BBB dys-
4 The Evolving Role of Specialized Pro-resolving Mediators in Modulating Neuroinflammation… 31
function in turn facilitates the migration of LPS-induced TNFα release, NF-κB nuclear
macrophages into the hippocampus [6, 12, 41, translocation, superoxide generation and M1-like
43]. Inhibition of the TNFα signaling by anti- phenotype surface markers expression in primary
TNFα antibody or genetic abrogation of bone marrow derived macrophages [43]. These
macrophage-specific IKKβ prevent postoperative modulatory effects on macrophage function/phe-
BBB disruption and macrophage infiltration in notype may be a key mechanism for SPMs to pre-
the hippocampus [6, 12]. This work demon- vent surgery induced BBB disruption, ensuing
strated the primary impact of innate immune sys- neuroinflammation, and cognitive decline [43].
tem and systemic inflammation on the Importantly, because SPMs exert both anti-
development of neuroinflammation and cognitive inflammatory and pro-resolving effects, they are
decline. During resolution of inflammation, crucial to terminate inflammation but also stimu-
SPMs stimulate the cessation of PMN influx [22, late tissue repair [65], which is fundamental in
34] and macrophage clearance of cellular and the context of perioperative recovery. In fact,
toxic debris [16, 20, 63]. The regulation of mac- RvD1 and maresins have shown to accelerate
rophage phagocytosis function by SPMs may be wound healing in diabetic patients [66, 67].
mediated by the switch of macrophage pheno- AT-RvD1 delivered through nanoparticles also
type [16]. For example, Dalli et al. demonstrated enhance wound healing in a mice model of peri-
that 10 nM of MaR1 or RvD1 lead to significant tonitis [68]. We recently demonstrated that MaR1
reductions in CD54 and CD80 expression and a pretreatment boosts systemic levels of IL-10 with
concomitant up- regulation of CD163 and a long- lasting trend up to 14 days after sur-
CD206 in human macrophage [64]. Moreover, gery [43]. Further, we found no difference in cal-
M2-like macrophages are more efficient in con- lus formation in mice treated with MaR1
verting DHA into MaR1 [64]. Similarly, our in compared to vehicle, suggesting MaR1 may be a
vitro work indicated 10 nM MaR1 can inhibit safe option to be tested in future clinical trials.
BBB CNS
TNF
IL-1 GFAP+
IL-6 cld-5
IL-12
CXCL1
Iba-1+
MΦ
SPMs
pro-inflammatory pro-resolving
Fig. 4.1 Putative mechanisms of surgery-induced neuro- dependent memory function. Treatment with SPMs, such
inflammation and cognitive dysfunction. Surgery triggers as resolvins and maresins, actively promote resolution of
systemic pro-inflammatory cytokines that can impair the inflammation after surgical trauma and prevent central
blood-brain barrier, thus allowing peripheral cells into the nervous system dysfunction through the modulation of
brain parenchyma. Macrophage infiltration activates glia, systemic and central cell types, such as monocytes,
including resident microglia and astrocytes, that acutely microglia, and neurons
affect processes of synaptic plasticity and hippocampal-
32 T. Yang and N. Terrando
Fig. 4.2 Proposed mechanisms for neuroinflammation microglia but also by infiltration of peripheral cells into
and surgery-induced cognitive dysfunction. Surgery has the brain parenchyma via a disrupted blood-brain barrier.
been shown to engage the innate immune system and acti- This pro-inflammatory milieu and glia dysfunction impair
vate a cascade of pro-inflammatory mediators, including neuronal activity and synaptic plasticity, impinging on
alarmins, cytokines and eicosanoids. These molecules processes of long-term potentiation, neurotransmission,
exert effects on the humoral and neuronal signaling over- and receptor function at the synapse. In combination,
all contributing to the neuroinflammatory response. These these pathological hallmarks contribute to learning and
processes are mediated not only by activation of resident memory impairments following surgical trauma
Overall, we are starting to uncover the poten- intervene before an elective surgical procedure.
tial for resolution agonists in the context of peri- SPMs biomarkers may take us a step closer to
operative disorders and surgical recovery. Further personalized approaches and targeted interven-
research is needed to evaluate the exact mecha- tions to treat common complications, like post-
nisms of action of different SPMs on different operative pain and PNDs. As coined by Serhan
CNS cell types, and whether specific mediators and Savill ‘alpha’ programs ‘omega’ (i.e. the
may better target the immune response triggered beginning programs the end) [20] could not bet-
by surgical trauma. SPMs have been detected ter define the importance of the temporal events
in human cerebrospinal fluid, establishing proof- that orchestrate the acute inflammatory
of-principle that may serve as biomarkers for response in the perioperative space. Further char-
disease progression in neurological conditions acterizing these complex molecular events may
like multiple sclerosis [69] and clinical PND provide novel and urgently needed approaches to
[43]. These mediators can also be modified by safely treat PND and overall improve brain
dietary interventions, for example omega-3 health for the millions of patients that undergo
PUFA [70], thus providing attractive strategies to surgery every year.
4 The Evolving Role of Specialized Pro-resolving Mediators in Modulating Neuroinflammation… 33
M. Brianza-Padilla and R. Bojalil
brane, EPA and DHA substitute AA as the sub- erosclerotic plaques represents a risk to develop
strates of lipid mediators. The SPMs derived myocardial infarction and cerebrovascular events
from those n-3 fatty acids, exert powerful anti- [57]. In atherosclerosis, vascular smooth muscle
inflammatory and pro-resolving actions mainly cells have increased proliferative and chemotactic
by decreasing the influx of neutrophils to tissues, features, reduced expression of contractile pro-
by enhancing non-phlogistic recruitment of teins and increased production of proinflamma-
monocytes into tissues, and by decreasing the tory cytokines [4]; they show increased
production of pro-inflammatory mediators [11]. inflammation and oxidative stress, and extensive
Cell membrane surface receptors mediate their areas of necrosis composed of non-clarified apop-
signaling [29, 33]. Annexin A1, a stress response totic cells [19]. Some authors suggest that poor
endogenous protein, also mediates inflammatory resolution of inflammation merges the above [19],
resolution and can decrease the production of hence various researchers have tested diverse
leukotrienes and prostaglandins by an inhibitory experimental approaches to prove this hypothesis.
action on FLPA2 [15]. SPMs delay the progression of atherosclerosis
Annexin
PLP A2 Regulation
PUFA’s Omega 6 Inflammation
Resolution
Intake AA EPA DHA
Omega 3
Thromboxanes Protectins
Maresins
Leukotrienes
Lipoxins
5.4 Resolution and CVD and promote greater plaque stability [18]. A nota-
ble observation is that mouse atherosclerotic
Atherosclerosis is a disease that is a convincing plaque treated with SPMs increases its thickness
example of the consequences of an inefficient and the synthesis of collagen in the fibrous layer,
inflammatory resolution. In humans, having ath- thus gaining stability [18, 19].
40 M. Brianza-Padilla and R. Bojalil
Today there is a great interest in studying the proliferation, migration, and production of
resolvins in cardiac alterations because they seem superoxide anion, and the expression of proin-
to be crucial players in restoring lesions since flammatory genes [3, 59]. The phosphoinositide
they reduce the response of vascular smooth 3-kinase (PI3K)/serine/threonine kinase Akt
muscle cells and decrease the damage through (PI3K/Akt) signaling interactive pathway medi-
the local activation of resolution mechanisms [8]. ates the protective effects of RvD1 (and of lipox-
There are reports that resolvins can reduce the ins), as treating with its inhibitor can block the
risk of developing atherosclerosis and its compli- cardioprotective effect of RvD1 [23]. In a model
cations by preventing platelet aggregation, induc- of damage caused by hepatic ischemia, RvD1
ing vasodilation [13], inducing neutrophil inhibits inflammatory pathways, decreasing the
apoptosis and limiting the size of the infarct [8]. levels of IL-6, TNF, and myeloperoxidase, and
In humans, however, there is still a long way to increases phosphorylation of Akt, which favors
fully assess their therapeutic efficiency and pos- its protective effect. In addition, RvD1 reduces
sible toxic doses. the expression of pro-fibrotic genes and the depo-
Resolvin E1 was the first metabolite of EPA sition of collagen, which reduces post-infarction
identified, its biosynthesis depends on the inter- fibrosis [30]. In ischemic lesions, neutrophils
action between endothelial cells and leukocytes infiltrate and release reactive oxygen species
in the inflammatory process [48]. Several studies (ROS) and other mediators that damage tissue
suggest that RvE1 plays an important role in the integrity [56]. Exogenous administration of
restoration and repair of tissue damage, favoring RvD1 after myocardial infarction in mice reduces
resolution by decreasing the production of pro- both the accumulation of neutrophils and fibro-
inflammatory proteins and platelet aggregation. sis, which induces an improvement in cardiac
Administering RvE1 reduces the size of the function [30].
lesion and decreases aortic expression of tumor RvD2 has effects on cellular recruitment and
necrosis factor (TNF) and IFNγ in a murine proliferation. In an animal model, RvD2 limits
model of atherosclerosis [47]. In a model of peri- the recruitment of neutrophils to the left ventricle
odontitis and atherogenesis in mice, it decreases and reduces the density of macrophages in the
the levels of C-reactive protein (CRP) and the infarcted area [8]. This lipid reduces cell prolif-
infiltration of macrophages to the intima [27]. eration and lymphocyte recruitment in a model of
RvE1 takes part in reducing platelet aggregation arterial angioplasty in rabbits [38]. Similarly, in
induced by thromboxane [20] and, in a model of an intimal arterial neoformation model in mice
ischemia-reperfusion of the myocardium in rats, induced by carotid ligation, RvD2 reduced the
its intravenous administration reduces the size of proliferation of smooth muscle cells and the
the infarct in a dose-dependent manner [31]. recruitment of neutrophils and macrophages. It
The mechanisms of action of resolvins of the also induced lower levels of TNF and of granulo-
D series in the inflammatory resolution of cardiac cyte and macrophage colony-stimulating factor
alterations seem to be different. In a model of (GM-CSF) [3]. Acting in a similar way of RvD2,
arterial damage in rats, treatment with RvD1 the administration of LXA4 in rats with myocar-
attenuates both the levels of oxidative stress and ditis reduces the infiltration of inflammatory cells
the activation of the NFκB signaling pathway [9]. [53].
Intact human arteries incubated with DHA Not being an SPM, but because of its role in
ex vivo, produce precursors of resolvins of the D inhibiting FLPA2, annexin A1 has been studied
series, which inhibit the adhesion of monocytes in atherosclerosis. Asymptomatic plaques of the
induced by stimulation of endothelial cells with carotid artery have elevated levels of the protein,
TNF [9], suggesting that the local production of and its plasmatic concentrations correlate nega-
SPMs is necessary to promote inflammatory res- tively with the total areas of the plaques in ath-
olution. The in vitro treatment with RvD1 and erosclerosis models, which suggests an important
RvD2 of vascular smooth muscle cells decreases role of annexin in resolving cardiac inflammation
5 Relationship Between Specialized Pro-resolving Mediators and Inflammatory Markers in Chronic… 41
[15]. Its action on regulating the production of tion (few months to more than 6 years), the great
leukotrienes and prostaglandins may account for variability in doses (<1–4 g/d), the diseases stud-
its capacity to inhibit the migration of leukocytes ied, comorbidities (diabetes, hypertension and
into the plaques and its early protective effects stroke), and the interaction with drugs. [1, 2].
[19]. To reach effective blood levels to reduce the
risk of CVD or prevent secondary complications,
various studies have concluded that the daily con-
5.5 iet PUFAs as Precursors
D sumption of EPA plus DHA must be of at least
of SPMs and CVD 1 g [5, 36, 44]. Consistently, in studies with lower
doses of PUFAs, no protection against a possible
Independent of the use of pure SPMs in the cur- cardiovascular event is achieved [14]. Some
rent approaches, there is a long history of use of authors suggest higher doses to prevent compli-
their precursors n-6 and n-3 PUFAs derived from cations in patients with cardiac disease, for
the diet, specifically by consuming seafoods or example, consuming 3.36 g of EPA plus DHA in
their purified compounds. Being essential in the patients with stable coronary disease, helps to
development and function of the organism [8], restore the levels of SPMs and promotes the
the human body can synthesize only small phagocytosis of clots. [16]. Administering high
amounts of EPA and DHA [60]. Some studies doses of n-3 PUFAs in patients with atherosclero-
observe that consuming fatty acids changes the sis and type 2 diabetes, accompanied by adequate
profile of the membrane phospholipids, and the medical therapy, increases the serum concentra-
composition of the fatty acids of different cells tions of EPA and DHA, although it does not
and tissues, affecting their availability [32]. Other induce changes in the concentrations of RvD1
studies have shown beneficial effects of n-3 on [45]. In rats, a diet enriched with DHA decreases
various pathological conditions [52]. The quality pro-inflammatory proteins such as CRP, IL-6,
of life of people with chronic inflammatory dis- TNF, and IL-1β, and favors an increase in the
eases such as arthritis and asthma [35], the sever- concentrations of RvD2 and RvD3 [39]. Fish oil
ity of chronic inflammation [7] and the risk for treatment favors protection against thrombosis
cardiovascular diseases [26] improve with a diet and damage induced by vascular remodeling in
with n-3 PUFAs. The incidence of heart failure in mice, reducing local inflammation and increasing
older adults associates negatively with the total resolution by increasing RvE1 [24].
plasma concentration of n-3 [40]. Coincidently, a
meta-analysis shows that supplementing with
EPA and DHA has positive effects on risk factors 5.6 Conclusions
for cardiovascular diseases because they have
hypolipemic, hypotensive, antiarrhythmic and Many studies suggest that uncontrolled inflam-
anti-inflammatory actions [1]. mation and failure in the resolution response are
Until today it is not fully clear the role of diets the basis of a myriad of human diseases. SPMs
that include DHA and EPA to protect against car- seem to play a crucial role in restoring damaged
diovascular diseases. There is not a consensus tissue and in the recovery of function. In fact, it is
despite a wide range of studies that show their possible that the success of the inflammatory
beneficial effects, in part because many factors resolution largely depends on the capacity of pro-
are involved and influence the outcome. For ducing SPMs and how these mediators can con-
example, a diet enriched with n-6 and n-3 in a 4:1 tain chronic inflammation. The study of
ratio (as opposed to the 15:1 obtained with a typi- inflammatory resolution in cardiovascular
cal occidental diet) decreases the mortality rate diseases still represents a challenge. It is still nec-
for cardiovascular diseases [55]. Other factors to essary to find out if administering SPMs or their
consider include the different EPA/DHA ratios precursors will have the beneficial therapeutic
used (0.5:1–1.4:1), different lengths of consump- use they promise. Indeed, finding successful
42 M. Brianza-Padilla and R. Bojalil
ways of resolving inflammation can provide 12. Dalli J, Serhan CN (2012) Specific lipid media-
tor signatures of human phagocytes: microparticles
important advances in preventing cardiometa- stimulate macrophage efferocytosis and pro-resolving
bolic diseases. mediators. Blood 120(15):e60–e72. https://doi.
org/10.1182/blood-2012-04-423525
13. Das UN (2008) Can endogenous lipid molecules
serve as predictors and prognostic markers of coro-
References nary heart disease? Lipids Health Dis 7(1):19. https://
doi.org/10.1186/1476-511X-7-19
1. AbuMweis S, Jew S, Tayyem R, Agraib L (2018) 14. de Goede J, Geleijnse JM, Boer JMA, Kromhout D,
Eicosapentaenoic acid and docosahexaenoic acid Verschuren WMM (2010) Marine (n-3) fatty acids,
containing supplements modulate risk factors for car- fish consumption, and the 10-year risk of fatal and
diovascular disease: a meta-analysis of randomised nonfatal coronary heart disease in a large population of
placebo-control human clinical trials. J Hum Nutr Dutch adults with low fish intake. J Nutr 140(5):1023–
Diet 31(1):67–84. https://doi.org/10.1111/jhn.12493 1028. https://doi.org/10.3945/jn.109.119271
2. Ajith TA, Jayakumar TG (2019) Omega-3 fatty acids 15. Drechsler M, de Jong R, Rossaint J, Viola JR, Leoni
in coronary heart disease: recent updates and future G, Wang JM et al (2015) Annexin A1 counteracts
perspectives. Clin Exp Pharmacol Physiol 46(1):11– chemokine-induced arterial myeloid cell recruitment.
18. https://doi.org/10.1111/1440-1681.13034 Circ Res 116(5):827–835. https://doi.org/10.1161/
3. Akagi D, Chen M, Toy R, Chatterjee A, Conte MS CIRCRESAHA.116.305825
(2015) Systemic delivery of proresolving lipid media- 16. Elajami TK, Colas RA, Dalli J, Chiang N, Serhan CN,
tors resolvin D2 and maresin 1 attenuates intimal Welty FK (2016) Specialized proresolving lipid medi-
hyperplasia in mice. FASEB J 29(6):2504–2513. ators in patients with coronary artery disease and their
https://doi.org/10.1096/fj.14-265363 potential for clot remodeling. FASEB J 30(8):2792–
4. Bennett MR, Sinha S, Owens GK (2016) Vascular 2801. https://doi.org/10.1096/fj.201500155R
smooth muscle cells in atherosclerosis. Circ 17. Frantz S, Nahrendorf M (2014) Cardiac macrophages
Res 118(4):692–702. https://doi.org/10.1161/ and their role in ischaemic heart disease. Cardiovasc
CIRCRESAHA.115.306361 Res 102(2):240–248. https://doi.org/10.1093/cvr/
5. Breslow JL (2006) N−3 fatty acids and cardiovascular cvu025
disease. Am J Clin Nutr 83(6):1477S–1482S. https:// 18. Fredman G, Hellmann J, Proto JD, Kuriakose G,
doi.org/10.1093/ajcn/83.6.1477S Colas RA, Dorweiler B et al (2016) An imbalance
6. Calabrò P, Golia E, Maddaloni V, Malvezzi M, between specialized pro-resolving lipid mediators and
Casillo B, Marotta C, Golino P (2009) Adipose pro-inflammatory leukotrienes promotes instability of
tissue-mediated inflammation: the missing link atherosclerotic plaques. Nat Commun 7(1):12859.
between obesity and cardiovascular disease? Intern https://doi.org/10.1038/ncomms12859
Emerg Med 4(1):25–34. https://doi.org/10.1007/ 19. Fredman G, Spite M (2017) Specialized pro-resolving
s11739-008-0207-2 mediators in cardiovascular diseases. Mol Asp Med
7. Calder PC, Grimble RF (2002) Polyunsaturated 58:65–71. https://doi.org/10.1016/j.mam.2017.02.003
fatty acids, inflammation and immunity. Eur J Clin 20. Fredman G, Van Dyke TE, Serhan CN (2010)
Nutr 56(Suppl 3):S14–S19. https://doi.org/10.1038/ Resolvin E1 regulates adenosine diphosphate acti-
sj.ejcn.1601478 vation of human platelets. Arterioscler Thromb Vasc
8. Capó X, Martorell M, Busquets-Cortés C, Tejada S, Biol 30(10):2005–2013. https://doi.org/10.1161/
Tur JA, Pons A, Sureda A (2018) Resolvins as pro- ATVBAHA.110.209908
resolving inflammatory mediators in cardiovascular 21. Frostegård J (2013) Immunity, atherosclerosis and
disease. Eur J Med Chem 153:123–130. https://doi. cardiovascular disease. BMC Med 11(1):117. https://
org/10.1016/j.ejmech.2017.07.018 doi.org/10.1186/1741-7015-11-117
9. Chatterjee A, Komshian S, Sansbury BE, Wu B, 22. Fujiu K, Nagai R (2013) Contributions of cardio-
Mottola G, Chen M, Conte MS (2017) Biosynthesis myocyte–cardiac fibroblast–immune cell inter-
of proresolving lipid mediators by vascular cells actions in heart failure development. Basic Res
and tissues. FASEB J 31(8):3393–3402. https://doi. Cardiol 108(4):357. https://doi.org/10.1007/
org/10.1096/fj.201700082R s00395-013-0357-x
10. Chin BSP, Blann AD, Gibbs CR, Chung NAY, Conway 23. Gilbert K, Bernier J, Bourque-Riel V, Malick M,
DG, Lip GYH (2003) Prognostic value of interleukin- Rousseau G (2015) Resolvin D1 reduces infarct size
6, plasma viscosity, fibrinogen, von Willebrand factor, through a phosphoinositide 3-kinase/protein kinase
tissue factor and vascular endothelial growth factor B mechanism. J Cardiovasc Pharmacol 66(1):72–79.
levels in congestive heart failure. Eur J Clin Investig https://doi.org/10.1097/FJC.0000000000000245
33(11):941–948 24. Gong Y, Lin M, Piao L, Li X, Yang F, Zhang J et al
11. Dalli J, Colas RA, Serhan CN (2013) Novel n-3
(2015) Aspirin enhances protective effect of fish
Immunoresolvents: structures and actions. Sci Rep oil against thrombosis and injury-induced vascular
3(1):1940. https://doi.org/10.1038/srep01940
5 Relationship Between Specialized Pro-resolving Mediators and Inflammatory Markers in Chronic… 43
48. Schwanke RC, Marcon R, Bento AF, Calixto JB (2016) 54. Sica A, Mantovani A (2012) Macrophage plastic-
EPA- and DHA-derived resolvins’ actions in inflam- ity and polarization: in vivo veritas. J Clin Investig
matory bowel disease. Eur J Pharmacol 785:156–164. 122(3):787–795. https://doi.org/10.1172/JCI59643
https://doi.org/10.1016/j.ejphar.2015.08.050 55. Simopoulos AP (2008) The importance of the Omega-6/
49. Serhan CN (2014) Pro-resolving lipid mediators are Omega-3 fatty acid ratio in cardiovascular disease and
leads for resolution physiology. Nature 510(7503):92– other chronic diseases. Exp Biol Med 233(6):674–
101. https://doi.org/10.1038/nature13479 688. https://doi.org/10.3181/0711-MR-311
50. Serhan CN, Chiang N, Dalli J (2015) The resolution 56. Swirski FK, Nahrendorf M (2013) Leukocyte behav-
code of acute inflammation: novel pro-resolving lipid ior in atherosclerosis, myocardial infarction, and
mediators in resolution. Semin Immunol 27(3):200– heart failure. Science 339(6116):161–166. https://doi.
215. https://doi.org/10.1016/j.smim.2015.03.004 org/10.1126/science.1230719
51. Serhan CN, Chiang N, Van Dyke TE (2008)
57. Virmani R, Burke AP, Farb A, Kolodgie FD (2006)
Resolving inflammation: dual anti-inflammatory and Pathology of the vulnerable plaque. J Am Coll
pro-resolution lipid mediators. Nat Rev Immunol Cardiol 47(8):C13–C18. https://doi.org/10.1016/j.
8(5):349–361. https://doi.org/10.1038/nri2294 jacc.2005.10.065
52. Shaikh NA, Yantha J, Shaikh S, Rowe W, Laidlaw 58. Wang A, Luan HH, Medzhitov R (2019) An evolu-
M, Cockerline C et al (2014) Efficacy of a unique tionary perspective on immunometabolism. Science
omega-3 formulation on the correction of nutritional 363(6423):eaar3932. https://doi.org/10.1126/science.
deficiency and its effects on cardiovascular disease aar3932
risk factors in a randomized controlled VASCAZEN® 59. Wu B, Mottola G, Chatterjee A, Lance KD, Chen
REVEAL trial. Mol Cell Biochem 396(1–2):9–22. M, Siguenza IO et al (2017) Perivascular delivery of
https://doi.org/10.1007/s11010-014-2132-1 resolvin D1 inhibits neointimal hyperplasia in a rat
53. Shi Y, Pan H, Zhang H-Z, Zhao X-Y, Jin J, Wang model of arterial injury. J Vasc Surg 65(1):207–217.
H-Y (2017) Lipoxin A4 mitigates experimental e3. https://doi.org/10.1016/j.jvs.2016.01.030
autoimmune myocarditis by regulating inflam- 60. Yadav NS, Wierzbicki A, Aegerter M, Caster CS,
matory response, NF-κB and PI3K/Akt signal- Pérez-Grau L, Kinney AJ et al (1993) Cloning of
ing pathway in mice. Eur Rev Med Pharmacol Sci higher plant omega-3 fatty acid desaturases. Plant
21(8):1850–1859 Physiol 103(2):467–476
Specialized Pro-resolving
Mediators Directs Cardiac Healing 6
and Repair with Activation
of Inflammation and Resolution
Program in Heart Failure
Ganesh V. Halade and Bochra Tourki
Abstract Keywords
After myocardial infarction, splenic leukocytes Cardiac repair · Heart failure · Leukocytes ·
direct biosynthesis of specialized pro-resolving Myocardial infarction · Resolution of
mediators (SPMs) that are essential for the res- inflammation · Specialized pro-resolving
olution of inflammation and tissue repair. In a mediators
laboratory environment, after coronary ligation
of healthy risk free rodents (young adult mice)
leukocytes biosynthesize SPMs with induced
activity of lipoxygenases and cyclooxygenases,
which facilitate cardiac repair. Activated mono-
cytes/macrophages drive the biosynthesis of Abbreviations
SPMs following experimental myocardial
infarction in mice during the acute heart failure. AA Arachidonic acid
In the presented review, we provided the recent AT-LXA4 Aspirin-triggered LXA4
updates on SPMs (resolvins, lipoxins and DHA docosahexaenoic acid
maresins) in cardiac repair that may serve as EPA eicosapentaenoic acid
novel therapeutics for future heart failure ther- H and E hematoxylin and eosin
apy/management. We incorporated the under- HF heart failure
lying causes of non-resolving inflammation LV left ventricle
following cardiac injury if superimposed with LX4 lipoxin A4
obesity, hypertension, diabetes, disrupted circa- LXB4 lipoxin B4
dian rhythm, co-medication (painkillers or MaR1 maresin 1
oncological therapeutics), and/or aging that MaR2 maresin 2
may delay or impair the biosynthesis of SPMs, MI myocardial infarction
intensifying pathological remodeling in heart RvD1 resolvin D1
failure. RvD4 resolvin D4
Fig. 6.1 Heart attack induced left ventricle (LV) 1 to day 56 compared to day 0 naïve control. Hematoxylin
structural and fibrotic remodeling from acute (day 1 and Eosin (H and E; 40X) staining of LV showing the
and 5) to chronic (day 28–56) heart failure in mice organized myocardium structure before MI compared to
model of permanent coronary ligation. From the top to necrotic and apoptotic disorganized myocardium from
the bottom: difference in ECG from normal mice to day 5 to day 56; Picrosirius red (PSR) staining confirmed
MI-induced heart after coronary ligation; Periodic acid– the deposition of collagen and compact fibrotic remodel-
Schiff (PAS; 1.25X) staining of the LV after MI from day ing in the infarcted area initiated at day 5 and progressive
to day 56
6 SPMs and Cardiac Repair 47
properly treat and identify novel target signaling out signal’). The synchronicity of leukocyte
pathways to resolve inflammation without the entry, coordinated phagocyte activity and an on
adverse effects of medication or therapies, which time exit prevents undesirable consequences of
frequently occurs with current anti-inflammatory an excessive inflammatory process through the
treatments [8, 9]. Aspirin, often considered the resolution process, which enhances the healing
safest anti-inflammatory treatment, has failed to and tissue repair mechanism [15].
limit the event of coronary heart disease and Although HF is a multifactorial and hetero-
stroke in elderly individuals and is often associ- geneous end stage disease, the mechanisms of
ated with high incidences of intracranial and initial inflammation and subsequent initiation
extracranial bleeding within the same risk group of tissue repair are relatively undefined in the
[10]. acute setting of MI and the role of these critical
In this review, we summarized the leukocyte- pathways in the chronic setting remains largely
directed biosynthesis of SPMs in myocardial unexplored [16]. HF is predominantly superim-
healing and the beginning of future applications posed on other risk factors, like aging and renal
of novel SPMs to limit chronic inflammation in failure; however, it is not only risk factors that
the setting of HF with potential enhancement to amplify the complexity of this disease. There is
the reparative program [11]. SPMs are biosynthe- an inter-organ crosstalk between the heart and
sized by leukocytes triggering endogenous car- peripheral organs in HF that delays the cardiac
diac healing as part of the acute innate response healing program (Fig. 6.1). We defined a clear
involved in the inflammation-resolution process. interaction between the spleen and heart, as ‘the
The successful biosynthesis of SPMs terminates splenocardiac axis’ as well as the cardio-renal
inflammation and facilitates repair by stimulating axis [17]. In this context, the spleen is not act-
distinct resolution processes necessary for tissue ing as a leukocyte reservoir but instead as a site
repair and regeneration from MI to advanced HF for the continuous activation of SPMs in car-
[12–14]. diac injury. The spleen is considered an integra-
tive organ in cardiac healing through the
production of multiple families of SPMs to acti-
6.2 Immune-Responsive vate the cardiac resolution program because
Systems in Myocardium splenic leukocytes define the resolution of
Wound Healing inflammation in HF [18, 19]. In order to prevent
the progression of HF pathology from acute to
Pathological inflammation and physiological chronic inflammation, the inflammatory
inflammation are two distinct and primary chal- response must be actively resolved (Fig. 6.2). In
lenges in identifying and treating injury, infec- this review, we highlighted the role of leuko-
tion, or stress (physical or neuro-hormonal cytes, particularly macrophages, that biosynthe-
mental exhaustion). Physiological inflammation, size SPMs in cardiac repair and remodeling
although necessary, needs to be both activated after injury.
and inactivated within a specified time in order to
avoid damage caused by sustained pathological
inflammation. Distinct differences include the 6.3 Immune-Responsive
phenotype and/or biomarker of injured cells and and Leukocytes Directed
tissues and the nature and timing of immune sig- Cardiac Repair
nals. Successful physiological inflammation
depends on the infiltration of neutrophils and Cardiac repair is based on the timely activation
monocytes (‘get-in signal’), phagocytic activity of cellular and molecular pathways that delay
of leukocytes (‘eat-me signal’), and the on time reactive fibrosis signaling, resulting in the forma-
departure, or efferocytosis, of neutrophils (‘get tion of scar tissue [20]. Both innate and adaptive
48 G. V. Halade and B. Tourki
Fig. 6.2 SPMs balance or imbalance defines the enzymes (Jadapalli JK et al., AJP-HC 2018). Likewise,
inflammation and resolution outcome. Resolution of pain-killer caprofen, when administrated before MI for
post-MI inflammation is an active process that requires a 2 weeks to mice, impairs the splenocardiac axis, dysregu-
balance between proresolving mediators (SPMs) and lated resolving mediators, over activates neutrophils, and
clearance of leukocytes after acute inflammatory response. decrease the reparative macrophages at the site of the
In case of heart failure associated to co-medication, this injury therefore non-resolving inflammation (Halade GV
balance is impaired. For example doxorubicin, an onco- et al., J Leukoc Biol 2018). LV Left Ventricle, MI
logical drug induced a spleen atrophy with a decrease of Myocardial Infarction, MØ macrophages, Neu
splenic macrophages that impairs SPMs biosynthesizing neutrophils
immune mechanisms are involved in this process of HF [24]. Following cardiac or other tissue
[21]. Recently, the belief that activated splenic injury, the resolution of inflammation is a prime
leukocytes play a critical role in both repair and indicator and precursor of tissue repair [25]. In a
remodeling has been validated. Specialists in clinical setting, the hematological profile of a HF
adaptive immune responses demonstrated that patient shows elevated levels of leukocytes, par-
the depletion of mature B lymphocytes impeded ticularly neutrophils and inflammation of the
monocyte mobilization, limited myocardial arterial wall in addition to the infarcted myocar-
injury, and improved heart function post-MI and dium [26, 27]. In mice, the density of leukocytes
Treg function in cardiac repair [22, 23]. However, (neutrophils/monocytes) increase in the infarcted
cardiac repair alone is insufficient due to various LV in response to myocardial injury [28]. This
additional risk factors, such as obesity, insulin spike is due to the depletion of splenic leukocytes
resistance, diabetes, dyslipidemia, and hyperten- that occurs while a steady flow of leukocytes are
sion. These risk factors can hinder healing, pro- provided to the infarcted LV after MI [18]. In
moting defective tissue repair or unresolved fact, splenic leukocytes mobilize to injured myo-
inflammation that can progress to adverse ven- cardium enriched with fatty acids, such as ara-
tricular remodeling thereby leading to end stage chidonic acid (AA), docosahexaenoic acid
6 SPMs and Cardiac Repair 49
[42, 43]. According to macrophage biology found in resolving exudates referred to as ‘reso-
research, many studies are devoted to the descrip- lution phase macrophages’. These macrophages
tion of mouse cardiac monocyte and macrophage possess characteristics of both M1 and M2 cells
subsets and the refinement of macrophage clas- and we believe that they represent cardiac repara-
sifications are ongoing [44]. With respect to tive macrophages, especially with the recent clin-
recent data, the composition of macrophage sub- ical strategies focused on improving macrophage
sets in the myocardium undergo dynamic changes function to improve healing outcomes [55–57].
throughout the course of a lifetime [45]. Fate Macrophage biology and phagocytic systems
mapping studies confirmed that the monocyte/ likely vary with the magnitude of injury and
macrophage systems placed in the heart prior to therefore the monocyte and macrophage
birth maintain self-renewal properties indepen- responses to injury can differ depending on the
dently of a circulation feedback system [46]. experimental model. For example, in a model of
Unfortunately, this discovery has limited transla- neonatal mice, the depletion of macrophages
tional potential since it is still unclear whether after MI impaired cardiac function and angiogen-
generation and differentiation of macrophages esis. The heart can fully regenerate without scar-
from precursors are impaired with age. However, ring following MI in neonatal mice; however, the
it is well proven that macrophages become either regenerative capacity of macrophages is lost
partially or totally dysfunctional in wound heal- 7 days after birth [28]. Under other conditions,
ing within the context of obesity and diabetes like in a model of adult mice, after MI, Ly6Chi
superimposed on aging in both humans and macrophages (M1 macrophages), and Ly6Clow
rodents [47]. (M2 macrophages) are the main effectors of car-
We know that macrophages have polarization diac remodeling.
profiles similar to neutrophils; within 1 day post- The kinetic and detailed macrophage and neu-
MI, macrophage exhibit a pro-inflammatory M1 trophil profiling studies demonstrated that after
profile [48–50]. At the post-MI “healing stage” cardiac injury, splenic monocyte-enriched leuko-
(up to 7 days post-MI), there is a transition from cytes mobilized to the infarct area and biosynthe-
predominately M1 macrophages to primarily sized SPMs. Macrophages are a major contributor
anti-inflammatory macrophages, and then to in SPMs biosynthesis [58]. In addition, lipid
reparative M2 cells [51, 52]. Cardiac reparative mediator profiling of the spleen indicates that the
M2-like macrophages are a complicated mixture spleen also receives feedback signal for biosyn-
of heterogeneous subsets that have strengthened thesis of lipid mediators, which promote self-
their tissue-repairing abilities by upregulating resolution of inflammation at the left ventricular
various anti-inflammatory and repair-associated site [59]. In cardiac injury, SPMs are biosynthe-
genes after MI [42, 53]. These two types of cells sized immediately by primarily activated macro-
(M1 and M2) are derived from Ly6Chi monocytes phages with the initiation of inflammation [60].
and Ly6Clow monocytes, respectively. Reparative In response to cardiac injury or stress, the
M2 macrophages are recruited simultaneously resolution-phase activated macrophages also
with inflammatory leukocytes in the post-infarct express ALOX-15, indicating that they may also
LV to mediate myocardial healing through the contribute to SPMs production during resolution
secretion of anti-inflammatory cytokines and [61, 62]. M2 macrophages produce more pro-
growth factors, such as VEGF and TGF-ß, that resolving lipid mediators than M1 macrophages
contribute to myofibroblast activation and neoan- [63]. SPMs initiate the switch from inflammation
giogenesis [54]. Deceased cardiac fibroblasts to resolution by reducing neutrophil recruitment
emit signals causing the activation of monocytes and T cell cytokine production and increasing
via MCP-1-mediated chemotaxis and adhesion to recruitment of phagocytic monocytes [64].
ICAM-1/VCAM-1, and induce their differentia- Resident macrophages express TGF-β1 and
tion to M1 or M2 macrophages [44]. Some IL-10, presumably because of phagocytizing
researches include a third macrophage subtype apoptotic neutrophils; these cells also expressed
6 SPMs and Cardiac Repair 51
ALOX-15 and TIMD4 (T-cell immunoglobulin phocytes [72]. Although data on SPMs primarily
domain and mucin domain 4) to facilitate the rec- focuses on innate immune cells involved in the
ognition and uptake of apoptotic cells [65, 66]. resolution of acute inflammation, resolution cre-
Recent results indicate that treatment with DHA ates a microenvironment conducive for the opti-
derived pro-resolving mediators, like exogenous mal development, tissue repair, and modulation
RvD1, stimulate a switch in macrophage pheno- of the adaptive immunity [19, 58, 73]. We con-
type from pro-inflammatory to pro-resolving, a clude that a better understanding of SPMs bio-
M2-like phenotype [67, 68]. Studies found that synthesis and the pharmacology of SPMs
human M2 macrophages are associated with receptors, especially in chronic inflammatory set-
increased MaR1 levels [61]. This increase is due tings like HF, could minimize the gap between
to the ability of this macrophage subtype to con- the initiation of inflammation and initiation of
vert the 13S, 14S epoxide intermediate to MaR1 resolution, capitalizing the fundamental actions
[60]. As critical regulators of the resolution pro- of these effectors of resolution.
gram, targeting the actions of macrophages could
be an effective strategy to control inflammation
and cardiac remodeling. 6.4 SPMs Biosynthesis
and Resolution
of Inflammation in Cardiac
6.3.3 Knowledge Gap Repair
of Inflammation
and Resolution as Part Splenic leukocytes mobilize to the site of
of Cardiac Reparative infarcted injury and biosynthesize SPMs to limit
Program the excessive inflammation that can be detrimen-
tal to healthy tissues, particularly in sterile
After cardiac injury, the resolution phase is an inflammation. In addition to the stop signal in
active biosynthetic process that overlaps or coin- acute inflammatory response, SPMs also stimu-
cides with the first response of the innate immune late distinct processes necessary for tissue repair
system. This was confirmed using structural elu- and regeneration [74]. SPMs have been associ-
cidation of the SPMs in a variety of wound heal- ated with positive feedback loops, where one
ing models [19]. Inflammation-resolution pro-resolving mediator induces the biosynthesis
process-derived bioactive metabolites implicate of another [15]. The administration of RvD1 3 h
an array of receptors that transduce pro-resolving after MI in mice induces lipoxin A4 and MaR1 to
action. Among them, the most studied receptors facilitate the resolution of inflammation and car-
are GPCRs as effectors of resolution shown indi- diac regeneration [75]. SPMs are produced
rectly through use of the receptor knockout cells mainly by the inter-organ interactions of
and mice. These GPCR receptors (ChemR23, monocyte-derived macrophages and neutrophils
GPR32, and FPR2) activate signals and trans- via distinct enzymatic pathways from essential
duce the pro-resolving signals of chemerin pep- fatty acids, such as omega-3 and omega 6, poly-
tides, resolvin E1 (RvE1) and resolvin D1 unsaturated fatty acids (PUFAs), eicosapentae-
(RvD1) and lipoxins [69–71]. noic acid (EPA), and docosahexaenoic acid
Recent, emerging data added confirmation to (DHA) [76]. Several endogenous SPMs have
previous thoughts that resolution is a primarily, been discovered, including lipoxins, resolvins,
active stage of the immune response to cardiac protectins, and maresins [36], which are directly
injury and not an inactive, secondary stage. This and indirectly involved in driving the initiation
stage acts as an overlap with the innate response, of inflammation-resolution and successfully ter-
adding a third phase after the initiation of inflam- minating inflammation. Healthy and risk
mation and resolution, creating a post-resolution free animals, such as young or adult mice, effec-
response dominated by macrophages and lym- tively mobilize circulating neutrophils, platelets,
52 G. V. Halade and B. Tourki
and monocytes/macrophages from the spleen changes of ECM in acute HF suggest that RvD1
after cardiac injury to promote left ventricle orchestrates mature scar formation and stabilizes
healing, which is essential to delay HF [59]. the ECM by resolving inflammation. An experi-
After cardiac injury, monocytes/macrophages mental model of hind limb ischemia (HLI) in
serve as a major contributor for the biosynthesis mice, RvD2 stimulated arteriogenic revascular-
of SPMs that have multiple cellular targets in the ization suggesting that resolvins may be a novel
inflammatory response, including immune cells, class of mediators that both resolve inflammation
platelets, and vascular cells [74]. Leukocyte- and promote arteriogenesis [84] (Table 6.1).
directed SPMs actively counter-regulate and Presented experimental studies in rodent models
control the production of pro-inflammatory strongly indicate that SPMs offer pharmacologi-
mediators, including cytokines, leukotrienes, and cal action in the range of ng or pg doses sugges-
eicosanoids, and regulate leukocyte trafficking tive of outstanding potency [85], in contrast to
and phenotype after both sterile and infectious nonsteroidal anti-inflammatory drugs (NSAID)
challenges [77]. which effective in a range of mg/kg doses that are
immune suppressive and impact the cardiovascu-
lar physiology and pathobiology [86].
6.4.1 Resolvins SPMs are produced by the coordinated activ-
ity of lipoxygenases (LOXs) in the heart and
These are novel bioactive autacoids termed cyclooxygenases (COXs) in the spleen; these are
“resolvins” because they are endogenously gen- the key enzymes for generating potent bioactive
erated by resolution phase interaction products lipid mediators, which primarily synchronizes
classified as either E-series resolvins, if leuko- the resolution of inflammation in pathophysio-
cyte directed biosynthesis is initiated from EPA, logical settings [87]. Fredman et al. reported that
or D-series resolvins, when resolvins are biosyn- RvD1 favors synthesis of the pro-resolving lipid
thesized from DHA [78, 79]. Subcutaneous mediator, LXA4, via limiting nuclear transloca-
administration of RvD1 limits the activation of tion of 5-LOX in atherosclerotic inflammation
neutrophils in the spleen and promotes the clear- [88]. RvD1 critically balanced LOX enzymes by
ance of macrophages and neutrophils from the stimulating 5-LOX expression, which is essen-
infarcted site to facilitate cardiac healing. After tial for wound healing post-MI [89]. RvD1 has
ischemic injury, treatment of RvD1 primarily the potential to delay HF by limiting cytoplas-
activates the ALX/FPR2 receptor in both the mic to nucleus translocation to delay atheroscle-
spleen and left ventricle to promote effective res- rotic inflammation and facilitate neutrophil
olution of inflammation and limit residual time of clearance. Future long-term studies focused on
neutrophils at the site of infarction (‘get-out’ sig- RvD1 are warranted in order to prove its utiliza-
nal) [30, 80]. tion in chronic HF management in patients with
A reperfusion study in rats confirmed that heterogeneous cardiovascular pathology [75].
intraventricular administration of RvD1 reduces Another compound of the D-series resolvins
myocardium infarct size; however, presence of family, RvD3, was used as a model of self-
excess linoleic acid attenuates the myocardial resolving peritonitis in aging mice. The study,
protection offered by RvD1 [81, 82]. In another performed by Arnardottir and colleagues,
experimental study, after cardiac injury using reported that increased levels of RvD1 and
permanent coronary ligation model, RvD1 RvD3 treatment stimulated increases in mono-
administration in mice not only resolves the cyte-accelerated resolution of acute inflamma-
inflammatory response, but also reduced the tion via efferocytosis [90]. RvD3 has been
expression of fibrotic genes, such as colla1, argued to be one of the most potent of the
coll2a1 and tnc, in the early phase of cardiac D-series resolvins [91].
healing. Thus, RvD1 limits fibrotic signaling and Regarding the E-series resolvins, Arita et al.
facilitate organ homeostasis [75, 83]. These reported leukocyte-directed actions of RvE1 as
6 SPMs and Cardiac Repair 53
one of the local mediators of tissue homeostasis nanomolar concentrations, inhibiting chemokine-
during inflammation-resolution [92, 93]. The driven recruitment of both granulocytes and
physiological functions of RvE1, and RvE2, both monocytes [98, 99]. At the site of inflammation,
biosynthesized by neutrophils via the 5LOX lipoxins also stimulate macrophages to ingest
pathway and RvE3 biosynthesized by 12/15- and clear apoptotic neutrophils, exerting pro-
LOXs in eosinophils, have been studied in the resolving actions through the activation of ALX/
last few years [94, 95]. The myocardial protective FPR2 [100, 101]. In correlation with lipoxins
action of RvE1 action is quite different from generation, LTB4 was proved higher in the spleen
other resolvins and protectins. RvE1 specifically at a naïve state and increases along with lipoxins
binds to ChemR23 and BLT1 to offer pro- in the LV at day 1 post-MI [18]. Recent studies
resolving responses in human polymorphonu- demonstrated that 15-epi-LXA4 coordinates the
clear (PMN) cells [96]. This binding enhances upregulation of IL-6 and IL-1β at day 1 post-MI
macrophage phagocytosis via phosphoprotein- to facilitate an unaltered, active inflammatory
mediated signaling [97]. It blocks LTB4 binding phase post-MI without altering the acute inflam-
and signals via BLT1 to promote apoptosis of matory response. Liposomal delivery of 15-epi-
PMN for their clearance by macrophages, while LXA4 showed a similar protective function as
LTB4-BLT1 signals PMN survival. free 15-epi-LXA4, improving ventricular func-
tion through the activation of neutrophil clear-
ance in the LV during the resolving phase with an
6.4.2 Lipoxins increase of ALX/FPR2 and Ccl2 during the acute
inflammatory phase in a murine model of HF
Lipoxins A4 and B4 are lipoxygenase-interaction [102]. Additionally, 15-epi-LXA4 promotes the
products of arachidonic acid and are biosynthe- resolution of inflammation by activating GPR120
sized at the site of injury to facilitate resolution of and inhibiting GPR40 in a translational HF model
inflammation. Lipoxins are endogenous that (Table 6.1) [3, 102, 103].
offers anti-inflammation and pro-resolution at
54 G. V. Halade and B. Tourki
receptors. It has been proven that RvD1, RvD2, local adipokine production and monocyte accu-
and MaR1 exert a non-cytotoxic regulatory role mulation in obesity-induced adipose inflamma-
on cells; it represents a promising beginning for a tion [90]. However, it has been demonstrated,
new avenue of resolution physiology research. that the formation of SPMs is severely dysregu-
Biosynthesized maresins counter-regulate the lated in inflamed, obese adipose tissue [120]. A
pro-inflammatory cytokines, such as IL-1β, IL-6, deficiency in pro-resolving mediators in obese
and TNF-α. They also regulate nuclear factor adipose tissue within the setting of obesity could
kappa B (NF-κ B) gene products, increase the be the consequence of a structural deficiency in
regulation of T cell de novo synthesis and intra- the tissue content of omega-3-PUFAs, which are
cellular levels of cyclic adenosine monophos- established substrates for SPM biosynthesis
phate, regenerate tissue, and play a role in [122]. Alternatively, the loss of SPMs in obesity
anti-nociceptive action [36, 117]. SPMs might may reflect accelerated tissue SPMs conversion
possibly act on the balance between pathogenic and clearance to inactive further metabolites
TH1/TH17 and tolerogenic Treg cells, which are because 15-PG-dehydrogenase/eicosanoid oxi-
typically altered during chronic inflammation doreductase, the key enzyme in SPM inactiva-
[58]. However, these endogenous pathways of tion, is markedly up-regulated in obese adipose
resolution could be disrupted or dysregulated tissue [123]. Moreover, not only is activated
with co-medication. In fact, treatment with pain- lipoxygenase essential for SPM biosynthesis, but
killer caprofen or oncologic drug doxorubicin also the substrate product of these enzymes is
facilitate splenocardiac resolution deficiency in essential for precise immune responses. A study
mice with cardiac injury, creating non-resolving involving mice found lipidomic analysis showed
inflammation (Fig. 6.2) [118, 119]. Other factors, higher levels of arachidonic acid and
like metabolic dysfunction, are major contribu- 12(S)-hydroxyeicosatetraenoic acid (12-HETE)
tors of SPMs dysregulation. For example, obe- at day 1 post-MI an obese group compared with
sity, hypertension, diabetes, and aging are the non-obese group (Fig. 6.3) [124]. However,
associated with HF and could give an extra rise to studies involving the obesity paradox recently
unresolved chronic inflammation [120]. Next, we provided evidence that both mouse and human
discussed the impact of SPMs dysregulation in adipose tissue have the capacity to generate
cardiometabolic disorder thereby impact on reso- LXA4 [111].
lution physiology.
6.5.2 Diabetes
6.5.1 Obesity
Uncontrolled inflammation plays an essential
The unprecedented, continuous rise in the preva- role in the pathogenesis of diabetes and its asso-
lence of obesity and obesity-related disorders is ciated pathologies, like HF [125]. Also, the key
causally linked to a chronic state of low-grade sequela of adipose tissue inflammation is insulin
inflammation in adipose tissue and many other resistance leading to type 2 diabetes (T2D) [126–
organs [120, 121]. In the case of obesity, the mol- 128]. In this setting, a known complication of
ecules and signaling pathways have dual roles as T2D is impaired wound healing; the administra-
inflammatory mediators as well as regulators of tion of RvD1 to diabetic mice enhances wound
energy storage and metabolism [109]. The on healing, compared with control or vehicle treat-
time resolution of inflammation and the return of ment [129]. Therefore, SPMs may limit factors
this tissue to homeostasis are key components to associated with T2D via its ability to quell
reducing obesity-induced metabolic dysfunc- inflammation and promote adipose and liver tis-
tions. Results indicate that in inflammatory adi- sue homeostasis. RvD1 improved insulin sensi-
pose tissue, RvD1, and RvD2 are potent tivity, reduced adipose inflammation, and reduced
pro-resolving mediators that counteract both steatosis in mouse models of T2D [130, 131]. In
56 G. V. Halade and B. Tourki
Fig. 6.3 Physiological and pathological healing in like obesity and aging that induce a low-grade chronic
mice delineates the effective and defective resolution in state of inflammation with a high levels of COXs and
heart failure. Physiological or pathological healing LOXs-derived pro-inflammatory mediators with low lev-
depends on resolution of inflammation process. els of SPMs and reparative macrophages along with the
Physiological cardiac remodeling is a consequence of an increase of VCAM-1 (vascular cell adhesion molecule 1),
effective resolution, with the production of necessary MPO (myeloperoxidase), CD40 (cluster of differentiation
SPMs at the right time by reparative macrophages. 40), PGs (prostaglandins), LTs (leukotrienes), and TxA2
However, many factors can lead to a defective resolution (thromboxane A2)
addition to resolvins, lipoxins also showed prom- levels of pro-inflammatory mediators, 5-HETE,
inent results in both mice and human and may 12-HETE and 15-HETE, were observed in pre-
have therapeutic potential in the context of diabe- eclamptic placentae, a disorder of a multifactorial
tes associated vascular complications [132]. etiology that compromise maternal and fetal
However, it is unknown if T2D affects the resolu- well-being as well as cardiovascular health later
tion of inflammation and whether treatment with in life [135]. Endothelial and vascular inflamma-
pro-resolving mediators would stimulate resolu- tion also contribute in sustaining “low-grade
tion and ameliorate clinical complications of inflammation” and have shown to be more asso-
T2D such as impaired wound healing. ciated with pulmonary hypertension (PH) [136].
Recent data demonstrated that in vitro RvD1 pre-
vents arterial wall over-activity in human (HPH).
6.5.3 Hypertension This effect appears to be related to a decrease in
Ca2+ sensitivity in HPA smooth muscle cells.
Hypertension remains a significant risk factor for RvD1 also decreases the expression of trans-
the development of HF with various mechanisms membrane protein member 16A (TMEM16A), a
contributing to both systolic and diastolic dys- specific marker of PH [137]. In the same context,
functions [133]. Much less is known about the Jannaway et al. demonstrated that low nanomolar
role of anti-inflammatory molecules in the regu- concentrations of RvE1, RvD1, and RvD2 can
lation of blood pressure in general or in the devel- prevent constriction in rat and human arteries
opment of hypertension [134]. In contrast, higher induced by a thromboxane mimetic [138].
6 SPMs and Cardiac Repair 57
6.5.4 Disruption of Circadian Clock One study measured SPMs in human blood dur-
ing post-exercise recovery. It was demonstrated
Many physiological functions, including leuko- that lipoxins (LXA4 and LXB4), resolvins (RvE1
cyte and platelet responses, are responsive to day and RvD1), and the protectin D1 isomer increased
and light schedules in order to regulate cell and in human serum during the early hours of post-
organ specific circadian clocks. Results from exercise recovery (0–3 h post-exercise). However,
several recent studies demonstrate that many
ibuprofen treatment with exercise reduced the
metabolic functions are controlled by circadian response of pro-resolving lipid mediators to exer-
rhythm genes, enzymes, and proteins [139]. cise [146]. These results are consistent with those
Disturbances to various aspects of these funda- reported earlier by Gangemi et al. in which LXA4
mental mechanisms are thought to be responsible in human urine was found to increase with tread-
for many of the diseases that affect modern soci- mill exercise [147]. In contrast to the former
eties, including cardiovascular and metabolic dis- belief, exercise training in HF patients has proven
orders [140, 141]. In context to cardiac biology, to be safe and has no adverse effect on left ven-
the direct influence of circadian rhythms on tricular remodeling [148]. However, an exercise
SPMs and endogenous mechanisms in the resolu- paradox exists, and recently a study showed that
tion of inflammation remains largely unexplored. exercise alters β-alanine, augments histidyl
A study demonstrated that cardiomyocte-specific dipeptide levels, and scavenges lipid peroxida-
deletion of the circadian clock component, tion products in human skeletal muscle [149].
Bmal1, leads to age-dependent dilated cardiomy-
opathy and decreased lifespan in mice [142, 143].
Additionally, the circadian release of glucocorti- 6.5.6 Aging and Microbiome
coids and their link to downstream anti-
inflammatory and pro-resolution mediator Aging and age-associated chronic and unre-
annexin A1 facilitates the return to homeostasis solved low-grade inflammation, “inflammaging,”
[140]. A recent study suggests that patients with are progressive and all-time predominant factors
risk of coronary vascular disease (CVD) have for CVD [150, 151]. The underlying mechanisms
defective diurnal regulation of vascular n-3 doc- of inflammaging remain of interest, but a plausi-
osapentaenoic acid (DPA) derived resolvins. ble hypothesis for the non-resolving inflamma-
RvDn-3 DPA is involved in regulating peripheral tion in the elderly is a defect in the
blood cell responses and tissue protection. The inflammation–resolution process. The baseline
marked reductions in plasma RvDn-3 DPA dur- pre- and post-MI environments are also altered
ing the early morning hours indicate that altera- with age [94, 152, 153]. A recent study in a con-
tions in biosynthesis due to dysregulation of text of muscle generation showed a correlation
lipoxygenase and may contribute to CVD onset between aging and CCR2. The data explained
and propagation. This finding can be explained that in young WT CCR2−/−, both dysfunctional
by the increase of adenosine, a regulator of macrophages and a pro-inflammatory environ-
5-LOX activity in plasma from patients with ment were observed. Moreover, this study con-
CVD [144]. cludes that CCR2−/− in mice have a positive
feedback loop that promotes inflammaging in
young mice when compared to aging WT mice
6.5.5 Exercise [154]. In humans, urinary lipoxins (LXs) were
decreased in the elderly, resulting in a profound
Exercise limitation is one of the hallmarks of imbalance between pro-resolving LXs and LTs
heart failure, and an increasing degree of intoler- [147]. In line with these results, a study demon-
ance is associated with a poor prognosis [145]. strated that aging mice dysregulate the formation
58 G. V. Halade and B. Tourki
of pro-inflammatory and pro- resolving mole- of pro-resolving mediators and therefore non-
cules with a decrease in LOXs expression, resolving inflammation in cardiovascular dis-
depending on the influx or availability of sub- eases. Future studies are warranted to determine
strates that temper acute inflammatory-resolving the mechanism of action, additional receptors for
phases, leading to the promotion or repression of SPMs, and interactions with lifestyles or medica-
inflammation post-MI (Fig. 6.3) [155]. Aged tions in the cardiac repair program.
mice have increased inflammation, LTs, and
decreased SPMs compared to young mice [91]. A Acknowledgements Authors acknowledge the support
recent, aging, metabolome human study from National Institutes of Health (NIH)-NCCIH (for-
merly known as NCCAM) AT006704, HL132989 and
described that RvD6 levels were decreased in an UAB Pittman Scholar Award to GVH. The authors would
aging individuals, indicating the role of lipid like to thank Servier Medical Art images bank that used to
metabolism in progressive aging [156]. Thus, create the illustrations in Figs. 6.1, 6.2 and 6.3.
there is evidence of defective SPMs in aging but
the mechanisms remain of interest. Compared to
evidence related to aging and inflammation, less References
is known regarding associations between aging
and the microbiome; however, along with age 1. Keulenaer GWB DL (2008) Heart failure with pre-
served ejection fraction. Eur Cardiol 4(1):31–33
related HF, current evidence has found links 2. Tannenbaum S, Sayer GT (2015) Advances in the
between HF in general and specifically HF in pathophysiology and treatment of heart failure
elderly women with alterations in microbial with preserved ejection fraction. Curr Opin Cardiol
composition and function [157]. Since HF has 30(3):250–258
3. Halade GV, Kain V, Ingle KA (2018) Heart func-
been associated with impaired intestinal barrier tional and structural compendium of cardiosplenic
function and bacterial translocation, leading to and cardiorenal networks in acute and chronic heart
inflammatory and immune responses [158]. failure pathology. Am J Physiol Heart Circ Physiol
Studies have shown that significant alterations of 314(2):H255–Hh67
4. Westman PC, Lipinski MJ, Luger D, Waksman R,
the intestinal bacterial microbiome, like a signifi- Bonow RO, Wu E et al (2016) Inflammation as a
cant decrease of Coriobacteriaceae, driver of adverse left ventricular remodeling after
Erysipelotrichaceae and Ruminococcaceae in acute myocardial infarction. J Am Coll Cardiol
the familiar level and Blautia, Collinsella, uncl. 67(17):2050–2060
5. Duarte Vera YC, Caceres Vinueza SV, Daher Nader
Erysipelotrichaceae and uncl. Ruminococcaceae JE, Lara Teran JF (2018) A novel agent in the treat-
at the genus level [159]. Recently, a study showed ment of heart failure with depressed systolic function.
that resolvins biosynthesizing substrates Archivos de cardiologia de Mexico 88(4):287–297
improved gut microbiome in elderly women 6. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK,
Blaha MJ, Cushman M et al (2016) Executive
[160]. summary: heart disease and stroke statistics--2016
update: a report from the American Heart
Association. Circulation 133(4):447–454
6.6 Conclusion 7. Savarese G, Lund LH (2017) Global public health
burden of heart failure. Card Fail Rev 3(1):7–11
8. Halade GV, Kain V, Wright GM, Jadapalli JK (2018)
After cardiac injury, leukocyte-directed biosyn- Subacute treatment of carprofen facilitate spleno-
thesis of SPMs is necessary for cardiac healing in cardiac resolution deficit in cardiac injury. J Leukoc
order to return to a hemostatic state. Leukocyte- Biol 104(6):1173–1186
9. Krishnan V, Booker D, Cunningham G, Jadapalli
derived SPM actions successfully operate the JK, Kain V, Pullen AB et al (2019) Pretreatment
transition from inflammation to resolution. of carprofen impaired initiation of inflammatory-
However, the dysregulation due to physical inac- and overlapping resolution response and promoted
tivity, co-medication, aging or cardiometabolic cardiorenal syndrome in heart failure. Life Sci
218:224–232
disorders like obesity, hypertension, and diabe- 10. McNeil JJ, Wolfe R, Woods RL, Tonkin AM, Donnan
tes, or even disruption of sleep/wake up cycles GA, Nelson MR et al (2018) Effect of aspirin on
impact the resolution process and the production
6 SPMs and Cardiac Repair 59
cardiovascular events and bleeding in the healthy tion, and heart failure. Science (New York, NY)
elderly. N Engl J Med 379(16):1509–1518 339(6116):161–166
11. Banovic M, Pusnik-Vrckovnik M, Nakou E, Vardas 28. Aurora AB, Porrello ER, Tan W, Mahmoud AI,
P (2018) Myocardial regeneration therapy in heart Hill JA, Bassel-Duby R et al (2014) Macrophages
failure: current status and future therapeutic implica- are required for neonatal heart regeneration. J Clin
tions in clinical practice. Int J Cardiol 260:124–130 Invest 124(3):1382–1392
12. Fredman G, Spite M (2017) Specialized pro- 29. Halade GV, Dorbane A, Ingle KA, Kain V, Schmitter
resolving mediators in cardiovascular diseases. Mol JM, Rhourri-Frih B (2018) Comprehensive tar-
Asp Med 58:65–71 geted and non-targeted lipidomics analyses in
13. Buckley CD, Gilroy DW, Serhan CN (2014) failing and non-failing heart. Anal Bioanal Chem
Proresolving lipid mediators and mechanisms in 410(7):1965–1976
the resolution of acute inflammation. Immunity 30. Tourki B, Halade G (2017) Leukocyte diversity in
40(3):315–327 resolving and nonresolving mechanisms of cardiac
14. Kohli P, Levy BD (2009) Resolvins and protectins: remodeling. FASEB J 31(10):4226–4239
mediating solutions to inflammation. Br J Pharmacol 31. Jadapalli JK, Halade GV (2018) Unified nexus of
158(4):960–971 macrophages and maresins in cardiac reparative
15. Sugimoto MA, Sousa LP, Pinho V, Perretti M, mechanisms. FASEB J 32(10):5227–5237
Teixeira MM (2016) Resolution of inflammation: 32. Frangogiannis NG (2014) The inflammatory
what controls its onset? Front Immunol 7:160 response in myocardial injury, repair, and remodel-
16. Dick SA, Epelman S (2016) Chronic heart failure ling. Nat Rev Cardiol 11(5):255–265
and inflammation: what do we really know? Circ 33. Frangogiannis NG (2012) Regulation of the
Res 119(1):159–176 inflammatory response in cardiac repair. Circ Res
17. Jahng JW, Song E, Sweeney G (2016) Crosstalk 110(1):159–173
between the heart and peripheral organs in heart fail- 34. Robinette CD, Fraumeni JF Jr (1977) Splenectomy
ure. Exp Mol Med 48:e217 and subsequent mortality in veterans of the 1939-45
18. Halade GV, Norris PC, Kain V, Serhan CN, Ingle war. Lancet (London, UK) 2(8029):127–129
KA (2018) Splenic leukocytes define the resolu- 35. Elajami TK, Colas RA, Dalli J, Chiang N, Serhan
tion of inflammation in heart failure. Sci Signal CN, Welty FK (2016) Specialized proresolving lipid
11(520):eaao1818 mediators in patients with coronary artery disease
19. Serhan CN (2014) Pro-resolving lipid media- and their potential for clot remodeling. FASEB
tors are leads for resolution physiology. Nature J 30(8):2792–2801
510(7503):92–101 36. Serhan CN (2017) Treating inflammation and infec-
20. Frangogiannis NG (2006) The mechanistic tion in the 21st century: new hints from decoding
basis of infarct healing. Antioxid Redox Signal resolution mediators and mechanisms. FASEB
8(11–12):1907–1939 J 31(4):1273–1288
21. Epelman S, Liu PP, Mann DL (2015) Role of innate 37. Frodermann V, Nahrendorf M (2017) Neutrophil-
and adaptive immune mechanisms in cardiac injury macrophage cross-talk in acute myocardial infarc-
and repair. Nat Rev Immunol 15(2):117–129 tion. Eur Heart J 38(3):198–200
22. Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, 38. Swirski FK (2015) Inflammation and repair in
Sage AP, Guerin C et al (2013) B lymphocytes trig- the ischaemic myocardium. Hamostaseologie
ger monocyte mobilization and impair heart func- 35(1):34–36
tion after acute myocardial infarction. Nat Med 39. Horckmans M, Ring L, Duchene J, Santovito D,
19(10):1273–1280 Schloss MJ, Drechsler M et al (2017) Neutrophils
23. Lai SL, Marin-Juez R, Stainier DYR (2018) orchestrate post-myocardial infarction healing by
Immune responses in cardiac repair and regenera- polarizing macrophages towards a reparative pheno-
tion: a comparative point of view. Cell Mol Life Sci type. Eur Heart J 38(3):187–197
CMLS76(7):1365–1380 40. Ma Y, Yabluchanskiy A, Iyer RP, Cannon PL, Flynn
24. Shinde AV, Frangogiannis NG (2014) Fibroblasts in ER, Jung M et al (2016) Temporal neutrophil polar-
myocardial infarction: a role in inflammation and ization following myocardial infarction. Cardiovasc
repair. J Mol Cell Cardiol 70:74–82 Res 110(1):51–61
25. Aurora AB, Olson EN (2014) Immune modula- 41. Eming SA, Wynn TA, Martin P (2017) Inflammation
tion of stem cells and regeneration. Cell Stem Cell and metabolism in tissue repair and regeneration.
15(1):14–25 Science (New York, NY) 356(6342):1026–1030
26. Madjid M, Fatemi O (2013) Components of the 42. Heidt T, Courties G, Dutta P, Sager HB, Sebas
complete blood count as risk predictors for coronary M, Iwamoto Y et al (2014) Differential contribu-
heart disease: in-depth review and update. Tex Heart tion of monocytes to heart macrophages in steady-
Inst J 40(1):17–29 state and after myocardial infarction. Circ Res
27. Swirski FK, Nahrendorf M (2013) Leukocyte 115(2):284–295
behavior in atherosclerosis, myocardial infarc- 43. Epelman S, Lavine KJ, Beaudin AE, Sojka DK,
Carrero JA, Calderon B et al (2014) Embryonic
60 G. V. Halade and B. Tourki
and adult-derived resident cardiac macrophages 59. Halade GV, Black LM, Verma MK (2018) Paradigm
are maintained through distinct mechanisms at shift – metabolic transformation of docosahexaenoic
steady state and during inflammation. Immunity and eicosapentaenoic acids to bioactives exemplify
40(1):91–104 the promise of fatty acid drug discovery. Biotechnol
44. Gombozhapova A, Rogovskaya Y, Shurupov V, Adv 36(4):935–953
Rebenkova M, Kzhyshkowska J, Popov SV et al 60. Dalli J, Zhu M, Vlasenko NA, Deng B, Haeggstrom
(2017) Macrophage activation and polarization in JZ, Petasis NA et al (2013) The novel 13S,14S-
post-infarction cardiac remodeling. J Biomed Sci epoxy-maresin is converted by human macrophages
24(1):13 to maresin 1 (MaR1), inhibits leukotriene A4 hydro-
45. Weinberger T, Schulz C (2015) Myocardial infarc- lase (LTA4H), and shifts macrophage phenotype.
tion: a critical role of macrophages in cardiac remod- FASEB J 27(7):2573–2583
eling. Front Physiol 6:107 61. Dalli J, Serhan CN (2012) Specific lipid media-
46. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, tor signatures of human phagocytes: micropar-
Breker M et al (2013) Fate mapping reveals origins ticles stimulate macrophage efferocytosis and
and dynamics of monocytes and tissue macrophages pro-resolving mediators. Blood 120(15):e60–e72
under homeostasis. Immunity 38(1):79–91 62. Sansbury BE, Spite M (2016) Resolution of acute
47. Linehan E, Fitzgerald DC (2015) Ageing and inflammation and the role of Resolvins in immu-
the immune system: focus on macrophages. Eu nity, thrombosis, and vascular biology. Circ Res
J Microbiol immunol 5(1):14–24 119(1):113–130
48. Chen B, Frangogiannis NG (2016) Macrophages 63. Marcon R, Bento AF, Dutra RC, Bicca MA, Leite
in the remodeling failing heart. Circ Res DF, Calixto JB (2013) Maresin 1, a proresolving
119(7):776–778 lipid mediator derived from omega-3 polyunsatu-
49. Frangogiannis NG (2015) Emerging roles for mac- rated fatty acids, exerts protective actions in murine
rophages in cardiac injury: cytoprotection, repair, models of colitis. J Immunol 191(8):4288–4298
and regeneration. J Clin Invest 125(8):2927–2930 64. Lannan KL, Spinelli SL, Blumberg N, Phipps RP
50. Honold L, Nahrendorf M (2018) Resident and (2017) Maresin 1 induces a novel pro-resolving phe-
monocyte-derived macrophages in cardiovascular notype in human platelets. J Thromb haemost: JTH
disease. Circ Res 122(1):113–127 15(4):802–813
51. Lindsey ML, Saucerman JJ, DeLeon-Pennell 65. Uderhardt S, Herrmann M, Oskolkova OV,
KY (2016) Knowledge gaps to understand- Aschermann S, Bicker W, Ipseiz N et al (2012)
ing cardiac macrophage polarization following 12/15-lipoxygenase orchestrates the clearance of
myocardial infarction. Biochim Biophys Acta apoptotic cells and maintains immunologic toler-
1862(12):2288–2292 ance. Immunity 36(5):834–846
52. Ma Y, Mouton AJ, Lindsey ML (2018) Cardiac mac- 66. Kobayashi N, Karisola P, Pena-Cruz V, Dorfman
rophage biology in the steady-state heart, the aging DM, Jinushi M, Umetsu SE et al (2007) TIM-1
heart, and following myocardial infarction. Trans and TIM-4 glycoproteins bind phosphatidylserine
Res J Lab Clin Med 191:15–28 and mediate uptake of apoptotic cells. Immunity
53. Shiraishi M, Shintani Y, Shintani Y, Ishida H, Saba 27(6):927–940
R, Yamaguchi A et al (2016) Alternatively activated 67. Hellmann J, Tang Y, Kosuri M, Bhatnagar A, Spite M
macrophages determine repair of the infarcted adult (2011) Resolvin D1 decreases adipose tissue macro-
murine heart. J Clin Invest 126(6):2151–2166 phage accumulation and improves insulin sensitivity
54. Fraccarollo D, Galuppo P, Bauersachs J (2012) in obese-diabetic mice. FASEB J 25(7):2399–2407
Novel therapeutic approaches to post-infarction 68. Titos E, Rius B, Gonzalez-Periz A, Lopez-Vicario
remodelling. Cardiovasc Res 94(2):293–303 C, Moran-Salvador E, Martinez-Clemente M
55. Ariel A, Serhan CN (2012) New lives given by cell et al (2011) Resolvin D1 and its precursor doco-
death: macrophage differentiation following their sahexaenoic acid promote resolution of adipose
encounter with apoptotic leukocytes during the reso- tissue inflammation by eliciting macrophage polar-
lution of inflammation. Front Immunol 3:4 ization toward an M2-like phenotype. J Immunol
56. Stables MJ, Shah S, Camon EB, Lovering RC, 187(10):5408–5418
Newson J, Bystrom J et al (2011) Transcriptomic 69. Cash JL, Norling LV, Perretti M (2014) Resolution
analyses of murine resolution-phase macrophages. of inflammation: targeting GPCRs that inter-
Blood 118(26):e192–e208 act with lipids and peptides. Drug Discov Today
57. Koh TJ, DiPietro LA (2011) Inflammation and 19(8):1186–1192
wound healing: the role of the macrophage. Expert 70. Serhan CN, Hong S, Gronert K, Colgan SP,
Rev Mol Med 13:e23 Devchand PR, Mirick G et al (2002) Resolvins: a
58. Chiurchiu V, Leuti A, Dalli J, Jacobsson A, Battistini family of bioactive products of omega-3 fatty acid
L, Maccarrone M et al (2016) Proresolving lipid transformation circuits initiated by aspirin treatment
mediators resolvin D1, resolvin D2, and maresin that counter proinflammation signals. J Exp Med
1 are critical in modulating T cell responses. Sci 196(8):1025–1037
Transl Med 8(353):353ra111
6 SPMs and Cardiac Repair 61
71. Serhan CN, Hamberg M, Samuelsson B (1984) Postischemic revascularization while resolving
Lipoxins: novel series of biologically active com- inflammation. Circulation 134(9):666–680
pounds formed from arachidonic acid in human leu- 85. Serhan CN, Petasis NA (2011) Resolvins and pro-
kocytes. Proc Natl Acad Sci U S A 81(17):5335–5339 tectins in inflammation resolution. Chem Rev
72. Newson J, Stables M, Karra E, Arce-Vargas F, 111(10):5922–5943
Quezada S, Motwani M et al (2014) Resolution of 86. Varga Z, Sabzwari SRA, Vargova V (2017)
acute inflammation bridges the gap between innate Cardiovascular risk of nonsteroidal anti-
and adaptive immunity. Blood 124(11):1748–1764 inflammatory drugs: an under-recognized public
73. Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang health issue. Cureus 9(4):e1144
N, Gronert K (2000) Novel functional sets of lipid- 87. Cheng Y, Austin SC, Rocca B, Koller BH, Coffman
derived mediators with antiinflammatory actions TM, Grosser T et al (2002) Role of prostacyclin in
generated from omega-3 fatty acids via cyclooxygen- the cardiovascular response to thromboxane A2.
ase 2-nonsteroidal antiinflammatory drugs and trans- Science (New York, NY) 296(5567):539–541
cellular processing. J Exp Med 192(8):1197–1204 88. Fredman G, Ozcan L, Spolitu S, Hellmann J,
74. Sansbury BE, Spite M (2016) Resolution of acute Spite M, Backs J et al (2014) Resolvin D1 lim-
inflammation and the role of Resolvins in immu- its 5- lipoxygenase nuclear localization and
nity, thrombosis, and vascular biology. Circ Res leukotriene B4 synthesis by inhibiting a calcium-
119(1):113–130 activated kinase pathway. Proc Natl Acad Sci USA
75. Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, 111(40):14530–14535
Serhan CN et al (2015) Resolvin D1 activates the 89. Blomer N, Pachel C, Hofmann U, Nordbeck P,
inflammation resolving response at splenic and ven- Bauer W, Mathes D et al (2013) 5-lipoxygenase
tricular site following myocardial infarction leading facilitates healing after myocardial infarction. Basic
to improved ventricular function. J Mol Cell Cardiol Res Cardiol 108(4):367
84:24–35 90. Clària J, Dalli J, Yacoubian S, Gao F, Serhan
76. Serhan CN, Chiang N, Van Dyke TE (2008) CN (2012) Resolvin D1 and resolvin D2 govern
Resolving inflammation: dual anti-inflammatory and local inflammatory tone in obese fat. J Immunol
pro-resolution lipid mediators. Nat Rev Immunol 189(5):2597–2605
8(5):349–361 91. Arnardottir HH, Dalli J, Colas RA, Shinohara M,
77. Colas RA, Souza PR, Walker ME, Burton M, Serhan CN (2014) Aging delays resolution of acute
Zaslona Z, Curtis AM et al (2018) Impaired produc- inflammation in mice: reprogramming the host
tion and diurnal regulation of vascular RvDn-3 DPA response with novel nano-proresolving medicines.
increase systemic inflammation and cardiovascular J Immunol 193(8):4235–4244
disease. Circ Res 122(6):855–863 92. Arita M, Bianchini F, Aliberti J, Sher A, Chiang N,
78. Serhan CN, Yacoubian S, Yang R (2008) Anti- Hong S et al (2005) Stereochemical assignment,
inflammatory and proresolving lipid mediators. antiinflammatory properties, and receptor for the
Annu Rev Pathol 3:279–312 omega-3 lipid mediator resolvin E1. J Exp Med
79. Hong S, Lu Y, Yang R, Gotlinger KH, Petasis NA, 201(5):713–722
Serhan CN (2007) Resolvin D1, protectin D1, 93. Arita M, Bianchini F, Aliberti J, Sher A, Chiang N,
and related docosahexaenoic acid-derived prod- Hong S et al (2005) Stereochemical assignment,
ucts: analysis via electrospray/low energy tandem antiinflammatory properties, and receptor for the
mass spectrometry based on spectra and fragmen- omega-3 lipid mediator resolvin E1. J Exp Med
tation mechanisms. J Am Soc Mass Spectrom 201(5):713–722
18(1):128–144 94. Schwab JM, Chiang N, Arita M, Serhan CN (2007)
80. Jadapalli JK, Halade GV (2018) Unified nexus of Resolvin E1 and protectin D1 activate inflammation-
macrophages and maresins in cardiac reparative resolution programmes. Nature 447(7146):869–874
mechanisms. FASEB J 32(10):5227–5237 95. Oh SF, Dona M, Fredman G, Krishnamoorthy S,
81. Gilbert K, Bernier J, Bourque-Riel V, Malick M, Irimia D, Serhan CN (2012) Resolvin E2 formation
Rousseau G (2015) Resolvin D1 reduces infarct size and impact in inflammation resolution. J Immunol
through a phosphoinositide 3-kinase/protein kinase 188(9):4527–4534
B mechanism. J Cardiovasc Pharmacol 66(1):72–79 96. Levy BD (2010) Resolvins and protectins:
82. Gilbert K, Malick M, Madingou N, Bourque-Riel natural pharmacophores for resolution biol-
V, Touchette C, Rousseau G (2016) Linoleic acid ogy. Prostaglandins Leukot Essent Fatty Acids
attenuates cardioprotection induced by resolvin D1. 82(4–6):327–332
J Nutr Biochem 31:122–126 97. Chiang N, de la Rosa X, Libreros S, Serhan CN
83. Halade GV, Kain V, Serhan CN (2018) Immune (2017) Novel Resolvin D2 receptor axis in infectious
responsive resolvin D1 programs myocardial inflammation. J Immunol 198(2):842–851
infarction-induced cardiorenal syndrome in heart 98. Serhan CN (2007) Resolution phase of inflamma-
failure. FASEB J 32(7):3717–3729 tion: novel endogenous anti-inflammatory and pro-
84. Zhang MJ, Sansbury BE, Hellmann J, Baker JF, Guo resolving lipid mediators and pathways. Annu Rev
L, Parmer CM et al (2016) Resolvin D2 enhances Immunol 25:101–137
62 G. V. Halade and B. Tourki
99. Samuelsson B, Hammarstrom S, Hamberg M, Serhan 113. Jove M, Mate I, Naudi A, Mota-Martorell N, Portero-
CN (1985) Structural determination of leukotrienes Otin M, De la Fuente M et al (2016) Human aging is
and lipoxins. Adv Prostaglandin Thromboxane a metabolome-related matter of gender. J Gerontol A
Leukot Res 14:45–71 Biol Sci Med Sci 71(5):578–585
100. Gronert K, Maheshwari N, Khan N, Hassan IR, 114. Dalli J, Serhan CN (2017) Pro-resolving mediators
Dunn M, Laniado Schwartzman M (2005) A role for in regulating and conferring macrophage function.
the mouse 12/15-lipoxygenase pathway in promot- Front Immunol 8:1400
ing epithelial wound healing and host defense. J Biol 115. Ho KJ, Spite M, Owens CD, Lancero H, Kroemer
Chem 280(15):15267–15278 AHK, Pande R et al (2010) Aspirin-triggered lipoxin
101. Stables MJ, Gilroy DW (2011) Old and new genera- and resolvin E1 modulate vascular smooth muscle
tion lipid mediators in acute inflammation and reso- phenotype and correlate with peripheral atheroscle-
lution. Prog Lipid Res 50(1):35–51 rosis. Am J Pathol 177(4):2116–2123
102. Kain V, Liu F, Kozlovskaya V, Ingle KA, Bolisetty 116. Serhan CN, Takano T, Clish CB, Gronert K, Petasis
S, Agarwal A et al (2017) Resolution agonist 15-epi- N (1999) Aspirin-triggered 15-epi-lipoxin A4 and
Lipoxin A4 programs early activation of resolving novel lipoxin B4 stable analogs inhibit neutrophil-
phase in post-myocardial infarction healing. Sci Rep mediated changes in vascular permeability. Adv Exp
7(1):9999 Med Biol 469:287–293
103. Kain V, Liu F, Kozlovskaya V, Ingle KA, Bolisetty 117. Lee CH (2012) Resolvins as new fascinating drug
S, Agarwal A et al (2017) Resolution agonist 15-epi- candidates for inflammatory diseases. Arch Pharm
Lipoxin A4 programs early activation of resolving Res 35(1):3–7
phase in post-myocardial infarction healing. Sci Rep 118. Halade GV, Kain V, Wright GM, Jadeapalli JK
7(1):9999 (2018) Subacute treatment of carprofen facilitate
104. Maderna P, Cottell DC, Toivonen T, Dufton N, splenocardiac resolution deficit in cardiac injury.
Dalli J, Perretti M et al (2010) FPR2/ALX receptor J Leukoc Biol 104(6):1173–1186
expression and internalization are critical for lipoxin 119. Jadapalli JK, Wright GW, Kain V, Sherwani MA,
A4 and annexin-derived peptide-stimulated phagocy- Sonkar R, Yusuf N et al (2018) Doxorubicin triggers
tosis. FASEB J 24(11):4240–4249 splenic contraction and irreversible dysregulation
105. Akagi D, Chen M, Toy R, Chatterjee A, Conte MS of COX and LOX that alters the inflammation-
(2015) Systemic delivery of proresolving lipid medi- resolution program in the myocardium. Am J Physiol
ators resolvin D2 and maresin 1 attenuates intimal Heart Circ Physiol 315(5):H1091–Hh100
hyperplasia in mice. FASEB J 29(6):2504–2513 120. Halade GV, Kain V (2017) Obesity and
106. Serhan CN, Yang R, Martinod K, Kasuga K, Pillai Cardiometabolic defects in heart failure pathology.
PS, Porter TF et al (2009) Maresins: novel macro- Compr Physiol 7(4):1463–1477
phage mediators with potent antiinflammatory and 121. Tourki B, Halade GV (2018) The failing of the obe-
proresolving actions. J Exp Med 206(1):15–23 sity paradox in the failing heart. Am J Physiol Heart
107. Liu G, Liu Q, Shen Y, Kong D, Gong Y, Tao B et al Circ Physiol 315:H1353–H1355
(2018) Early treatment with Resolvin E1 facilitates 122. Spite M, Clària J, Serhan CN (2014) Resolvins,
myocardial recovery from ischaemia in mice. Br specialized proresolving lipid mediators, and their
J Pharmacol 175(8):1205–1216 potential roles in metabolic diseases. Cell Metab
108. Colas RA, Dalli J, Chiang N, Vlasakov I, Sanger JM, 19(1):21–36
Riley IR et al (2016) Identification and Actions of the 123. Krishnamoorthy S, Recchiuti A, Chiang N,
Maresin 1 Metabolome in Infectious Inflammation. Yacoubian S, Lee C-H, Yang R et al (2010) Resolvin
J Immunol 197(11):4444–4452 D1 binds human phagocytes with evidence for
109. Viola JR, Lemnitzer P, Jansen Y, Csaba G, proresolving receptors. Proc Natl Acad Sci U S A
Winter C, Neideck C et al (2016) Resolving lipid 107(4):1660–1665
mediators Maresin 1 and Resolvin D2 prevent 124. Lopez EF, Kabarowski JH, Ingle KA, Kain V, Barnes
Atheroprogression in mice. Circ Res 119(9): S, Crossman DK et al (2015) Obesity superimposed
1030–1038 on aging magnifies inflammation and delays the
110. Tang S, Wan M, Huang W, Stanton RC, Xu Y (2018) resolving response after myocardial infarction. Am
Maresins: specialized Proresolving lipid mediators J Physiol Heart Circ Physiol 308(4):H269–H280
and their potential role in inflammatory-related dis- 125. Martín-Timón I, Sevillano-Collantes C, Segura-
eases. Mediat Inflamm 2018:2380319 Galindo A, Del Cañizo-Gómez FJ (2014) Type
111. Claria J, Lopez-Vicario C, Rius B, Titos E (2017) 2 diabetes and cardiovascular disease: have all
Pro-resolving actions of SPM in adipose tissue biol- risk factors the same strength? World J Diabetes
ogy. Mol Asp Med 58:83–92 5(4):444–470
112. Lee HN, Surh YJ (2013) Resolvin D1-mediated 126. Freire MO, Dalli J, Serhan CN, Van Dyke TE
NOX2 inactivation rescues macrophages under- (2017) Neutrophil Resolvin E1 receptor expres-
taking efferocytosis from oxidative stress-induced sion and function in type 2 diabetes. J Immunol
apoptosis. Biochem Pharmacol 86(6):759–769 198(2):718–728
6 SPMs and Cardiac Repair 63
127. Ferrante AW Jr (2007) Obesity-induced inflamma- 142. Ingle KA, Kain V, Goel M, Prabhu SD, Young ME,
tion: a metabolic dialogue in the language of inflam- Halade GV (2015) Cardiomyocyte-specific Bmal1
mation. J Intern Med 262(4):408–414 deletion in mice triggers diastolic dysfunction, extra-
128. Ouchi N, Parker JL, Lugus JJ, Walsh K (2011) cellular matrix response, and impaired resolution
Adipokines in inflammation and metabolic disease. of inflammation. Am J Physiol Heart Circ Physiol
Nat Rev Immunol 11(2):85–97 309(11):H1827–H1836
129. Hotamisligil GS (2006) Inflammation and metabolic 143. Young ME, Brewer RA, Peliciari-Garcia RA, Collins
disorders. Nature 444(7121):860–867 HE, He L, Birky TL et al (2014) Cardiomyocyte-
130. Hellmann J, Tang Y, Kosuri M, Bhatnagar A, Spite M specific BMAL1 plays critical roles in metabolism,
(2011) Resolvin D1 decreases adipose tissue macro- signaling, and maintenance of contractile function of
phage accumulation and improves insulin sensitivity the heart. J Biol Rhythm 29(4):257–276
in obese-diabetic mice. FASEB J 25(7):2399–2407 144. Colas RA, Souza PR, Walker ME, Burton M,
131. Titos E, Rius B, González-Périz A, López-Vicario Zasłona Z, Curtis AM et al (2018) Impaired produc-
C, Morán-Salvador E, Martínez-Clemente M tion and diurnal regulation of vascular RvDn-3 DPA
et al (2011) Resolvin D1 and its precursor doco- increase systemic inflammation and cardiovascular
sahexaenoic acid promote resolution of adipose disease. Circ Res 122(6):855–863
tissue inflammation by eliciting macrophage polar- 145. Alvarez P, Hannawi B, Guha A (2016) Exercise
ization toward an M2-like phenotype. J Immunol and heart failure: advancing knowledge and
187(10):5408–5418 improving care. Methodist Debakey Cardiovasc
132. Brennan EP, Mohan M, McClelland A, de Gaetano J 12(2):110–115
M, Tikellis C, Marai M et al (2018) Lipoxins protect 146. Markworth JF, Vella L, Lingard BS, Tull DL,
against inflammation in diabetes-associated athero- Rupasinghe TW, Sinclair AJ et al (2013) Human
sclerosis. Diabetes 67(12):2657–2667 inflammatory and resolving lipid mediator
133. Tang Y, Zhang MJ, Hellmann J, Kosuri M,
responses to resistance exercise and ibuprofen treat-
Bhatnagar A, Spite M (2013) Proresolution therapy ment. Am J Physiol Regul Integr Comp Physiol
for the treatment of delayed healing of diabetic 305(11):R1281–R1296
wounds. Diabetes 62(2):618–627 147. Gangemi S, Pescara L, D'Urbano E, Basile G,
134. Kannan A, Janardhanan R (2014) Hypertension as Nicita-Mauro V, Davì G et al (2005) Aging is charac-
a risk factor for heart failure. Curr Hypertens Rep terized by a profound reduction in anti-inflammatory
16(7):447 lipoxin A4 levels. Exp Gerontol 40(7):612–614
135. Pearson T, Zhang J, Arya P, Warren AY, Ortori C, 148. Taylor RS, Sagar VA, Davies EJ, Briscoe S, Coats
Fakis A et al (2010) Measurement of vasoactive AJ, Dalal H et al (2014) Exercise-based rehabilita-
metabolites (hydroxyeicosatetraenoic and epoxye- tion for heart failure. Cochrane Database Syst Rev
icosatrienoic acids) in uterine tissues of normal (4):Cd003331
and compromised human pregnancy. J Hypertens 149. Hoetker D, Chung W, Zhang D, Zhao J, Schmidtke
28(12):2429–2437 VK, Riggs DW et al (2018) Exercise alters and
136. Didion SP (2017) Unraveling the role and complexi- beta-alanine combined with exercise augments
ties of inflammation in hypertension, Hypertension histidyl dipeptide levels and scavenges lipid per-
(Dallas, Tex: 1979). 70(4):700–702 oxidation products in human skeletal muscle.
137. Pullamsetti SS, Savai R, Janssen W, Dahal BK, J Appl Physiol (1985). https://doi.org/10.1152/
Seeger W, Grimminger F et al (2011) Inflammation, japplphysiol.00007.2018
immunological reaction and role of infection in 150. Meschiari CA, Ero OK, Pan H, Finkel T, Lindsey
pulmonary hypertension. Clin Microbiol Infect ML (2017) The impact of aging on cardiac extracel-
17(1):7–14 lular matrix. GeroScience 39(1):7–18
138. Hiram R, Rizcallah E, Sirois C, Sirois M, Morin C, 151. Franceschi C, Campisi J (2014) Chronic inflamma-
Fortin S et al (2014) Resolvin D1 reverses reactiv- tion (inflammaging) and its potential contribution to
ity and Ca2+ sensitivity induced by ET-1, TNF- age-associated diseases. J Gerontol A Biol Sci Med
alpha, and IL-6 in the human pulmonary artery. Am Sci 69(Suppl 1):S4–S9
J Physiol Heart Circ Physiol 307(11):H1547–H1558 152. Yabluchanskiy A, Ma Y, DeLeon-Pennell KY, Altara
139. McAlpine CS, Swirski FK (2016) Circadian influ- R, Halade GV, Voorhees AP et al (2016) Myocardial
ence on metabolism and inflammation in atheroscle- infarction superimposed on aging: MMP-9 deletion
rosis. Circ Res 119(1):131–141 promotes M2 macrophage polarization. J Gerontol A
140. Perretti M, D'Acquisto F (2009) Annexin A1 and Biol Sci Med Sci 71(4):475–483
glucocorticoids as effectors of the resolution of 153. Yabluchanskiy A, Ma Y, Chiao YA, Lopez EF,
inflammation. Nat Rev Immunol 9(1):62–70 Voorhees AP, Toba H et al (2014) Cardiac aging is
141. Krump E, Picard S, Mancini J, Borgeat P (1997) initiated by matrix metalloproteinase-9-mediated
Suppression of leukotriene B4 biosynthesis by endothelial dysfunction. Am J Physiol Heart Circ
endogenous adenosine in ligand-activated human Physiol 306(10):H1398–H1407
neutrophils. J Exp Med 186(8):1401–1406
64 G. V. Halade and B. Tourki
154. Melton DW, Roberts AC, Wang H, Sarwar Z, Wetzel 157. Buford TW. (Dis)Trust your gut: the gut microbiome
MD, Wells JT et al (2016) Absence of CCR2 results in age-related inflammation, health, and disease.
in an inflammaging environment in young mice with Microbiome. 2017;5(\):80
age-independent impairments in muscle regenera- 158. Zabell A, Tang WHW (2017) Targeting the microbi-
tion. J Leukoc Biol 100(5):1011–1025 ome in heart failure. Curr Treat Options Cardiovasc
155. Halade GV, Kain V, Black LM, Prabhu SD, Ingle Med 19(4):27
KA (2016) Aging dysregulates D- and E-series 159. Luedde M, Winkler T, Heinsen F-A, Rühlemann
resolvins to modulate cardiosplenic and cardiore- MC, Spehlmann ME, Bajrovic A et al (2017) Heart
nal network following myocardial infarction. Aging failure is associated with depletion of core intestinal
8(11):2611–2634 microbiota. ESC Heart Fail 4(3):282–290
156. Jové M, Maté I, Naudí A, Mota-Martorell N, Portero- 160. Menni C, Zierer J, Pallister T, Jackson MA, Long T,
Otín M, De la Fuente M et al (2016) Human aging is Mohney RP et al (2017) Omega-3 fatty acids cor-
a metabolome-related matter of gender. J Gerontol A relate with gut microbiome diversity and production
Biol Sci Med Sci 71(5):578–585 of N-carbamylglutamate in middle aged and elderly
women. Sci Rep 7(1):11079
Novel n-3 Docosapentaneoic Acid-
Derived Pro-resolving Mediators 7
Are Vasculoprotective and Mediate
the Actions of Statins
in Controlling Inflammation
Jesmond Dalli, Kimberly Pistorius,
and Mary E. Walker
upregulated during acute inflammation, to an cells and chemotaxis towards IL-8, to a similar
extent that was comparable to essential fatty extent as the DHA-derived RvD2 (7S,16R,17S-
acids that are involved in the biosynthesis of lipid trihydroxy- 4Z,8E,10Z,12E,14E,19Z- docosa-
mediators, including arachidonic acid and hexaenoic acid) [20, 23, 24]. RvD2n-3 DPA and
DHA. Using a systematic approach coupling RvD5n-3 DPA also regulate the expression of adhe-
structure elucidation with functional readouts we sion molecules on peripheral blood leukocytes
found that endogenous n-3 DPA is converted to and platelets including the expression of CD11b
bioactive mediators in both mice and human leu- on neutrophils and monocytes as well as CD62P
kocytes that carried pro-resolving properties and CD63 on platelets [25]. Furthermore, they
[20]. These mediators are congenerous with also regulate the formation of leukocyte-platelet
DHA products, namely D-series resolvins heterotypic aggregates in both human and mouse
(RvDn-3 DPA), protectins (PDn-3 DPA) and maresins peripheral blood. The biological actions of these
(MaRn-3 DPA), with unique stereochemistries [20]. molecules extend beyond the regulation of mech-
In self resolving exudates, the production of these anisms in leukocyte trafficking. Indeed, n-3
molecules was temporally regulated where for DPA-derived SPMs also regulate the uptake of
example RvD1n-3 DPA (7S,8R,17S-trihydroxy- apoptotic cells by macrophages, a key biological
9E,11E,13Z,15E,19Z -docosapentaenoic acid) action in the resolution of inflammation, with
and PD2n-3 DPA (16,17-dihydroxy-7Z,10,13, increases in macrophage efferocytosis of up to
14,19Z- docosapentaenoic acid) displayed a 70% at doses as low as 1 nM [20, 23]. These
bi-phasic profile, reaching a maximum during mediators also display endothelial directed
peak neutrophil infiltration and late into actions, counteracting the TNF-α- mediated
resolution. PD1n-3 DPA (10R,17S-dihydroxy-upregulation of adhesion molecules, such as
7Z,11E,13E,15Z,19Z-docosapentaenoic acid), Intercellular Adhesion Molecule 1 (ICAM-1/
MaR2n-3 DPA (13,14-dihydroxy-7Z,9,11, 16Z, CD54), on endothelial cells [20].
19Z- docosapentaenoic acid) and MaR3n-3 DPA
(4, 21-dihydroxy-7Z,10Z,12E,16Z,19Z-docosa
pentaenoic acid) levels were each found to 7.3 Diurnal Regulation of RvDn-3
reach a maximum at the 4 h interval and DPA Controls Vascular
gradually decreased over the next 20 h. The peak Leukocyte and Platelet
in exudate RvD2n-3 DPA (7S,16,17S-trihydroxy- Activation
8,10Z,12,14,19Z-docosapentaenoic acid) levels
coincided with the onset of resolution (the point Circadian mechanisms are at the heart of a num-
where PMN levels reach ~50% of maximim neu- ber of physiological functions, including leuko-
trophil counts). RvD5n-3 DPA (7S,17S-dihydroxy- cyte and platelet responses [26, 27]. Disturbances
4Z,8,10Z,13Z,15,19Z-docosahexaenoic acid) to various aspects of these fundamental mecha-
levels were found to gradually increase over the nisms are thought to be responsible for many of
course of inflammation-resolution, with a maxi- the diseases that afflict modern societies, includ-
mum being reached late in the resolution phase. ing cardiovascular and metabolic disorders [26–
The n-3 DPA product corresponding to MaR1n-3 28]. These conditions are characterized by a
DPA (7S,14S-dihydroxy-8E, 10E, 12Z, 16Z, dysregulated inflammatory response, although
19Z-docosapentaenoic acid) gave levels that the exact mechanisms that underlie this inflam-
were elevated in the peritoneum of naive mice, matory state remain of interest. Recent studies
where upon challenge with zymosan these levels demonstrate that the production of RvDn-3 DPA are
drastically decreased [20]. Of note, each of these diurnally regulated in the peripheral blood of
molecules displayed leukocyte directed actions healthy volunteers [25] (Fig. 7.1). Multivariate
whereby incubation of human neutrophils with analysis of plasma lipid mediator profiles demon-
RvD5n-3 DPA or PD1n-3 DPA markedly reduced neu- strated a diurnal shift in plasma LM-SPM con-
trophil adhesion to TNF-α activated endothelial centrations. This shift was associated with an
68 J. Dalli et al.
Fig. 7.1 Diurnal changes in RvDn-3 DPA regulates monocytes, neutrophils and platelets during the early
peripheral blood leukocyte and platelet activation pro- morning hours. Increases in circulating adenosine concen-
tecting from cardiovascular disease. In peripheral blood trations in patients with cardiovascular disease inhibit
diurnal changes in acetylcholine (ACh) upregulates ALOX5 activity disrupting the diurnal changes in plasma
15-lipoxygenase (ALOX15) activity promoting RvDn-3 DPA RvDn-3 DPA and increasing peripheral blood leukocyte and
biosynthesis that limit the physiological activation of platelet activation
increase in the amounts of n-3 DPA derived tion of whole blood with ACh increased RvDn-3
mediators, including RvD1n-3 DPA and RvD5n-3 DPA DPA concentrations, including RvD2n-3 DPA, under
from the evening (18:00 h) to morning intervals both static and flow conditions.
(7:00 and 9:00 h). These diurnal changes in Assessment of the production of these media-
peripheral blood RvDn-3 DPA concentrations were tors in patients with cardiovascular disease
abrogated in mice lacking the main orchestrator (CVD) demonstrated significant decreases in
of the molecular clock, Aryl hydrocarbon recep- plasma RvDn-3 DPA concentrations and a marked
tor nuclear translocator-like protein 1, in myeloid impairment in their diurnal regulation when
cells. Of note, the fluctuations in plasma RvDn-3 compared with healthy volunteers. Flow cyto-
DPA were associated with a regulation of leukocyte metric analysis of peripheral blood leukocyte
and platelet activation that reaches a maximum from patients with CVD demonstrated increases
between 7:00 and 9:00 h coincident with an in the expression of CD11b on both neutrophils
increase in RvDn-3 DPA concentrations. The pro- and monocytes when compared with healthy vol-
duction of these mediators was found to be under unteers. This was coupled with increases in
the control of acetylcholine (ACh), with periph- platelet–neutrophil and platelet–monocyte aggre-
eral blood concentrations of this neurotransmitter gates in peripheral blood from patients with CVD
also reaching a maximum during the early hours [25]. In addition, we found a significant relation-
of the morning (i.e. 7:00 h). Furthermore, incuba- ship between peripheral blood RvDn-3 DPA
7 Novel n-3 Docosapentaneoic Acid-Derived Pro-resolving Mediators Are Vasculoprotective… 69
c oncentrations and leukocyte and platelet activa- 7.4 PDn-3 DPA Regulates
tion, as demonstrated by a negative correlation Macrophage Phenotype
between RvDn-3 DPA and neutrophil CD41, mono- and Function
cyte CD41, and platelet CD63 and CD42b During Monocyte
expression. Investigations into mechanisms that to Macrophage
lead to the downregulation of peripheral blood Differentiation
RvDn-3 DPA in patients with CVD demonstrated a
link between peripheral blood adenosine concen- It is now well appreciated that in chronic inflam-
trations and the activity of one of the RvDn-3 DPA matory conditions, such as atherosclerosis and
biosynthetic enzymes, ALOX5. Adenosine, rheumatoid arthritis, monocytes play a central
which via the activation of the A2a receptor, role in the initiation, propagation and termination
downregulates the activity of ALOX5 [29], was of inflammation [32, 33]. Upon recruitment to
increased in peripheral blood from patients with the site of inflammation, these cells either differ-
CVD. The role of adenosine in downregulating entiate to inflammatory macrophages that propa-
RvDn-3 DPA concentrations was further under- gate the inflammatory response or to resolution
scored by experiments where peripheral blood phase macrophages that promote the termination
from CVD patients was incubated with adenos- of inflammation and restitution of tissue func-
ine deaminase, leding to the restoration of tion. Studies investigating mechanism that regu-
peripheral blood RvDn-3 DPA concentrations [25]. late the differentiation of macrophages
These findings led us to propose RvDn-3 DPA as demonstrate a role for the PDn-3 DPA pathway in
endogenous protective signals that control phys- regulating the phenotype and function of
iological platelet and leukocyte activation. This monocyte-derived macrophages [34] (Fig. 7.2).
is further supported by observations made in Using a total organic synthetic approach coupled
Apolipoprotein E deficient mice (ApoE−/−) mice with lipid mediator profiling, human ALOX15
fed a western diet. Treatment of these mice with and ALOX15B were identified as the enzymes
RvD5n-3 DPA reduced platelet-leukocytes aggre- that catalyse the first two steps in the PDn-3 DPA
gates in vivo and modulated vascular lipid medi- biosynthetic pathway yielding an allylic epoxide.
ator profiles reducing concentrations of the Using total organic synthesis we established the
pro-thrombogenic mediator Thromboxane (Tx) absolute stereochemistry of this epoxide as
A2 (measured as its metabolite TxB2) and upreg- 1 6 S , 1 7 S - e p o x y - 7 Z , 1 0 Z , 1 2 E , 1 4 E , 1 9 Z -
ulating the formation of pro-resolving mediators docosapentaenoic acid [34, 35]. This intermedi-
including MaR1 (7S,14S-dihydroxy-4Z, 8E, ate was in turn converted to PD1n-3 DPA and PD2n-3
10E, 12Z, 16Z, 19Z-docosahexaenoic acid) and DPA by distinct epoxide hydrolase enzymes, where
aspirin triggered (AT)-LXA4 (5S,6R,15R- in human cells epoxide hydrolase 2 (EPHX2)
trihydroxy-7 E,9E,11Z,13E-eicosatetraenoic was found to catalyse the conversion of 16S,17S-
acid). Furthermore RvD5n-3 DPA also decreased ePDn-3 DPA to PD2n-3 DPA. Experiments investigat-
early aortic lesions in ApoE−/− mice. The present ing the expression of the PDn-3 DPA biosynthetic
findings are in line with published findings dem- pathway during monocyte to macrophage differ-
onstrating an altered production of vascular entiation found the expression of all three
DHA-derived SPMs including RvD2 and MaR1 enzymes was upregulated during monocyte to
and impaired resolution responses in the patho- macrophage differentiation. Of note, ALOX15
genesis of atherosclerosis [30, 31]. Together expression was higher in M2 differentiated cells,
these findings demonstrate that alterations in the whereas the expression of ALOX15B was higher
diurnal regulation of vascular RvDn-3 DPA may in M1 cells [34].
occur early in the pathogenesis of cardiovascu- Pharmacological inhibition and genetic dele-
lar diseases that results in vascular inflammation tion of ALOX15 enzymes in monocytes led to
and impaired biosynthesis of DHA derived phenotypic and functional changes in monocyte-
SPM. derived macrophages. In cells where ALOX15
70 J. Dalli et al.
was inhibited there was a downregulation of sev- ALOX15 deficient mice also restored the pheno-
eral lineage markers including CD206, CD163 type and efferocytic activity of macrophages in
and CD64 and a shift in macrophage phenotype vivo [34]. Thus, these findings identify the PDn-3
[34]. This downregulation in phagocytic recep- DPA pathway as a component in the monocyte-to-
tors was of functional consequence since inhibi- macrophage differentiation program that regu-
tion of the PDn-3 DPA biosynthetic pathway also lates their phenotype and function.
significantly downregulated the ability of human
macrophages to uptake apoptotic cells, a key pro-
resolving action [36, 37]. Of note, this alteration 7.5 vTs Are Produced
R
in macrophage phenotype and function was During the Early Stages
recovered with the reconstitution of components of Acute Inflammation
within the PDn-3 DPA pathway. Incubation of and Temper Host Immune
human monocyte-derived macrophages with Responses
either PD1n-3 DPA or 16S, 17S-epoxy-PDn-3 DPA led
to a restoration of several phagocytic receptors Recent studies have described and characterized
and increased macrophage efferocytosis. a new family of bioactive mediators produced
Furthermore, administration of PD1n-3 DPA to from n-3 DPA. This new family of four resolvins
7 Novel n-3 Docosapentaneoic Acid-Derived Pro-resolving Mediators Are Vasculoprotective… 71
is termed the 13-series resolvins (RvT) given that when compared to healthy volunteers. RvTs are
all four molecules display potent host protective protective in mice during acute inflammation
actions and carry a hydroxyl group on carbon 13 where a mixture of RvT1, RvT2, RvT3 and
[38]. RvTs are biosynthesized from n-3 DPA in a RvT4 immediately before or 2 h after intraperi-
process that requires both endothelial cell COX-2 toneal Escherichia coli inoculation resulted in
and neutrophil lipoxygenase (ALOX) activity. host- protection, increasing survival by >60%.
The role for ALOX enzymes in the biosynthesis Indeed, RvT limited neutrophil recruitment to
of RvT was established using heavy oxygen the site of inflammation, increased phagocytosis
incorporation [38] with the identity of the ALOX and intracellular ROS levels and upregulated
enzymes catalyzing this reaction remaining of macrophage efferocytosis. Of note, the protec-
interest. tive actions of RvTs resulted from the repro-
During acute inflammation, cytokines such as gramming of the innate host response since RvT
TNF-α and IL-1β are released, activating did not display direct bactericidal actions at bio-
endothelial cells that upregulate the expression logically-relevant concentrations. Additionally,
of COX-2. Endothelial COX-2 converts n-3 DPA RvT reduced monocyte and macrophage expres-
to 13R-hydro(peroxy)-docosa-7Z,10Z,14E,16Z, sion of inflammasome components, decreasing
19Z-pentaenoic acid (13R-HpDPA); 13R-HpDPA caspase-1 and IL-1β expression and lactate
or its reduced alcohol form 13R-hydroxy- dehydrogenase activity, a marker of pyroptosis.
docosa-7Z,10Z,14E,16Z,19Z-pentaenoic acid RvT also reduced peripheral blood platelet-leu-
(13R-HDPA) is then donated to neutrophils dur- kocyte aggregates, an observation associated
ing neutrophil-endothelial cell interactions, with reduced systemic inflammation. Exudate
whereby it is first converted to 7-hydro(peroxy)- macrophage efferocytosis was also increased
13R-hydroxy-docosa-8,10Z,14E,16Z,19Z- and a significant reduction in local and systemic
pentaenoic acid [38]. This molecule can eicosanoid levels was found in mice given RvT
then undergo a second lipoxygenation [38].
reaction to yield 7,13R,20-trihydroxy-docosa- Using a total organic synthetic approach, we
8,10,14E,16Z,18-pentaenoic acid that was coined recently established the complete stereochemis-
as RvT1. The hydroperoxide can also, in a try and biosynthetic role of 13R-HDPA in the
lipoxygenase- dependent manner, undergo an RvT pathway [39]. Chirally pure precursors were
epoxidation reaction to yield the allylic used in conjunction with stereoselective reac-
epoxide 7,8-epoxy-13-hydroxy-docosa-9,11, tions that installed the configuration at the carbon
14E,16Z,19Z-pentaenoic acid. This is then enzy- 13 atom as R and formed geometrically pure
matically hydrolysed to 7,12,13-trihydroxy- double-bond moieties.
docosa-8,10,14E,16Z,19Z-pentaenoic acid and Synthetic 13R-HDPA was matched with bio-
7,8,13-trihydroxy-docosa-9,11,14E,16Z,19Z- genic 13-HDPA, obtained by incubating n-3 DPA
pentaenoic acid, coined RvT2 and RvT3, respec- with human recombinant COX-2. Structural eval-
tively. Finally, 7-hydro(peroxy)-13R-hydroxy- uation of synthetic 13R-HDPA and biogenic
docosa- 8,10Z,14E,16Z,19Z-pentaenoic acid is 13-HDPA was carried out using liquid chroma-
also reduced to 7,13R-dihydroxy-docosa-tography tandem mass spectrometry (LC-MS/
8,10Z,14E,16Z,19Z-pentaenoic acid that is MS) to attain MRM chromatograms to match
coined RvT4 (Fig. 7.3). retention times and MS-MS spectra to identify
RvTs are produced in humans during exer- matching daughter ions. Chiral LC-MS/MS was
cise, that is now seen as a self-resolving inflam- used to confirm the stereochemistry around car-
matory state, marked by an increase in bon 13, and UV-Vis spectrophotometry was used
neutrophil-endothelial interactions with upregu- to match the double bond conjugation system.
lation of plasma RvT [38]. These mediators are Incubation of the synthetic material with human
also produced during infection when RvT levels leukocytes demonstrated that this was rapidly
are upregulated in plasma from septic patients converted to all four RvTs [39]. These findings
72 J. Dalli et al.
Fig. 7.3 RvT biosynthesis and actions in mediating expression yielding 13-HpDHA, that can then be con-
the protective actions of statins in infections and verted to 13-HDPA and donated to neutrophils where via
inflammatory arthritis. Production of RvT is initiated ALOX activity this is converted to RvT1–4. Statin medi-
via the conversion of n-3 DPA by the endothelial COX-2 ated S-nitrosylation of COX-2 upregulates 13-HDPA pro-
duction contributing to the upregulation of RvTs
confirmed that the stereochemistry around the ple, upregulates the biosynthesis of 15-epi-LXA4
hydroxyl group on carbon 13 was in the R [41]. In addition, atorvastatin was recently found
orientation and the double bond geometry to increase RvT formation during human
around the conjugated double bond system neutrophil-endothelial cell interactions, as well
was found to be in E, Z with the complete as in mice during infections. This increase in RvT
stereochemistry established as 13(R)-hydroxy- production resulted from the S-nitrosylation of
7Z,10Z,13R,14E,16Z,19Z-docosapentaenoic COX-2 leading to increased 13R-HDPA levels,
acid as well as the role of this intermediate in the suggesting that the S-nitrosylation of COX-2
RvT biosynthetic pathway. increased catalytic activity of the enzyme. This
finding was in concordance with the
S-nitrosylation of COX-2 cysteine residues in the
7.6 vT Mediate the Biological
R presence of atorvastatin [38]. Inhibition of induc-
Actions of Statins ible nitric oxide synthase (iNOS) by L-NG-
in Infectious-Inflammation nitroarginine (L-NAME) reduced the
and Inflammatory Arthritis atorvastatin-mediated increases in plasma RvT
levels after E. coli inoculation. A similar reduc-
SPMs are implicated in mediating the protective tion was observed when mice were given cele-
actions of a number of clinically relevant thera- coxib, a COX-2 specific inhibitor. This highlights
peutics including aspirin and statins, whereby the complex regulatory axis of COX-2, as post-
aspirin initiates the biosynthesis of epimeric translational modification of the enzyme may
forms of SPMs [40], while lovastatin, for exam-
7 Novel n-3 Docosapentaneoic Acid-Derived Pro-resolving Mediators Are Vasculoprotective… 73
yield mediators with distinct biological activities ~34% respectively in mice given atorvastatin
to the classic eicosanoids. compared to mice given vehicle. In mice given
This mechanism was recently also found to be pravastatin, CD11b expression was decreased by
protective in arthritic inflammation where admin- ~40% and platelet-monocyte aggregation reduced
istration of atorvastatin upregulated RvT concen- by ~35%. Compared to vehicle, neutrophil acti-
trations during inflammatory arthritis in both vation markers were significantly reduced by
peripheral blood and joints [42]. Of note, this atorvastatin and pravastatin, reducing CD11b
protective mechanism was not unique to atorvas- expression by ~30% and platelet-neutrophil
tatin and was shared with pravastatin. Atorvastatin aggregation by ~24%. Of note, administration of
administration during ongoing arthritis led to a celecoxib that inhibits the upregulation of RvT
43% increase in total RvT amounts in arthritic by pravastatin and atorvastatin reverses the pro-
paws compared to vehicle-treated mice. tective actions of these statins on both disease
Pravastatin also increased paw RvT by ~20% severity and leukocyte responses [42].
with increases in RvT1 and RvT2.
These increases in joint RvT concentrations
were also linked with decreases in tissue pros- 7.7 Conclusion
tanoids and LTB4 concentrations. Prostaglandins
were reduced by 20–40% by all statins tested The identification of n-3 DPA as a substrate to
when compared to vehicle. Exceptionally, PGF2α novel, structurally distinct, mediators that display
was reduced by ~75% in mice given pravastatin. potent host protective activities demonstrates that
LTB4 concentrations were reduced ~50% by ator- complex mediator networks become activated
vastatin and ~15% by pravastatin. Additionally, during acute inflammation to ensure tissue
TxB2 was reduced 20–50% both statins [42]. The homeostasis. This is further underscored by the
upregulation in RvT concentrations were linked observation of a selective regulation of distinct
with a reduction in disease severity where in mice lipid mediator pathways in a tissue and cell spe-
administered atorvastatin, disease progression cific manner. In addition, mounting evidence
was dampened at day 4 post disease initiation, suggests that some of the beneficial actions of a
with disease scores reaching a maximum of number of widely used drugs, including statins,
9.1 ± 1.2 at day 5 which was sustained until day is mediated via the regulation of these protective
7. When mice were administered pravastatin, dis- pathways. Given the potent actions of n-3 DPA-
ease activity at day 5 was lower compared with derived SPMs in regulating systemic and periph-
mice administered vehicle, with a reduction in eral inflammatory responses, utilizing analogues
disease activity maintained until day 7 measured and mimetics may be useful therapeutics in the
both as reduction in of clinical score and edema. prevention and treatment of chronic inflamma-
In addition, both statins also lead to a reduction in tory diseases. In addition, strategies to boost their
joint damage at a histological level. endogenous production potentially via supple-
In inflammatory arthritis atorvastatin and mentation with n-3 DPA may also be useful in
pravastatin administration also regulates both cir- controlling inflammation. While the clinical evi-
culating and tissue resident leukocyte responses. dence for this approach is currently limited,
In non-classical monocytes, atorvastatin reduced recent studies in healthy volunteers provide evi-
the expression of CD11b by ~18% and platelet- dence for its effectiveness, whereby administra-
monocyte aggregation (measured by a decrease tion of n-3 DPA upregulates peripheral blood
in CD62P) was reduced by ~24% compared to concentrations of RvD5n-3 DPA [43]. Future studies
mice given vehicle [42]. Pravastatin significantly will need to identify the patient populations that
reduced platelet-monocyte aggregation by ~35%, will be responsive to this approach and the sup-
and decreased CD11b expression by ~10%. In plement forms that will be effective in regulating
classical monocytes, expression of CD11b and n-3 DPA-derived SPM concentrations.
CD62P were significantly reduced by ~42% and Furthermore, changes in the observation that
74 J. Dalli et al.
tissue concentrations of these SPM are altered in macrophage mediators with potent antiinflammatory
and proresolving actions. J Exp Med 206(1):15–23.
disease and that their levels can be upregulated https://doi.org/10.1084/jem.20081880
by certain therapeutics (e.g. atorvastatin and 12. Dona M, Fredman G, Schwab JM, Chiang N, Arita
pravastatin) suggest that these pathways may also M, Goodarzi A, Cheng G, von Andrian UH, Serhan
be useful as biomarkers in both patient stratifica- CN (2008) Resolvin E1, an EPA-derived mediator in
whole blood, selectively counterregulates leukocytes
tion and measuring the effectiveness of treatment and platelets. Blood 112(3):848–855. https://doi.
efficacy. org/10.1182/blood-2007-11-122598
13. Morita M, Kuba K, Ichikawa A, Nakayama M,
Katahira J, Iwamoto R, Watanebe T, Sakabe S, Daidoji
T, Nakamura S, Kadowaki A, Ohto T, Nakanishi
References H, Taguchi R, Nakaya T, Murakami M, Yoneda Y,
Arai H, Kawaoka Y, Penninger JM, Arita M, Imai
1. Malagoli D (2016) The evolution of the immune Y (2013) The lipid mediator protectin D1 inhibits
system: conservation and diversification. Elsevier, influenza virus replication and improves severe influ-
Amsterdam enza. Cell 153(1):112–125. https://doi.org/10.1016/j.
2. Majno G (1991) The ancient riddle of sigma eta psi cell.2013.02.027
iota sigma (sepsis). J Infect Dis 163(5):937–945 14. Dalli J, Vlasakov I, Riley IR, Rodriguez AR, Spur
3. Dalli J, Serhan CN (2018) Identification and struc- BW, Petasis NA, Chiang N, Serhan CN (2016)
ture elucidation of the pro-resolving mediators pro- Maresin conjugates in tissue regeneration biosyn-
vides novel leads for resolution pharmacology. Br thesis enzymes in human macrophages. Proc Natl
J Pharmacol 176:1024–1037. https://doi.org/10.1111/ Acad Sci U S A 113(43):12232–12237. https://doi.
bph.14336 org/10.1073/pnas.1607003113
4. de Gaetano M, McEvoy C, Andrews D, Cacace A, 15. Chiang N, Serhan CN (2017) Structural elucidation
Hunter J, Brennan E, Godson C (2018) Specialized and physiologic functions of specialized pro-resolving
pro-resolving lipid mediators: modulation of mediators and their receptors. Mol Asp Med 58:114–
diabetes-associated cardio-, reno-, and retino-vascular 129. https://doi.org/10.1016/j.mam.2017.03.005
complications. Front Pharmacol 9:1488. https://doi. 16. Colas RA, Dalli J, Chiang N, Vlasakov I, Sanger JM,
org/10.3389/fphar.2018.01488 Riley IR, Serhan CN (2016) Identification and actions
5. Perretti M, Norling LV (2017) Actions of SPM of the Maresin 1 metabolome in infectious inflam-
in regulating host responses in arthritis. Mol mation. J Immunol 197(11):4444–4452. https://doi.
Asp Med 58:57–64. https://doi.org/10.1016/j. org/10.4049/jimmunol.1600837
mam.2017.04.005 17. Calder PC (2011) Fatty acids and inflammation:
6. Quiros M, Nusrat A (2019) Saving problematic the cutting edge between food and pharma. Eur
mucosae: SPMs in intestinal mucosal inflammation J Pharmacol 668(Suppl 1):S50–S58. https://doi.
and repair. Trends Mol Med 25:124–135. https://doi. org/10.1016/j.ejphar.2011.05.085
org/10.1016/j.molmed.2018.12.004 18. De Caterina R (2011) n-3 fatty acids in cardiovascular
7. Serhan CN, Levy BD (2018) Resolvins in inflamma- disease. N Engl J Med 364(25):2439–2450. https://
tion: emergence of the pro-resolving superfamily of doi.org/10.1056/NEJMra1008153
mediators. J Clin Invest 128(7):2657–2669. https:// 19. Crawford MA, Broadhurst CL, Guest M, Nagar
doi.org/10.1172/JCI97943 A, Wang Y, Ghebremeskel K, Schmidt WF (2013)
8. Serhan CN (2017) Discovery of specialized pro- A quantum theory for the irreplaceable role of
resolving mediators marks the dawn of resolution docosahexaenoic acid in neural cell signalling
physiology and pharmacology. Mol Asp Med 58:1– throughout evolution. Prostaglandins Leukot Essent
11. https://doi.org/10.1016/j.mam.2017.03.001 Fatty Acids 88(1):5–13. https://doi.org/10.1016/j.
9. Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang plefa.2012.08.005
N, Gronert K (2000) Novel functional sets of lipid- 20. Dalli J, Colas RA, Serhan CN (2013) Novel n-3
derived mediators with antiinflammatory actions gen- immunoresolvents: structures and actions. Sci Rep
erated from omega-3 fatty acids via cyclooxygenase 3:1940. https://doi.org/10.1038/srep01940
2-nonsteroidal antiinflammatory drugs and transcel- 21. Lemaitre RN, Tanaka T, Tang W, Manichaikul A, Foy
lular processing. J Exp Med 192(8):1197–1204 M, Kabagambe EK, Nettleton JA, King IB, Weng LC,
10. Serhan CN, Hong S, Gronert K, Colgan SP, Devchand Bhattacharya S, Bandinelli S, Bis JC, Rich SS, Jacobs
PR, Mirick G, Moussignac RL (2002) Resolvins: a DR Jr, Cherubini A, McKnight B, Liang S, Gu X,
family of bioactive products of omega-3 fatty acid Rice K, Laurie CC, Lumley T, Browning BL, Psaty
transformation circuits initiated by aspirin treatment BM, Chen YD, Friedlander Y, Djousse L, Wu JH,
that counter proinflammation signals. J Exp Med Siscovick DS, Uitterlinden AG, Arnett DK, Ferrucci
196(8):1025–1037 L, Fornage M, Tsai MY, Mozaffarian D, Steffen LM
11. Serhan CN, Yang R, Martinod K, Kasuga K, Pillai (2011) Genetic loci associated with plasma phospho-
PS, Porter TF, Oh SF, Spite M (2009) Maresins: novel lipid n-3 fatty acids: a meta-analysis of genome-wide
7 Novel n-3 Docosapentaneoic Acid-Derived Pro-resolving Mediators Are Vasculoprotective… 75
association studies from the CHARGE consortium. 32. Buckley CD, McGettrick HM (2018) Leukocyte traf-
PLoS Genet 7(7):e1002193. https://doi.org/10.1371/ ficking between stromal compartments: lessons from
journal.pgen.1002193 rheumatoid arthritis. Nat Rev Rheumatol 14(8):476–
22. Serhan CN, Savill J (2005) Resolution of inflamma- 487. https://doi.org/10.1038/s41584-018-0042-4
tion: the beginning programs the end. Nat Immunol 33. Tabas I, Lichtman AH (2017) Monocyte-macrophages
6(12):1191–1197. https://doi.org/10.1038/ni1276 and T cells in atherosclerosis. Immunity 47(4):621–
23. Aursnes M, Tungen JE, Vik A, Colas R, Cheng CY, 634. https://doi.org/10.1016/j.immuni.2017.09.008
Dalli J, Serhan CN, Hansen TV (2014) Total synthe- 34. Pistorius K, Souza PR, De Matteis R, Austin-
sis of the lipid mediator PD1n-3 DPA: configurational Williams S, Primdahl KG, Vik A, Mazzacuva F,
assignments and anti-inflammatory and pro-resolving Colas RA, Marques RM, Hansen TV, Dalli J (2018)
actions. J Nat Prod 77(4):910–916. https://doi. PDn-3 DPA pathway regulates human monocyte dif-
org/10.1021/np4009865 ferentiation and macrophage function. Cell Chem
24. Gobbetti T, Dalli J, Colas RA, Federici Canova D, Biol 25(6):749–760 e749. https://doi.org/10.1016/j.
Aursnes M, Bonnet D, Alric L, Vergnolle N, Deraison chembiol.2018.04.017
C, Hansen TV, Serhan CN, Perretti M (2017) 35. Primdahl KG, Tungen JE, De Souza PRS, Colas RA,
Protectin D1n-3 DPA and resolvin D5n-3 DPA are Dalli J, Hansen TV, Vik A (2017) Stereocontrolled
effectors of intestinal protection. Proc Natl Acad Sci synthesis and investigation of the biosynthetic trans-
U S A 114(15):3963–3968. https://doi.org/10.1073/ formations of 16(S),17(S)-epoxy-PDn-3 DPA. Org
pnas.1617290114 Biomol Chem 15(40):8606–8613. https://doi.
25. Colas RA, Souza PR, Walker ME, Burton M, Zaslona org/10.1039/c7ob02113e
Z, Curtis AM, Marques RM, Dalli J (2018) Impaired 36. Chiang N, Serhan CN (2017) Structural elucidation
production and diurnal regulation of vascular RvDn-3 and physiologic functions of specialized pro-resolving
DPA increase systemic inflammation and cardiovas- mediators and their receptors. Mol Asp Med 58:114–
cular disease. Circ Res 122(6):855–863. https://doi. 129. https://doi.org/10.1016/j.mam.2017.03.005
org/10.1161/CIRCRESAHA.117.312472 37. Dalli J, Serhan C (2016) Macrophage proresolv-
26. Ingle KA, Kain V, Goel M, Prabhu SD, Young ME, ing mediators-the when and where. Microbiol
Halade GV (2015) Cardiomyocyte-specific Bmal1 Spectr 4 (3). https://doi.org/10.1128/microbiolspec.
deletion in mice triggers diastolic dysfunction, extra- MCHD-0001-2014
cellular matrix response, and impaired resolution 38. Dalli J, Chiang N, Serhan CN (2015) Elucidation of
of inflammation. Am J Physiol Heart Circ Physiol novel 13-series resolvins that increase with atorvas-
309(11):H1827–H1836. https://doi.org/10.1152/ tatin and clear infections. Nat Med 21(9):1071–1075.
ajpheart.00608.2015 https://doi.org/10.1038/nm.3911
27. McAlpine CS, Swirski FK (2016) Circadian influence 39. Primdahl KG, Aursnes M, Walker ME, Colas RA,
on metabolism and inflammation in atherosclerosis. Serhan CN, Dalli J, Hansen TV, Vik A (2016) Synthesis
Circ Res 119(1):131–141. https://doi.org/10.1161/ of 13(R)-Hydroxy-7Z,10Z,13R,14E,16Z,19Z
CIRCRESAHA.116.308034 Docosapentaenoic acid (13R-HDPA) and its biosyn-
28. Puttonen S, Oksanen T, Vahtera J, Pentti J, Virtanen thetic conversion to the 13-series Resolvins. J Nat
M, Salo P, Kivimaki M (2010) Is shift work a risk fac- Prod 79(10):2693–2702. https://doi.org/10.1021/acs.
tor for rheumatoid arthritis? The Finnish public sec- jnatprod.6b00634
tor study. Ann Rheum Dis 69(4):779–780. https://doi. 40. Claria J, Serhan CN (1995) Aspirin triggers previ-
org/10.1136/ard.2008.099184 ously undescribed bioactive eicosanoids by human
29. Krump E, Picard S, Mancini J, Borgeat P (1997)
endothelial cell-leukocyte interactions. Proc Natl
Suppression of leukotriene B4 biosynthesis by endog- Acad Sci U S A 92(21):9475–9479
enous adenosine in ligand-activated human neutro- 41. Planaguma A, Pfeffer MA, Rubin G, Croze R, Uddin
phils. J Exp Med 186(8):1401–1406 M, Serhan CN, Levy BD (2010) Lovastatin decreases
30. Viola JR, Lemnitzer P, Jansen Y, Csaba G, Winter acute mucosal inflammation via 15-epi-lipoxin
C, Neideck C, Silvestre-Roig C, Dittmar G, Doring A4. Mucosal Immunol 3(3):270–279. https://doi.
Y, Drechsler M, Weber C, Zimmer R, Cenac N, org/10.1038/mi.2009.141
Soehnlein O (2016) Resolving lipid mediators 42. Walker ME, Souza PR, Colas RA, Dalli J (2017)
Maresin 1 and Resolvin D2 prevent atheroprogres- 13-series resolvins mediate the leukocyte-platelet
sion in mice. Circ Res 119(9):1030–1038. https://doi. actions of atorvastatin and pravastatin in inflamma-
org/10.1161/CIRCRESAHA.116.309492 tory arthritis. FASEB J 31(8):3636–3648. https://doi.
31. Fredman G, Hellmann J, Proto JD, Kuriakose G, Colas org/10.1096/fj.201700268
RA, Dorweiler B, Connolly ES, Solomon R, Jones 43. Markworth JF, Kaur G, Miller EG, Larsen AE,
DM, Heyer EJ, Spite M, Tabas I (2016) An imbalance Sinclair AJ, Maddipati KR, Cameron-Smith D (2016)
between specialized pro-resolving lipid mediators Divergent shifts in lipid mediator profile following
and pro-inflammatory leukotrienes promotes instabil- supplementation with n-3 docosapentaenoic acid and
ity of atherosclerotic plaques. Nat Commun 7:12859. eicosapentaenoic acid. FASEB J 30(11):3714–3725.
https://doi.org/10.1038/ncomms12859 https://doi.org/10.1096/fj.201600360R
Aspects of Prostaglandin Glycerol
Ester Biology 8
Philip J. Kingsley, Carol A. Rouzer,
Amanda J. Morgan, Sachin Patel,
and Lawrence J. Marnett
Abstract
analogous to canonical prostaglandins. This
The Cyclooxygenase enzymes (COX-1 and chapter reviews the literature detailing the
COX-2) incorporate 2 molecules of O2 into production, metabolism, and bioactivity of
arachidonic acid (AA), resulting in an array of these compounds, as well as their detection in
bioactive prostaglandins. However, much intact animals.
work has been done showing that COX-2 will
perform this reaction on several different Keywords
AA-containing molecules, most importantly, Endocannabinoid · Cannabinoid ·
the endocannabinoid 2-arachidonoylglycerol Cyclooxygenase · COX-2 · Inflammation ·
(2-AG). The products of 2-AG oxygenation, Prostaglandin · Prostaglandin glyceryl ester ·
prostaglandin glycerol esters (PG-Gs), are Transgenic mouse · Anandamide ·
2-Arachidonoyl glycerol
COR
COR
R = OH OH
OH COR
COX-2
N
H
OH HO
O
15- & 11-HETE O
OH PGF2a COR
Thromboxane
N
OH HO
Synthase O
X Synthasea
H OH
O COR O
COOH
OH O
O OH HO
N OH PGD2 OH
O P O +
Synthase PGI2
O
Synthase COR
OH
+ PGE2
O OH HO
NH3 Synthase
O P O
O COR
O
O
OH
O
COR HO OH
HO OH
Scheme 8.1 Schematic of COX-2-mediated oxygen- (a – PGH2 derived from AEA or 2-AG is not a substrate
ation of various arachidonoyl-containing lipid species. for thromboxane synthase. It is unknown if PGH2-LPC or
PGH2-LPE are acted upon by this enzyme)
8.2 PG-Gs in in Vitro and Cellular Rouzer and Marnett [12] further investigated
Systems the cellular production of PG-Gs in a more physi-
ologically relevant setting by isolating murine
The majority of reports regarding non-AA- resident peritoneal macrophages (RPMs). These
derived products of COX-2 oxygenation used cells are known to have a relatively high level of
both reconstituted enzymatic and cellular sys- AA in their phospholipid pool and are able to
tems to characterize the oxygenated products as synthesize COX-2 and generate large amounts of
well as the mechanism of their production. In the PGs in response to a variety of inflammatory
initial report of PG-G formation, Kozak et al. [2] stimuli. Here, RPMs were pretreated with LPS to
used purified murine and human COX-2 for induce COX-2 expression and then challenged
assessment of the kinetics (via O2 uptake) of with zymosan, which stimulated the release of
2-AG oxygenation and the identification of 2-AG. The authors showed that PGE2-G and
COX-2 reaction products. These studies estab- PGI2-G were produced by RPMs without the
lished that, when assayed individually, COX-2 addition of exogenous 2-AG, and these PG-Gs
utilizes AA and 2-AG with similar efficiencies were released from the cells. As in the case of
(kcat/Km = 2.4 s−1 μM−1 and 4.0 s−1 μM−1 for AA RAW264.7 cells, the primary PG-G products in
and 2-AG, respectively, for human COX-2 and RPMs were consistent with the primary PG prod-
2.5 s−1 μM−1 and 2.3 s−1 μM−1 for AA and 2-AG, ucts, which were PGE2 and PGI2.
respectively, for murine COX-2). The authors Based on kinetic analyses of COX-2-mediated
used the RAW264.7 murine macrophage-like cell oxygenation of AA and 2-AG in vitro, which had
line to confirm that the observed enzymatic reac- shown similar enzymatic efficiency for the two
tions could also take place in a cellular setting, substrates, one would predict that the ratio of
where 2-AG would be subject to metabolism by PG-Gs to PGs produced in cells should be simi-
several different pathways. Since RAW264.7 lar to the ratio of available 2-AG to AA. However,
cells do not express COX-2 under basal condi- in zymosan-stimulated RPMs, the ratio of total
tions, they were pretreated with interferon-γ PG to PG-G production was roughly 1000:1,
(INF-γ) and bacterial lipopolysaccharide (LPS) whereas the ratio of free AA to 2-AG in these
to induce the COX-2 enzyme. The investigators cells was 10:1. The much lower than expected
established that exogenous 2-AG can be taken up production of PG-Gs relative to PGs based on
by cells, processed to PGD2-G via COX-2, and substrate availability could not be explained on
secreted into the extracellular medium. They fur- the basis of PG-G hydrolysis [12]. These obser-
ther demonstrated the formation of PGD2-G from vations led to more extensive kinetic analyses of
endogenous 2-AG following stimulation of LPS- 2-AG and AA oxygenation by COX-2 via exper-
and IFN-γ-pretreated RAW264.7 cells with iono- iments in which both substrates were present.
mycin. The formation of PGD2-G by the cells Results of these kinetic studies were consistent
was consistent with the fact that they produce with previously published data suggesting that
predominantly PGD2 from AA. In subsequent the homodimeric COX-2 enzyme behaves as a
studies, Kozak et al. demonstrated the formation heterodimer with one subunit acting as the cata-
of PGE2-G and PGF2α-G from exogenous 2-AG lytic site and the other serving an allosteric func-
by human HCA-7 colon adenocarcinoma cells, tion. Specifically, the results supported the
which constitutively express COX-2 [10]. In hypothesis that AA and 2-AG compete with each
addition, Siegmund et al. reported the synthesis other for both the catalytic and allosteric sites
of PGD2-G from exogenously provided 2-AG by and that binding of either substrate in the alloste-
hepatic stellate cells isolated from mice pre- ric site suppresses 2-AG oxygenation and pro-
treated with bile duct ligation to induce liver motes AA oxygenation. Consequently, when
fibrosis. The investigators confirmed that bile both substrates are present, oxygenation of AA is
duct ligation led to induction of COX-2 expres- favored over that of 2-AG [13]. It is not clear
sion in the hepatic stellate cells [11]. whether this phenomenon fully explains the
80 P. J. Kingsley et al.
observed relatively low yield of PG-Gs produced PG-G-associated biological activity immediately
by zymosan-stimulated RPMs. In fact, COX-2 led to the question of whether or not the effects
requires peroxide-dependent activation before were mediated by known endocannabinoid or
oxygenation of any substrate can occur, and a prostanoid receptors as opposed to distinct recep-
report from Musee and Marnett indicates that tors. An initial pharmacologic study ruled out sig-
higher levels of peroxide are required to activate nificant activity of PGE2-G at the four EP receptors
2-AG oxygenation compared to AA oxygenation that modulate the effects of PGE2, as well as the
[14]. Other factors, such as the actual availability FP, DP, TP, and IP receptors [16]. More recently,
of free 2-AG and AA at the COX-2 active site Woodward et al. used the more metabolically and
within the cell, may also play a role. chemically stable amide derivatives of the 1(3)-
While investigating the phenomenon of and 2-glyceryl esters of PGE2 and PGF2α to show
substrate-selective inhibition, in which an that neither compound has significant activity at
inhibitor blocks COX-2-dependent oxygen- any of the prostanoid receptors. The highest activ-
ation of 2-AG more strongly than that of AA, ity was noted in the case of PGE2-serinolamide
Duggan et al. [15] employed a model utilizing (the stable analog of the 2-glyceryl ester of PGE2)
dorsal root ganglia (DRG). DRGs were har- at the EP3 receptor, for which an EC50 of 500 nM
vested, plated, and treated with a cocktail of was observed. EC50 values were > 3000 nM for all
compounds that included LPS and IFN-γ to tested analogs at all other receptors [17].
induce COX-2. Ionomycin was subsequently The earliest report of PG-G-mediated biologi-
added to stimulate release of 2-AG, resulting in cal activity came from Nirodi et al., who found
the production of PG-Gs. The predominant that PGE2-G, but not PGF2α-G or PGD2-G, caused
products of 2-AG oxygenation were PGE2-G Ca2+ mobilization in the murine RAW264.7
and PGF2α-G. Interestingly, ethanolamide macrophage-like cell line at picomolar concen-
derivatives of PGs were also produced by the trations. Ca2+ mobilization was secondary to
cells, in addition to free acid PGs. The absolute increases in intracellular inositol
quantities of the various products were not 1,4,5-trisphospate, and it resulted in the activa-
reported. With this model, the authors showed tion of protein kinase C and, subsequently, extra-
that within the physiological system of DRGs, cellular signal regulated kinases (ERKs) 1 and 2.
the R-profen class of inhibitors acted as sub- Importantly, the authors also showed that PGE2
strate-selective inhibitors. did not elucidate these effects [16]. In follow-up
It should be noted that other reports have dem- studies, Richie-Jannetta et al. demonstrated
onstrated COX-2-dependent effects of 2-AG in a PGE2-G-mediated Ca2+ mobilization in the
variety of cell systems, suggesting that these H1819 human non-small-cell lung cancer cell
effects may be due to conversion of the endocan- line. In these studies, similar activity was
nabinoid to a PG-G. However, in many of these observed regardless of whether the PGE2 moiety
studies, the actual synthesis of a PG-G in the tis- was attached at the sn-1 or sn-2 position of the
sue or cells of interest was not confirmed. We glycerol, and PGF2α-G exhibited activity similar
have limited this discussion to cases in which to that of PGE2-G. In contrast, the corresponding
PG-G formation was demonstrated analytically. free acid prostanoids were inactive [18].
A number of studies have suggested a potential
role for PG-Gs in the nervous system. Sang et al.
8.3 Biological Activity of PG-Gs [19] demonstrated that PGE2-G, PGF2α-G, and
PGD2-G increased miniature inhibitory postsyn-
Concurrent with the elucidation of the production aptic currents (mIPSCs) in murine hippocampal
and control of PG-Gs, several investigators have neurons. Shortly thereafter, the same group
reported interesting and potent biological activi- reported that these three PG-Gs enhanced minia-
ties of PG-Gs, both in cellular systems and in ture excitatory postsynaptic currents (mEPSCs) in
intact mammals. However, the discovery of any hippocampal neurons, an indication of enhanced
8 Aspects of Prostaglandin Glycerol Ester Biology 81
glutamatergic synaptic transmission [20]. In both tis model; the related compounds PGD2 and
cases, 2-AG had the opposite effect, a role for the PGD2-EA did not have similar effects [25].
CB1 receptor was ruled out, and the involvement Raman and colleagues demonstrated that the
of both IP3 and ERKs was implicated. The PGE2- nonenzymatic breakdown product of PGD2-G,
G-mediated enhancement of mEPSCs was also 15-deoxy-Δ12,14-PGJ2-G (15d-PGJ2-G), activates
found to induce neuronal injury and/or death. Yang the PPARγ receptor leading to decreased tran-
et al. also reported an effect of PGE2-G, PGF2α-G, scriptional activity of nuclear factor of activated
and PGD2-G on hippocampal neurotransmission. T cells (NFAT), and ultimately, reduced IL-2
In their case, an increase in basal synaptic trans- expression in the human Jurkat T lymphoblas-
mission and long-term potentiation in hippocam- toid cell line. The researchers suggest that
pal slices was observed that, consistent with prior 15d-PGJ2-G mediates the 2-AG-dependent sup-
findings, was dependent on IP3-mediated Ca2+ pression of IL-2 production in these cells [26,
mobilization and ERK signaling [21]. 27]. In one of the few studies in intact animals,
Commensurate with the neurotoxic effects noted Hu et al. reported that intraplantar administra-
by Sang et al., Valdeolivas and colleagues reported tion of PGE2-G in rats produced hyperalgesia
that PGE2-G was neurotoxic to the M-213-2O fetal and allodynia (8). In this study, PGE2 induced
rat striatum-derived cell line. Their studies sug- hyperalgesia in a manner similar to
gested that PGE2-G is a mediator of malonate- PGE2-G. However, while EP1–4 receptor antago-
dependent neurotoxicity, which in M-213-2O nists blocked the hyperalgesic effect of PGE2,
cells, serves as a model of Huntington’s disease they did not block the hyperalgesic effects of
[22]. In studies involving tissues other than the PGE2-G, suggesting that the PGE2-G response is
hippocampus, Lindgren and colleagues reported mediated by a pathway distinct from that of
that PGE2-G induces an enhancement of stimulus- PGE2. Nevertheless, this study highlights one of
evoked neurotransmitter release by the presynap- the major challenges of elucidating the role of
tic neuron at the neuromuscular junction. They PG-Gs in vivo. The compounds are subject to
further demonstrated that the effect is dependent hydrolysis by multiple esterases such that the
on production of nitric oxide and does not involve half-life of PG-Gs in biological tissues is short
the EP1 or EP2 receptors for PGE2 [23]. Woodward (see below). Consequently, some reports of
et al. demonstrated that the amide derivatives of PG-G-mediated biological effects are actually
the 1(3)- and 2-glycerol esters of PGE2 lower due to free PGs obtained following hydrolysis.
intraocular pressure in canine eyes, whereas the We have limited this discussion to cases in which
comparable derivatives of PGF2α are much less the observed pharmacology appears to be due
effective. In the monkey eye, the derivative of the directly to the action of PG-Gs.
1(3)-glycerol ester of PGE2 was more effective A common finding in these studies of the bio-
than the 2-glycerol ester [17]. logical activities of PG-Gs is that neither 2-AG nor
Most other actions of PG-Gs have been iden- the free acid prostaglandins mediate the same
tified in various models of immune or inflamma- effects, and in many cases, a role for CB or pros-
tory responses. Alhouayek et al. [24] tanoid receptors was specifically ruled out. This
demonstrated that PGD2-G suppresses while strongly implies that there is one (or several)
PGE2-G and PGF2α-G increase IL-1β secretion receptors specific for PG-Gs. In support of this
by murine J774 macrophage-like cells. Their hypothesis, Bruser et al. recently reported that
accompanying exploration of LPS-stimulated PGE2-G is a potent ligand of the P2Y6 receptor
2-AG metabolism by the cells suggested that [28]. In this study, the authors identified G protein-
2-AG-dependent decreases in macrophage acti- coupled receptors (GPCRs) in PGE2-G response-
vation in response to LPS is mediated by positive cells via RNA sequence analysis. From a
PGD2-G. In later work, Alhouayek et al. reported pool of these candidate GPCRs, the authors estab-
that PGD2-G reduced symptoms of inflammation lished that the UDP receptor P2Y6 is also a recep-
in a murine dextran sodium sulfate-induced coli- tor for PGE2-G. In fact, they report that PGE2-G
82 P. J. Kingsley et al.
activates P2Y6 with an EC50 of 1.2 pM, compared Our most recent report [31] employed homog-
to an EC50 of 78 nM for the canonical ligand, UDP. enization of brain tissue via sonication in aceto-
nitrile. No secondary purification was employed.
Phospholipids are known to be less soluble than
8.4 Analytical Methodology eicosanoids in acetonitrile, and it has been our
for PG-G Quantitation experience that little interference is experienced
in the analysis of PG-Gs and related lipids with
While several types of analytical methodologies this method. PG-Gs were chromatographed on a
have been applied to lipid detection and quantita- C18 column with ammonium acetate in the
tion in the last several decades, reports of PG-G mobile phase to promote the generation of the
quantitation have employed almost exclusively [M + NH4]+ ion. For unknown reasons, PG-Gs
LC-MS techniques. Previously, our group pro- preferentially co-ordinate with the ammonium
duced protocols [29] and reviews [30] regarding cation, and this complex provides several candi-
this type of analysis, and other researchers have date fragment ions (Fig. 8.1), giving the
employed similar methodologies. The typical researcher several options for SRM analysis.
work flow involves homogenization of cells or PG-Gs will also coordinate with Na+, but typi-
animal tissue followed by a secondary purifica- cally sodium complexes give rise to relatively
tion step (often solid phase extraction (SPE)). weak product spectra compared to those of H+
Finally, an aliquot of the dried and reconstituted and NH4+ complexes, rendering this ionization
sample is injected onto an LC-MS system where method not useful for SRM detection.
the analytes are chromatographed on a reverse- In one of the few studies of PG-G detection in
phase column and detected by mass spectrometry samples from an intact animal, Hu et al. (8) mea-
via selected reaction monitoring (SRM) (Note: sured PGE2-G in rat hind paws by homogenizing
sometimes researchers use the term “multiple the tissues in methanol, then subjecting the
reaction monitoring” (MRM), which is equiva- diluted homogenates to purification via C18
lent to SRM). SPE. Samples were finally analyzed on a LC-MS/
MS system, where PGE2-G was ionized via
Fig. 8.1 Product ion spectrum of PGE2-G complexed to 444 and the Q3 m/z to any of the several product masses
with an ammonium cation (m/z of [M + NH4]+ = 444.2). observed
SRM analysis may be accomplished by setting the Q1 m/z
8 Aspects of Prostaglandin Glycerol Ester Biology 83
mouse brain setting. Towards this end, we acquired concurrently with the highly selective COX-2
the Thy-1 hCOX-2 mouse. These mice overexpress inhibitor lumiracoxib (LMX) [42]. The LMX-
human COX-2 (hCOX-2) primarily in the neurons treated animals showed LMX levels in brain tissue
of the striatum, amygdala, cerebral cortex, and hip- of 200–400 pmol/g and an almost complete elimi-
pocampus [40]. Litters resulting from the breeding nation of PG-G species (Fig. 8.4). Interestingly,
of these mice contain both wild-type and transgenic PGE2 and PGD2 were also markedly reduced.
(Tg) overexpressor animals. The experimental data
shown below were generated with 60–90 day-old
mice that were obtained in this way [31]. 8.5.2 PG-Gs in Response
Western blotting analysis verified the presence of to Systemic Inflammation
hCOX-2 in the brain tissue (prefontal cortex and hip-
pocampus) of the Tg animals (Fig. 8.2) [31]. Having established a model system in which
However, PG-Gs were not detected in analyses of Tg robust PG-Gs can be observed with Tg mice, we
or WT brain tissue (Fig. 8.3). We hypothesized that wanted to explore the possibility of observing
an increase in substrate levels (i.e., 2-AG) may afford PG-Gs without genetic manipulation. As previ-
detectable PG-G generation. Thus, WT and Tg mice ous reports of COX-2 products in vivo involved
were dosed with the monoacylglycerol lipase the administration of inflammatory stimuli, we
(MAGL) inhibitor, JZL184 [41]. In agreement with treated WT mice with LPS (3 mg/kg once per day
previous literature reports, 2-AG was increased in the for 2 days) with and without concurrent adminis-
JZL184-dosed subjects (data not shown), and PG-Gs tration of JZL184. Some of the LPS-treated mice
were detected (Fig. 8.3). Levels of PGF2α-G, PGE2-G, produced PG-Gs, albeit at levels 1–2 orders of
and PGD2-G were found to be in the range of magnitude lower than those of the Tg + JZL184
5–50 pmol per g of tissue (wet weight). subjects (Fig. 8.5). Note that tissues from animals
The COX-2 dependence of PG-G production treated with LPS alone exhibited a trend toward
was tested by dosing the Tg + JZL184 animals increased PG-G levels, though this did not reach
-2
17). (Figure reproduced Cerebellum PFC Pfc
OX
by permission from mC
Morgan et al. [31].
Copyright 2018, hCOX-2
American Chemical
Society)
Actin
Fig. 8.3 Detection and quantitation of PG-G species in permission from Morgan et al. [31]. Copyright 2018,
Tg mice. PG-Gs are observed only in the Tg + JZL184 American Chemical Society)
subjects (∗∗ indicated p < 0.01). (Figure reproduced by
8 Aspects of Prostaglandin Glycerol Ester Biology 85
Fig. 8.4 Effect of LMX on lipid species in brain tissue of Y-axis. (Figure reproduced by permission from Morgan
Tg + JZL184 animals. (∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ et al. [31]. Copyright 2018, American Chemical Society)
p < 0.001, ∗ p < 0.0001). Note: Figure d has a non-zero
Fig. 8.5 Effect of systemic LPS treatment on brain lipid levels of Tg and Tg + JZL184 subjects (∗p < 0.05, ∗∗
p < 0.01). (Figure reproduced by permission from Morgan et al. [31]. Copyright 2018, American Chemical Society)
86 P. J. Kingsley et al.
statistical significance when compared to con- synthesis can be detected and monitored under
trols. It is interesting to note, however, that there conditions that will allow us to assess their role in
was no statistical significance between PG-G lev- physiological or pathophysiological processes.
els in the brains of animals treated with LPS Such models will also allow further study of
alone and those treated with LPS and JZL184, additional aspects of COX-2 biology, such as the
although levels in the latter animals were statisti- in vivo relevance of substrate selective
cally different from those in the controls. These inhibition.
findings suggest that LPS treatment increased
PG-G levels in the brains of the mice even in the Acknowledgments This work was supported by
absence of JZL184; however, a larger study will CA89450, GM15431 and (cite Sachin’s grant(s)).
be necessary to test this hypothesis.
References
8.6 Conclusions and Future
1. Yu M, Ives D, Ramesha CS (1997) Synthesis of prosta-
Directions glandin E2 ethanolamide from anandamide by cyclo-
oxygenase-2. J Biol Chem 272(34):21181–21186
The ability of COX-2 to metabolize 2-AG and 2. Kozak KR, Rowlinson SW, Marnett LJ
other ester and amide derivatives of AA is now (2000) Oxygenation of the endocannabinoid,
2-arachidonylglycerol, to glyceryl prostaglandins
well established. However, the physiological rel- by cyclooxygenase-2. J Biol Chem 275(43):33744–
evance of this phenomenon remains unsettled. It 33749. https://doi.org/10.1074/jbc.M007088200
is clear that PG-Gs exert interesting biological 3. Prusakiewicz JJ, Kingsley PJ, Kozak KR, Marnett LJ
activities in tissues relevant to their site of pro- (2002) Selective oxygenation of N-arachidonylglycine
by cyclooxygenase-2. Biochem Biophys Res
duction. Furthermore, their high potencies in Commun 296(3):612–617
many systems (with EC50’s in the pM range) 4. Dong L, Zou H, Yuan C, Hong YH, Uhlson CL,
suggest that only very low levels of these lipid Murphy RC, Smith WL (2016) Interactions of
mediators may be necessary to exert important 2-O-arachidonylglycerol ether and ibuprofen with
the allosteric and catalytic subunits of human COX-2.
physiological functions. Clearly, a major impedi- J Lipid Res 57(6):1043–1050. https://doi.org/10.1194/
ment to elucidating the potential relevance of jlr.M067512
PG-G formation is our inability to detect these 5. Liu X, Moon SH, Jenkins CM, Sims HF, Gross
compounds in vivo. One should note that this RW (2016) Cyclooxygenase-2 mediated oxida-
tion of 2-Arachidonoyl-Lysophospholipids identi-
does not preclude relevance. For example, the fies unknown lipid signaling pathways. Cell Chem
highly relevant eicosanoids thromboxane A2 and Biol 23(10):1217–1227. https://doi.org/10.1016/j.
prostacyclin (PGI2) are not detected in tissue or chembiol.2016.08.009
cellular samples. Rather, their production is mon- 6. Gatta L, Piscitelli F, Giordano C, Boccella S, Lichtman
A, Maione S, Di Marzo V (2012) Discovery of pros-
itored through the detection of their nonenzy- tamide F2alpha and its role in inflammatory pain and
matic breakdown products, thromboxane B2 and dorsal horn nociceptive neuron hyperexcitability.
6-keto-PGF1α. Unfortunately, in the case of PLoS One 7(2):e31111. https://doi.org/10.1371/jour-
PG-Gs, their hydrolysis products are free acid nal.pone.0031111
7. Silvestri C, Martella A, Poloso NJ, Piscitelli F,
PGs. Thus, it is impossible to know whether PGs Capasso R, Izzo A, Woodward DF, Di Marzo V (2013)
detected in a biological sample may have initially Anandamide-derived prostamide F2alpha negatively
been synthesized from 2-AG or AA. regulates adipogenesis. J Biol Chem 288(32):23307–
Given these challenges, the Tg mice described 23321. https://doi.org/10.1074/jbc.M113.489906
8. Woodward DF, Liang Y, Krauss AH (2008)
here offer an intriguing model that can be used to Prostamides (prostaglandin-ethanolamides) and their
detect PG-G formation in vivo, albeit under con- pharmacology. Br J Pharmacol 153(3):410–419.
ditions that are not totally physiological. Of per- https://doi.org/10.1038/sj.bjp.0707434
haps greater interest are our results from 9. Woodward DF, Wang JW, Poloso NJ (2013) Recent
progress in prostaglandin F2alpha ethanolamide
LPS-stimulated animals suggesting that, eventu- (prostamide F2alpha) research and therapeutics.
ally, models may be found in which PG-G bio-
8 Aspects of Prostaglandin Glycerol Ester Biology 87
Pharmacol Rev 65(4):1135–1147. https://doi. 20. Sang N, Zhang J, Chen C (2006) PGE2 glycerol ester,
org/10.1124/pr.112.007088 a COX-2 oxidative metabolite of 2-arachidonoyl glyc-
10. Kozak KR, Crews BC, Morrow JD, Wang LH,
erol, modulates inhibitory synaptic transmission in
Ma YH, Weinander R, Jakobsson PJ, Marnett mouse hippocampal neurons. J Physiol 572. (Pt 3:735–
LJ (2002) Metabolism of the endocannabinoids, 745. https://doi.org/10.1113/jphysiol.2006.105569
2-arachidonylglycerol and anandamide, into prosta- 21. Yang H, Zhang J, Andreasson K, Chen C (2008)
glandin, thromboxane, and prostacyclin glycerol esters COX-2 oxidative metabolism of endocannabinoids
and ethanolamides. J Biol Chem 277(47):44877– augments hippocampal synaptic plasticity. Mol Cell
44885. https://doi.org/10.1074/jbc.M206788200 Neurosci 37(4):682–695. https://doi.org/10.1016/j.
11. Siegmund SV, Wojtalla A, Schlosser M, Schildberg mcn.2007.12.019
FA, Knolle PA, Nusing RM, Zimmer A, Strassburg 22. Valdeolivas S, Pazos MR, Bisogno T, Piscitelli
CP, Singer MV (2016) Cyclooxygenase-2 contributes F, Iannotti FA, Allara M, Sagredo O, Di Marzo
to the selective induction of cell death by the endocan- V, Fernandez-Ruiz J (2013) The inhibition of
nabinoid 2-arachidonoyl glycerol in hepatic stellate 2-arachidonoyl-glycerol (2-AG) biosynthesis, rather
cells. Biochem Biophys Res Commun 470(3):678– than enhancing striatal damage, protects striatal
684. https://doi.org/10.1016/j.bbrc.2016.01.083 neurons from malonate-induced death: a potential
12. Rouzer CA, Marnett LJ (2005) Glycerylprostaglandin role of cyclooxygenase-2-dependent metabolism of
synthesis by resident peritoneal macrophages in 2-AG. Cell Death Dis 4:e862. https://doi.org/10.1038/
response to a zymosan stimulus. J Biol Chem cddis.2013.387
280(29):26690–26700. https://doi.org/10.1074/jbc. 23. Lindgren CA, Newman ZL, Morford JJ, Ryan SB,
M501021200 Battani KA, Su Z (2013) Cyclooxygenase-2, prosta-
13. Mitchener MM, Hermanson DJ, Shockley EM, Brown glandin E2 glycerol ester and nitric oxide are involved
HA, Lindsley CW, Reese J, Rouzer CA, Lopez CF, in muscarine-induced presynaptic enhancement at
Marnett LJ (2015) Competition and allostery govern the vertebrate neuromuscular junction. J Physiol
substrate selectivity of cyclooxygenase-2. Proc Natl 591(19):4749–4764. https://doi.org/10.1113/
Acad Sci U S A 112(40):12366–12371. https://doi. jphysiol.2013.256727
org/10.1073/pnas.1507307112 24. Alhouayek M, Masquelier J, Cani PD, Lambert
14. Musee J, Marnett LJ (2012) Prostaglandin H synthase- DM, Muccioli GG (2013) Implication of the
2-catalyzed oxygenation of 2-arachidonoylglycerol is anti-inflammatory bioactive lipid prostaglandin
more sensitive to peroxide tone than oxygenation of D2-glycerol ester in the control of macrophage activa-
arachidonic acid. J Biol Chem 287(44):37383–37394. tion and inflammation by ABHD6. Proc Natl Acad Sci
https://doi.org/10.1074/jbc.M112.381202 U S A 110(43):17558–17563. https://doi.org/10.1073/
15. Duggan KC, Hermanson DJ, Musee J, Prusakiewicz pnas.1314017110
JJ, Scheib JL, Carter BD, Banerjee S, Oates JA, 25. Alhouayek M, Buisseret B, Paquot A, Guillemot-
Marnett LJ (2011) (R)-Profens are substrate-selective Legris O, Muccioli GG (2018) The endogenous bio-
inhibitors of endocannabinoid oxygenation by COX- active lipid prostaglandin D2-glycerol ester reduces
2. Nat Chem Biol 7(11):803–809 murine colitis via DP1 and PPARgamma receptors.
16. Nirodi CS, Crews BC, Kozak KR, Morrow JD,
FASEB J 32(9):5000–5011. https://doi.org/10.1096/
Marnett LJ (2004) The glyceryl ester of prostaglan- fj.201701205R
din E2 mobilizes calcium and activates signal trans- 26. Raman P, Kaplan BL, Kaminski NE (2012) 15-Deoxy-
duction in RAW264.7 cells. Proc Natl Acad Sci U Delta(1)(2),(1)(4)-prostaglandin J(2)-glycerol, a
S A 101(7):1840–1845. https://doi.org/10.1073/ putative metabolite of 2-arachidonyl glycerol and a
pnas.0303950101 peroxisome proliferator-activated receptor gamma
17.
Woodward DF, Poloso NJ, Wang JW (2016) ligand, modulates nuclear factor of activated T cells.
Prostaglandin E2-glyceryl Ester: in vivo evidence for J Pharmacol Exp Ther 342(3):816–826. https://doi.
a distinct pharmacological identity from intraocular org/10.1124/jpet.112.193003
pressure studies. J Pharmacol Exp Ther 358(2):173– 27. Raman P, Kaplan BL, Thompson JT, Vanden Heuvel
180. https://doi.org/10.1124/jpet.116.232512 JP, Kaminski NE (2011) 15-deoxy-delta12,14-
18. Richie-Jannetta R, Nirodi CS, Crews BC, Woodward prostaglandin J2-glycerol ester, a putative metabolite
DF, Wang JW, Duff PT, Marnett LJ (2010) Structural of 2-arachidonyl glycerol, activates peroxisome prolif-
determinants for calcium mobilization by prostaglan- erator activated receptor. Mol Pharmacol 80(1):201–
din E2 and prostaglandin F2alpha glyceryl esters in 209. https://doi.org/10.1124/mol.110.070441
RAW 264.7 cells and H1819 cells. Prostaglandins 28. Bruser A, Zimmermann A, Crews BC, Sliwoski G,
Other Lipid Mediat 92(1–4):19–24. https://doi. Meiler J, Konig GM, Kostenis E, Lede V, Marnett LJ,
org/10.1016/j.prostaglandins.2010.01.003 Schoneberg T (2017) Prostaglandin E2 glyceryl ester
19. Sang N, Zhang J, Chen C (2007) COX-2 oxidative is an endogenous agonist of the nucleotide receptor
metabolite of endocannabinoid 2-AG enhances excit- P2Y6. Sci Rep 7(1):2380. https://doi.org/10.1038/
atory glutamatergic synaptic transmission and induces s41598-017-02414-8
neurotoxicity. J Neurochem 102(6):1966–1977. 29. Kingsley PJ, Rouzer CA, Saleh S, Marnett LJ (2005)
https://doi.org/10.1111/j.1471-4159.2007.04668.x Simultaneous analysis of prostaglandin glyceryl
88 P. J. Kingsley et al.
esters and prostaglandins by electrospray tandem cells. J Biol Chem 289(49):33741–33753. https://doi.
mass spectrometry. Anal Biochem 343(2):203–211. org/10.1074/jbc.M114.582353
https://doi.org/10.1016/j.ab.2005.05.005 37. Savinainen JR, Kansanen E, Pantsar T, Navia-
30. Kudalkar SNKPJ, Marnett LJ (2016) Assay of
Paldanius D, Parkkari T, Lehtonen M, Laitinen T,
endocannabinoid oxidation by Cyclooxygenase-2. Nevalainen T, Poso A, Levonen AL, Laitinen JT
Methods Mol Biol 1412:205–216 (2014) Robust hydrolysis of prostaglandin glyc-
31. Morgan AJ, Kingsley PJ, Mitchener MM, Altemus erol esters by human monoacylglycerol lipase
M, Patrick TA, Gaulden AD, Marnett LJ, Patel S (MAGL). Mol Pharmacol 86(5):522–535. https://doi.
(2018) Detection of Cyclooxygenase-2-derived oxy- org/10.1124/mol.114.094284
genation products of the endogenous cannabinoid 38. Xie S, Borazjani A, Hatfield MJ, Edwards CC, Potter
2-Arachidonoylglycerol in mouse brain. ACS Chem PM, Ross MK (2010) Inactivation of lipid glyceryl
Neurosci 9(7):1552–1559. https://doi.org/10.1021/ ester metabolism in human THP1 monocytes/mac-
acschemneuro.7b00499 rophages by activated organophosphorus insecti-
32. Urquhart P, Wang J, Woodward DF, Nicolaou A
cides: role of carboxylesterases 1 and 2. Chem Res
(2015) Identification of prostamides, fatty acyl etha- Toxicol 23(12):1890–1904. https://doi.org/10.1021/
nolamines, and their biosynthetic precursors in rab- tx1002194
bit cornea. J Lipid Res 56(8):1419–1433. https://doi. 39. Chicca A, Gachet MS, Petrucci V, Schuehly W,
org/10.1194/jlr.M055772 Charles RP, Gertsch J (2015) 4′-O-methylhonokiol
33. Hu SS, Bradshaw HB, Chen JS, Tan B, Walker JM increases levels of 2-arachidonoyl glycerol in mouse
(2008) Prostaglandin E2 glycerol ester, an endog- brain via selective inhibition of its COX-2-mediated
enous COX-2 metabolite of 2-arachidonoylglycerol, oxygenation. J Neuroinflammation 12:89. https://doi.
induces hyperalgesia and modulates NFkappaB activ- org/10.1186/s12974-015-0307-7
ity. Br J Pharmacol 153(7):1538–1549. https://doi. 40. Andreasson KI, Savonenko A, Vidensky S, Goellner
org/10.1038/bjp.2008.33 JJ, Zhang Y, Shaffer A, Kaufmann WE, Worley PF,
34. Kozak KR, Crews BC, Ray JL, Tai HH, Morrow JD, Isakson P, Markowska AL (2001) Age-dependent cog-
Marnett LJ (2001) Metabolism of prostaglandin glyc- nitive deficits and neuronal apoptosis in cyclooxygen-
erol esters and prostaglandin ethanolamides in vitro ase-2 transgenic mice. J Neurosci 21(20):8198–8209
and in vivo. J Biol Chem 276(40):36993–36998. 41. Long JZ, Li W, Booker L, Burston JJ, Kinsey SG,
https://doi.org/10.1074/jbc.M105854200 Schlosburg JE, Pavon FJ, Serrano AM, Selley DE,
35. Vila A, Rosengarth A, Piomelli D, Cravatt B, Marnett Parsons LH, Lichtman AH, Cravatt BF (2009)
LJ (2007) Hydrolysis of prostaglandin glycerol Selective blockade of 2-arachidonoylglycerol hydro-
esters by the endocannabinoid-hydrolyzing enzymes, lysis produces cannabinoid behavioral effects. Nat
monoacylglycerol lipase and fatty acid amide hydro- Chem Biol 5(1):37–44. https://doi.org/10.1038/
lase. Biochemistry 46(33):9578–9585. https://doi. nchembio.129
org/10.1021/bi7005898 42. Marshall PJ BJ, Wasvary J, van Duzer J, Du Z, Scott
36. Manna JD, Wepy JA, Hsu KL, Chang JW, Cravatt BF, G, Rordorf C, Milosavljev S, and, RA, F (2002) The
Marnett LJ (2014) Identification of the major pros- in vitro and in vivo selectivity of COX189, A new and
taglandin glycerol ester hydrolase in human cancer highlyselective inhibitor of COX-2. Ann Rheum Dis
61 (259)
Targeting the COX/mPGES-1/PGE2
Pathway in Neuroblastoma 9
Karin Larsson, Anna Kock, Per Kogner,
and Per-Johan Jakobsson
describe upregulation of PGE2 in different tumors cells that are not targeted by chemotherapy and
[1–5] and the contribution of PGE2 to tumor pro- radiation, continue to produce important media-
gression is linked to several of the hallmarks of tors such as cytokines, chemokines, growth fac-
cancer including increased proliferation of tumor tors and PGE2 that sustains inflammation and
cells [6, 7], withstanding apoptosis signals [8], promote tumor repopulation [20]. It has therefore
increased angiogenesis [9], invasion and metasta- lately been suggested that therapies targeting
sis [10], and evasion of host immune response genetically stable cells in the stromal compart-
[11, 12]. ment will be a promising alternative or an addi-
Neuroblastoma is a neural crest derived tional therapeutic strategy [21, 22]. This review
childhood malignancy of the sympathetic ner- will address PGE2-driven inflammation, its con-
vous system. Neuroblastoma accounts for tribution to neuroblastoma progression and inhi-
approximately 6–10% of the childhood cancer bition of PGE2 as a new therapeutic strategy in
cases and 9–15% of the pediatric cancer deaths neuroblastoma treatment.
making it the single most common and deadly
tumor of childhood [13, 14]. The disease ranges
from localized tumors with favorable biology 9.2 Prostaglandin E2
and high survival to metastatic tumors with Biosynthesis and Signaling
aggressive biology and poor prognosis.
Neuroblastoma is stratified into three risk- Arachidonic acid (AA), the initial precursor for
groups, low-risk, intermediate-risk and high- prostanoid (prostaglandins and thromboxane)
risk groups, depending on clinical age, biosynthesis, is an omega-6 fatty acid and
metastatic stage and histologic appearance with/ released from cellular membranes by phospholi-
without differentiation and biology including pases A2 (PLA2). PGE2 is synthesized from AA
genetic aberrations [15]. Two of the most com- in a two-step enzymatic reaction. In the first step
mon genetic aberration in the unfavorable high- AA is converted by cyclooxygenases (COX)-1
risk neuroblastoma is amplification of the and − 2 into a reactive intermediate, prostaglan-
MYCN oncogene (MYCN-amplified) and loss of din (PG)H2 via a transition molecule PGG. PGH2
the long arm of chromosome 11 (11q-deletion) is then converted into PGE2, PGD2, PGF2α, pros-
[16]. Children with high-risk tumors only have a tacyclin (PGI2) and thromboxane (TXA2) by spe-
survival rate of 40–50% despite intensive multi- cific terminal synthases i.e. PGE synthases, PGD
modal treatment including intense induction synthases, PGF synthase, PGI synthase and TX
chemotherapy, ablative therapy with stem cell synthase respectively. The key PGE2 producing
rescue, radiation therapy, immune therapy and synthase is microsomal prostaglandin E syn-
attempts to complete surgical removal [17]. The thase-1 (mPGES-1) [23]. During normal condi-
remarkable progression in pediatric oncology tions mPGES-1 is expressed at low levels but
with increasing survival of most childhood then rapidly induced upon stimuli with pro-
malignancies during the second half of the inflammatory cytokines (IL-1β, TNF-α) [24, 25],
twentieth century has stagnated in the last endotoxins (LPS) [26], growth factors (EGF)
decade highlighting the need for new therapeu- [27] and hypoxia [28]. There is two additional
tic targets and strategies to further increase sur- PGE2 synthases reported, namely cytosolic pros-
vival. Intensification of current treatments is not taglandin E synthase (cPGES) and microsomal
an option since life long side effects and second- prostaglandin E synthase-2 (mPGES-2), with no
ary malignancies already pose an increasing structural relationship to mPGES-1 [29, 30].
problem in surviving children [18]. While the function of mPGES-2 as a PGE2 syn-
Chemotherapy and radiation therapy hit the thase has been shown both for recombinantly
fast proliferating tumor cells that are genetically expressed enzyme [31] and in mammalian cells
unstable resulting in clonal evolution and the [29], the function of mPGES-2 as a PGE2 syn-
development of therapy resistance [19]. Stromal thase in physiological and pathological condi-
9 Targeting the COX/mPGES-1/PGE2 Pathway in Neuroblastoma 91
EP receptors
PLA2
Leukotrienes LOX
HETEs Arachidonic acid
Lipoxins
Cyt P450
COX-1 PGE
COX-2 PGE 2
synthase
EETs
TXA 2
Fig. 9.1 Prostaglandin E2 biosynthesis. Arachidonic acid converts PGH2 into PGE2. PGH2 can also be converted
(AA) released from cellular membranes is converted to into other prostanoids (PGD2, PGF2α, prostacyclin (PGI2)
prostaglandin (PG)E2 in a two-step enzymatic reaction. In or thromboxane (TXA2)) by respective terminal syn-
the first step AA is oxidized by cyclooxygenase (COX) -1 thases. PLA2, phospholipase A2; EP, E prostanoid;
or -2 into the short lived intermediate PGH2. In the second HETEs, hydroxyeicosatetraenoic acids; EETs, epoxye-
step microsomal prostaglandin E synthase-1 (mPGES-1) icosatrienoic acids; LOX, lipoxygenase; Cyt, Cytochrome
EP1-4
EP2 EP3
EP1 EP4
PLA2 β γ β γ β γ
Gαq Gαs Gα i
AA PGH 2 PGE 2
COX-1 mPGES-1
COX-2
PLC AC AC
mPGES-1
NSAIDs
inhibitor
Coxibs
Ca2+ cAMP cAMP
Fig. 9.2 PGE2 signaling. PGE2 formed via COXs and results in increased Ca2+ flux mediated through phospholi-
mPGES-1 is released from cells and confer downstream pase C (PLC), EP2 and EP4 leads to increased cAMP lev-
signaling via four E prostanoid (EP) receptors, EP1-EP4, els and EP3 in decrease levels of cAMP via activation or
either on the same cell (autocrine) or neighboring cells inhibition of adenylyl cyclase (AC) respectively. To
(paracrine). The EP receptors are G-protein coupled inhibit PGE2 signaling, either COXs or mPGES-1 can be
receptors that leads to changes in intracellular Ca2+ or inhibited or EP receptor antagonists can be used.
cyclic adenosine monophosphate (cAMP) levels. EP1 NSAIDs, nonsteroidal anti-inflammatory drugs
suppressor cells [42–46]. PGE2 also promotes the PGE2 produced by CAFs, TAMs and tumor cells
polarization of tumor-associated macrophages can stimulate vascular endothelial growth factor
(TAMs) towards a phenotype close to M2 polar- (VEGF) production in both CAFs and tumor
ized macrophages with surface expression of cells and thereby promote angiogenesis [50, 51].
CD163 and CD206 [47]. The M2-like TAMs in
turn contribute to tumor progression and immune
suppression by the production of cytokines and 9.4 Tumor-Promoting
chemokines e.g. IL-6 and IL-10 [48]. CAFs, one Inflammation
of the most abundant cell type within the tumor in Neuroblastoma
microenvironment, are recruited and activated by
tumor cells and influence several important pro- Lately, several investigations about the stromal
cesses in tumor progression including ECM compartment of neuroblastoma tumors have
remodeling, migration and metastasis, immune improved our knowledge about the role of non-
modulation and angiogenesis via secretion of a malignant cells in the tumor microenvironment to
plethora of soluble factors, including PGE2 [49]. neuroblastoma progression [21]. In a study by
9 Targeting the COX/mPGES-1/PGE2 Pathway in Neuroblastoma 93
toma xenografts, and that the reduction of tumor decreased presence of PDGFRβ expressing
growth was associated with a reduction of PGE2 CAFs, thus not only inhibiting mPGES-1 enzyme
in the tumor tissue [2]. activity in CAFs already infiltrating the tumor
but, consistent with the in vitro data, reducing fur-
ther recruitment of CAFs. These data together
9.5.3 Selective Inhibition provide strong evidence that mPGES-1/PGE2
of mPGES-1 in Experimental play an important role in the developing tumor
Neuroblastoma Models in vivo microenvironment in neuroblastoma creating an
immunosuppressive milieu and contributing to
In a recently published paper, we investigated the CAF infiltration and angiogenesis.
effect of selective mPGES-1 inhibition on neuro-
blastoma growth [58]. We used two experimental
neuroblastoma mouse models, a xenograft model 9.6 linical Benefit of PGE2
C
reflecting the 11q-deleted subset of high-risk neu- Blocking Therapy in Cancer
roblastoma and an inflammatory MYCN-driven Treatment
transgenic model [52, 64]. In both models, daily
intraperitoneal injections of the cross- species All anti-tumor treatments strive to induce apopto-
human/murine mPGES-1 inhibitor Compound III sis or cell death, either by directly targeting the
(CIII, 50 mg/kg) for 8–10 days, resulted in tumor cells or via the immune system. The cel-
reduced tumor growth. We also found interesting lular stress that follow massive cell death, induced
changes in the tumor microenvironment upon by cytotoxic treatment or irradiation of cells, also
pharmacological mPGES-1 inhibition with CIII, leads to induction and release of PGE2, stimulat-
highlighting the importance of PGE2 in several ing tumor repopulation [65–67]. Proposed mech-
aspects of the tumor microenvironment all of anisms how PGE2 support tumor cell survival and
which probably contributed to the resulting repopulation include changing the apoptotic
decrease in tumor growth. Firstly, tumors from threshold [68], suppression of inflammatory
CIII treated mice were less vascularized com- responses [69] and by promotion of cancer stem
pared to controls, assessed by immunohistochem- cells [70, 71]. Several studies have illuminated
ical (IHC) analysis with an antibody towards the the benefits of PGE2 inhibition in combination
endothelial marker CD31. Secondly, IHC analy- with chemotherapy. Recently, Kurtova et al.
sis of macrophage polarization markers revealed showed that the cytotoxic treatment-induced
less pro-tumorigenic M2 phenotype of the TAMs apoptosis and the accompanied PGE2 release pro-
(CD206) in tumors from CIII treated mice com- moted cancer stem cells repopulation and chemo-
pared to controls. A concomitant increase in anti- resistance. Inhibition of PGE2 with celecoxib
tumorigenic M1 phenotype of the TAMs, detected (5 mg/kg/day, starting 2 days before cytotoxic
with flow cytometry, confirmed a shift towards a treatment and then continuing throughout
less immunosuppressive tumor microenviron- the experiment) in combination with gem-
ment in CIII treated mice compared to controls. citabine/cisplatin (GC) administered in several
Thirdly, host-derived PGE2 producing CAFs treatment cycles reduced chemotherapy resis-
infiltrating the tumors were positive for PDGFRβ, tance compared to GC treatment alone in a blad-
EP4 and the IL-1 receptor type I as well as phos- der cancer xenograft model [20]. Also in
phorylated STAT3, describing a pro-inflammatory neuroblastoma in vivo studies, there are reports
pro-tumorigenic CAF phenotype. In an in vitro about beneficial combination effects with cele-
migration assay, IL-1β treatment induced migra- coxib and cytotoxic drugs. Celecoxib (10 mg/day
tion of fibroblasts towards tumor cells, a migra- for 12 days) was found to potentiate the anti-
tion that was inhibited either by CIII or an EP4 tumor effect of irinotecan and doxorubicin in
antagonist, suggesting a role of PGE2 in recruit- neuroblastoma (SH-SY5Y) xenografted rats
ing stromal cells to the tumor microenvironment. [72]. Additional studies confirmed an enhanced
Inhibition of mPGES-1 in vivo also resulted in anti-tumor effect of irinotecan when adminis-
96 K. Larsson et al.
tered in combination with low-dose celecoxib cardiovascular system. (ii) A three amino acid
(5 mg/kg/day for 20 consecutive days) in three phylogenetic difference between human
patient-derived neuroblastoma xenografts [73]. mPGES-1 and murine mPGES-1 in the catalytic
Celecoxib (250 mg/kg/day for 20 days) and a cleft renders most candidate drugs developed to
PGE2 neutralizing antibody was also found to human mPGES-1 inefficient towards murine
enhance the effect of radiation in a head and neck mPGES-1 and thus obstructing pre-clinical test-
squamous carcinoma xenograft model [74]. ing [81]. (iii) Lack of differentiation to NSAIDs.
Several recent studies also indicate that inhibition Researchers have failed to provide compelling
of PGE2 could be a strategy to enhance the clini- evidence of the potential benefits of mPGES-1
cal outcome of immune-based therapies [75–77]. inhibition compared to COX-inhibition.
Weather mPGES-1 inhibitors will have the same In contrast to COX-2 inhibitors, mPGES-1
beneficial combinatorial effects as celecoxib inhibitors would not affect the thromboxane/
remains to be proven. prostacyclin balance in favor of thromboxane
production and would thereby not have the same
effect on the cardiovascular system. Indeed, phar-
9.7 A Niche for mPGES-1 macological inhibition of mPGES-1 with CIII
Inhibition in Cancer [82], a cross-species human/murine mPGES-1
Treatment inhibitor, was even shown to increase production
of prostacyclin and reduce vasoconstriction of
Although NSAIDs and Coxibs have proved anti- blood vessels [83]. There are also studies show-
tumor efficiency in mouse models and epidemi- ing chemoprotective effects of prostacyclin and
ology data supports the benefits of regular PGD2, effects that would be lost upon COX inhi-
NSAID intake to prevent cancer, the severe side bition but would be spared or even enhanced by
effects on the gastrointestinal tract and cardiovas- mPGES-1 inhibition and the potential shunting to
cular system has hampered their use as chemo- prostacyclin and PGD2. For comprehensive
preventing and chemopotentiating agents. In reviews of pre-clinical mPGES-1 inhibitors see
children, there is also a fear of Reye’s syndrome references [84, 85]. For a summary of the advan-
that has been associated with viral induced tages and disadvantages with mPGES-1 inhibi-
fever and intake of aspirin, which is why the use tion versus COX inhibition see Table 9.2.
of aspirin in children below 16 years of age is
usually not recommended [78, 79]. In addition, Table 9.2 Advantages and disadvantages with mPGES-1
some cytostatic drugs like the anthracyclines are inhibition vs COX inhibition in cancer treatment
cardiotoxic and therefore may increase the risk of mPGES-1 inhibition
cardiovascular adverse effects seen with Coxibs + Anti-tumor efficiency in vivo.
in adults, also in children [80]. + Selective PGE2 inhibition.
An mPGES-1 inhibitor would potentially have + Cardiovascular safe.
the same anti-tumor properties seen with COX − No available inhibitor in the clinic.
inhibitors without the severe side effects on the − Unknown shunting effects.
gastrointestinal tract and the cardiovascular sys- − Poor efficiency of human inhibitors in mouse
models.
tem, resulting from the inhibition of all prosta-
COX inhibition
glandins. Still, there are no mPGES-1 inhibitors + Anti-tumor efficiency in vivo.
available in the clinic to date. There are three + Supportive epidemiologic data.
main reasons why the development of mPGES-1 − Inhibit chemoprotective prostacyclin and PGD2.
inhibitors has failed to reach the clinic. (i) ‘Guilt − Gastrointestinal side effects.
of association’ to COX-2 inhibitors. Since − Cardiovascular side effects.
mPGES-1 preferentially couples with COX-2 − Fear that aspirin causes Reye’s syndrome in
there are concerns that an mPGES-1 inhibitor children.
would have the same deleterious effect on the − Could increase cardiotoxicity of cytotoxic drugs.
9 Targeting the COX/mPGES-1/PGE2 Pathway in Neuroblastoma 97
significant difference was found in the risk of mation biology include prostaglandin E2 (PGE2),
serious vascular events between those who were a pro-inflammatory molecule stimulated by
assigned to n-3 PUFA supplementation and those COX, and 5-LOX-produced leukotrienes, which
who were assigned to a placebo [92]. It is thus contribute to potent inflammation in asthma and
imperative to conduct further research not only to other allergic diseases [96]. CYP regulates
confirm the effectiveness of n-3 PUFAs in psy- inflammation by oxidizing ARA with its active
chiatric disorders but also to explore molecular heme iron to form anti-inflammatory HETE or
mechanisms driving clinical benefits in patients epoxy-eicosatrienoic acid (EETs), which is then
and develop biomarkers to classify individuals hydrolyzed into pro-inflammatory dihydroxy-
with high likelihood of benefiting from PUFA eicosatrienoic acids (DHET) by soluble epoxide
supplementation. To achieve these goals, the log- hydrolase (sEH) [97].
ical next step is to take advantage of targeted We have demonstrated the effectiveness of a
metabolomics technology to focus on a specific combined use of lipidomics and targeted metabo-
class of metabolites, termed eicosanoids [93, 94]. lomics in investigating anorexia nervosa, an ill-
ness characterized by rapid weight loss and
reduction in food consumption [63]. Higher
10.7 Eicosanoids as Biomarkers ratios of dihydroxy to epoxy fatty acids were
for Psychiatric Disorders found in anorexia nervosa patients than in con-
trols, suggesting an upregulation of sEH activity,
While the pattern of association between PUFA an elevation in pro-inflammatory eicosanoid pro-
and psychiatric disorders may at first seem file, and a reduction in anti-inflammatory epoxy
straightforward, one must understand the func- fatty acids [63]. Additionally, recovered anorexia
tions and mechanisms underlying beneficial nervosa patients showed a partial normalization
effects of any compound to use such benefits in PUFA and eicosanoids, implying the resolu-
clinically. The bioactive lipids family represents tion of inflammation and that it may be achieved
the next logical class of molecules to study to by dietary intervention [26]. This is clinically rel-
elucidate PUFA mechanisms, and to establish a evant as well for patients with other types of psy-
biomarker system that is biologically and clini- chiatric disorders because medication
cally informative to guide precision psychiatry. non-adherence rate is notoriously high, up to
Major n-6 and n-3 PUFA can be oxygenated 80% in schizophrenia [98]. Dietary intervention
by at least 3 different enzymes to synthesize over may be an important alternative treatment modal-
120 heterogeneous and pleiotropic bioactive ity for patients refusing medications.
molecules termed eicosanoids [59, 95]. While the The results of the anorexia nervosa study sug-
word eicosanoid was derived from the Greek gest that psychopathology and inflammatory pro-
word “eikosa,” meaning “20,” based on the deriv- cesses in eating disorders are affected by
atives of the 20-carbon ARA, here “eicosanoid” interactions between dietary PUFA and geneti-
is applied to also include the oxygenated prod- cally driven metabolism. With additional empiri-
ucts of other PUFA including LA, ALA, DHA, cal research, food-based treatment or a
and DPA. The 3 enzymatic families that affect nutraceutical strategy may be employed to
PUFA are cyclooxygenases 1 and 2 (COX-1/2); improve outcomes in clinical psychiatry.
5-, 12-, and 15-lypooxygenases (5/12/15-LOX); Furthermore, as eicosanoid variation reflects in
and P450 epoxygenase. The COX-1/2 are known vivo cellular inflammation, targeted metabolo-
to drive the synthesis of prostanoids such as pros- mics can be applied to develop improved progno-
taglandins and thromboxanes, while 5/12/15- sis biomarkers.
LOX produce leukotrienes, lipoxins, and Untargeted metabolomics emerged as a use-
hydroxyeicosatetraenoids, and P450 synthesize ful tool to uncover unsuspected pathways
HETEs and epoxyeicosatrienoids [95]. The eico- involved in psychiatric disorders, and a targeted
sanoids most well-studied for their link to inflam- metabolomics approach is particularly helpful in
108 Pei-an (Betty) Shih
characterizing the specificity, direction and mag- The discovery of an association between cyto-
nitude of disease-associated metabolites, which chrome P450-associated bioactive lipid media-
provide molecular insight helpful to develop tors and psychiatric disorders is made possible in
new treatments. In a pilot study of adolescent part because of advances in technology, but the
major depressive disorder, we characterized involvement of eicosanoids in psychiatric disor-
eicosanoids in fasting plasma at the baseline ders was reported as early as the 1980s. Using
visit and final visit after a 2 year follow-up low throughput techniques such as radioimmuno-
period. While all subjects displayed no differ- assay, elevated levels of PGE and PGE2 were
ence in depression severity or profile of depres- identified in schizophrenia [104, 105], whereas
sion risk factors at the baseline visit, half of the PGD2, PGE2, and PGF2α and TXB2 were found
subjects had progressed to significantly worse to be elevated in major depressive disorder [106–
depression (refractory group) while the other 109]. Almost 40 years later, the field can now
half remitted. Strikingly, the eicosanoids profile take advantage of both untargeted and targeted
in the refractory group revealed a pattern very liquid chromatography-mass spectrometry-based
similar to that found in patients with anorexia methods to monitor a much larger number of
nervosa [99], implicating an epoxy fatty acid potential markers. A recent schizophrenia study
catalyzing enzyme, soluble epoxide hydrolase that investigated 158 markers including PUFA,
(sEH), as a common risk factor for depression eicosanoids, and related mediators from enzyme-
and anorexia nervosa.In a study of seasonal dependent or independent pathways uncovered
major depression [31], quantitative changes of 23 metabolites that were significantly altered in
CYP450 pathway eicosanoids during the winter patients compared with healthy controls [64].
season (when subjects experienced severe While some abnormal markers were reversed
depression symptoms) were similar in pattern to after antipsychotic treatment, anandamide,
the eicosanoids profile we found in the refrac- oleoylethanolamine, and ARA were identified as
tory adolescent depression group [99], suggest- having the best potential for differentiating
ing that sEH-mediated metabolism of PUFA patients from controls [64].
eicosanoids underlies the psychopathology of Leveraging what is already known about the
depressive disorders [31]. biology of bioactive lipids and the plethora of
sEH is known as a regulator of inflammatory physiological and homeostatic processes they
resolution due to its potent and complex mecha- participate in, several drugs have already been
nisms in the formation/catabolism of epoxy- and developed to inhibit the production of pro-
diol eicosanoids [100], but its involvement with inflammatory mediators, including nonsteroidal
psychiatric phenotypes was only recently uncov- anti-inflammatory drugs (NSAIDS) that reduce
ered through MS-based discovery [31, 99] and the activity of both COX-1 and COX-2 [110],
sequencing [101]. Another group has since dem- cysteinyl leukotriene (cysLT) receptor antago-
onstrated that sEH inhibition showed antidepres- nists that reduce bronchoconstriction caused by
sant effects in both inflammation and social cycLT and pro-inflammatory cytokines in the
defeat stress models of depression [102] and pulmonary system [111], and COX-2 inhibitors
attenuated behavioral abnormalities (i.e., hyper- [112]. In fact, administration of COX-2 inhibitor
locomotion and prepulse inhibition deficits) in an celecoxib has been shown to improve symptoms
animal model of schizophrenia [103]. Moreover, in schizophrenia [113], possibly through inhibit-
a higher level of sEH was found in postmortem ing conversion of ARA into prostanoids.
brain samples from patients with depression, Additionally, COX-2 inhibitors may be effective
schizophrenia, and bipolar disorder compared as an adjunctive treatment by accelerating the
with control samples [102], strengthening the onset of antidepressant effects for bipolar depres-
role sEH plays in psychiatric pathology. sion and refractory major depression [114, 115].
10 Metabolomics Biomarkers for Precision Psychiatry 109
11. Howland RH (2008) Sequenced treatment alterna- 27. Gowda GA, Djukovic D (2014) Overview of mass
tives to relieve depression (STAR∗D). Part 2: study spectrometry-based metabolomics: opportunities
outcomes. J Psychosoc Nurs Ment Health Serv and challenges. Methods Mol Biol 1198:3–12
46(10):21–24 28. Markley JL, Bruschweiler R, Edison AS, Eghbalnia
12. National research council committee on a frame- HR, Powers R, Raftery D et al (2017) The future of
work for developing a new taxonomy of disease NMR-based metabolomics. Curr Opin Biotechnol
(2011) National Academies Press, Washington, DC 43:34–40
13. Collins FS, Varmus H (2015) A new initiative on 29. Grebe SK, Singh RJ (2011) LC-MS/MS in the
precision medicine. N Engl J Med 372(9):793–795 clinical laboratory – where to from here? Clinical
14. Hasin Y, Seldin M, Lusis A (2017) Multi-omics Biochem Rev 32(1):5–31
approaches to disease. Genome Biol 18(1):83 30. Johnson CH, Ivanisevic J, Siuzdak G (2016)
15. Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Metabolomics: beyond biomarkers and towards
Byrne EM, Abdellaoui A et al (2018) Genome-wide mechanisms. Nat Rev Mol Cell Biol 17(7):451–459
association analyses identify 44 risk variants and 31. Hennebelle M, Otoki Y, Yang J, Hammock BD,
refine the genetic architecture of major depression. Levitt AJ, Taha AY et al (2017) Altered soluble
Nat Genet 50(5):668–681 epoxide hydrolase-derived oxylipins in patients with
16. Schizophrenia Working Group of the Psychiatric seasonal major depression: an exploratory study.
Genomics Consortium (2014) Biological insights Psychiatry Res 252:94–101
from 108 schizophrenia-associated genetic loci. 32. Tasic L, Pontes JGM, Carvalho MS, Cruz G, Dal
Nature 511(7510):421–427 Mas C, Sethi S et al (2017) Metabolomics and
17. Cross-Disorder Group of the Psychiatric Genomics lipidomics analyses by (1)H nuclear magnetic reso-
C, Lee SH, Ripke S, Neale BM, Faraone SV, Purcell nance of schizophrenia patient serum reveal poten-
SM et al (2013) Genetic relationship between five tial peripheral biomarkers for diagnosis. Schizophr
psychiatric disorders estimated from genome-wide Res 185:182–189
SNPs. Nat Genet 45(9):984–994 33. Meltzer HY, Rajagopal L, Huang M, Oyamada
18. Ward ET, Kostick KM, Lazaro-Munoz G (2019) Y, Kwon S, Horiguchi M (2013) Translating the
Integrating genomics into psychiatric practice: ethi- N-methyl-D-aspartate receptor antagonist model of
cal and legal challenges for clinicians. Harv Rev schizophrenia to treatments for cognitive impair-
Psychiatry 27(1):53–64 ment in schizophrenia. Int J Neuropsychopharmacol
19. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy 16(10):2181–2194
MI, Brown MA et al (2017) 10 years of GWAS 34. Yang J, Chen T, Sun L, Zhao Z, Qi X, Zhou K et al
discovery: biology, function, and translation. Am (2013) Potential metabolite markers of schizophre-
J Hum Genet 101(1):5–22 nia. Mol Psychiatry 18(1):67–78
20. Sun YV, Hu YJ (2016) Integrative analysis of multi- 35. Holmes E, Tsang TM, Huang JT, Leweke FM,
omics data for discovery and functional studies of Koethe D, Gerth CW et al (2006) Metabolic profiling
complex human diseases. Adv Genet 93:147–190 of CSF: evidence that early intervention may impact
21. Brainstorm C, Anttila V, Bulik-Sullivan B, Finucane on disease progression and outcome in schizophre-
HK, Walters RK, Bras J et al (2018) Analysis of nia. PLoS Med 3(8):e327
shared heritability in common disorders of the brain. 36. Jin H, Shih P-aB, Golshan S, Mudaliar S, Henry R,
Science 360(6395) Glorioso DK et al (2013) Comparison of longer-
22. Schwarz E, Guest PC, Rahmoune H, Harris LW, term safety and effectiveness of 4 atypical anti-
Wang L, Leweke FM et al (2012) Identification of a psychotics in patients over age 40: a trial using
biological signature for schizophrenia in serum. Mol equipoise-stratified randomization. J Clin Psychiatry
Psychiatry 17(5):494–502 74(1):11–19
23. Biomarkers Definitions Working G (2001) 37. McEvoy J, Baillie RA, Zhu H, Buckley P, Keshavan
Biomarkers and surrogate endpoints: preferred defi- MS, Nasrallah HA et al (2013) Lipidomics reveals
nitions and conceptual framework. Clin Pharmacol early metabolic changes in subjects with schizophre-
Ther 69(3):89–95 nia: effects of atypical antipsychotics. PLoS One
24. Dalvie S, Koen N, McGregor N, O’Connell K, 8(7):e68717
Warnich L, Ramesar R et al (2016) Toward a global 38. Suvitaival T, Mantere O, Kieseppa T, Mattila I, Poho
roadmap for precision medicine in psychiatry: chal- P, Hyotylainen T et al (2016) Serum metabolite pro-
lenges and opportunities. OMICS 20(10):557–564 file associates with the development of metabolic
25. Beger RD, Dunn W, Schmidt MA, Gross SS, Kirwan co-morbidities in first-episode psychosis. Transl
JA, Cascante M et al (2016) Metabolomics enables Psychiatry 6(11):e951
precision medicine: “a white paper, community per- 39. Xuan J, Pan G, Qiu Y, Yang L, Su M, Liu Y et al
spective”. Metabolomics 12(10):149 (2011) Metabolomic profiling to identify potential
26. Shih PB (2017) Integrating multi-omics biomarkers serum biomarkers for schizophrenia and risperidone
and postprandial metabolism to develop personal- action. J Proteome Res 10(12):5433–5443
ized treatment for anorexia nervosa. Prostaglandins 40. Zheng P, Fang Z, Xu XJ, Liu ML, Du X, Zhang X
Other Lipid Mediat 132:69–76 et al (2016) Metabolite signature for diagnosing
10 Metabolomics Biomarkers for Precision Psychiatry 111
major depressive disorder in peripheral blood mono- and recurrent mood disorders: phenomenology,
nuclear cells. J Affect Disord 195:75–81 mechanisms, and clinical application. Prog Lipid
41. Chen JJ, Zhou CJ, Zheng P, Cheng K, Wang HY, Li Res 66:1–13
J et al (2017) Differential urinary metabolites related 54. Grosso G, Pajak A, Marventano S, Castellano S,
with the severity of major depressive disorder. Behav Galvano F, Bucolo C et al (2014) Role of omega-3
Brain Res 332:280–287 fatty acids in the treatment of depressive disorders: a
42. Paige LA, Mitchell MW, Krishnan KR, Kaddurah- comprehensive meta-analysis of randomized clinical
Daouk R, Steffens DC (2007) A preliminary metab- trials. PLoS One 9(5):e96905
olomic analysis of older adults with and without 55. Young JJ, Bruno D, Pomara N (2014) A review of
depression. Int J Geriatr Psychiatry 22(5):418–423 the relationship between proinflammatory cyto-
43. Steffens DC, Wei J, Krishnan KR, Karoly kines and major depressive disorder. J Affect Disord
ED, Mitchell MW, O’Connor CM et al (2010) 169:15–20
Metabolomic differences in heart failure patients 56. Tanaka T, Matsuda T, Hayes LN, Yang S, Rodriguez
with and without major depression. J Geriatr K, Severance EG et al (2017) Infection and inflam-
Psychiatry Neurol 23(2):138–146 mation in schizophrenia and bipolar disorder.
44. Zhu H, Bogdanov MB, Boyle SH, Matson W, Sharma Neurosci Res 115:59–63
S, Matson S et al (2013) Pharmacometabolomics 57. Valdes AM, Ravipati S, Pousinis P, Menni C,
of response to sertraline and to placebo in major Mangino M, Abhishek A et al (2018) Omega-6
depressive disorder – possible role for methoxyin- oxylipins generated by soluble epoxide hydrolase
dole pathway. PLoS One 8(7):e68283 are associated with knee osteoarthritis. J Lipid Res
45. Gupta M, Neavin D, Liu D, Biernacka J, Hall-Flavin 59(9):1763–1770
D, Bobo WV et al (2016) TSPAN5, ERICH3 and 58. Serhan CN, Chiang N, Van Dyke TE (2008)
selective serotonin reuptake inhibitors in major Resolving inflammation: dual anti-inflammatory and
depressive disorder: pharmacometabolomics- pro-resolution lipid mediators. Nat Rev Immunol
informed pharmacogenomics. Mol Psychiatry 8(5):349–361
21(12):1717–1725 59. Spector AA, Kim HY (2015) Cytochrome
46. Czysz AH, South C, Gadad BS, Arning E, Soyombo P450 epoxygenase pathway of polyunsaturated
A, Bottiglieri T et al (2019) Can targeted metabolo- fatty acid metabolism. Biochim Biophys Acta
mics predict depression recovery? Results from the 1851(4):356–365
CO-MED trial. Transl Psychiatry 9(1):11 60. Imig JD (2015) Epoxyeicosatrienoic acids, hyperten-
47. Sethi S, Pedrini M, Rizzo LB, Zeni-Graiff M, Mas sion, and kidney injury. Hypertension 65(3):476–482
CD, Cassinelli AC et al (2017) (1)H-NMR, (1) 61. Li N, Liu JY, Timofeyev V, Qiu H, Hwang SH, Tuteja
H-NMR T2-edited, and 2D-NMR in bipolar disorder D et al (2009) Beneficial effects of soluble epoxide
metabolic profiling. Int J Bipolar Disord 5(1):23 hydrolase inhibitors in myocardial infarction model:
48. Chen JJ, Liu Z, Fan SH, Yang DY, Zheng P, Shao insight gained using metabolomic approaches. J Mol
WH et al (2014) Combined application of NMR- and Cell Cardiol 47(6):835–845
GC-MS-based metabonomics yields a superior uri- 62. Panigrahy D, Kaipainen A, Greene ER, Huang S
nary biomarker panel for bipolar disorder. Sci Rep (2010) Cytochrome P450-derived eicosanoids: the
4:5855 neglected pathway in cancer. Cancer Metastasis Rev
49. Ribeiro HC, Klassen A, Pedrini M, Carvalho MS, 29(4):723–735
Rizzo LB, Noto MN et al (2017) A preliminary study 63. Shih PB, Yang J, Morisseau C, German JB, Zeeland
of bipolar disorder type I by mass spectrometry-based AA, Armando AM et al (2016) Dysregulation of
serum lipidomics. Psychiatry Res 258:268–273 soluble epoxide hydrolase and lipidomic profiles in
50. Young SN (1991) The 1989 Borden Award Lecture. anorexia nervosa. Mol Psychiatry 21(4):537–546
Some effects of dietary components (amino acids, 64. Wang D, Sun X, Yan J, Ren B, Cao B, Lu Q et al
carbohydrate, folic acid) on brain serotonin synthe- (2018) Alterations of eicosanoids and related media-
sis, mood, and behavior. Can J Physiol Pharmacol tors in patients with schizophrenia. J Psychiatr Res
69(7):893–903 102:168–178
51. Conklin SM, Gianaros PJ, Brown SM, Yao JK, 65. Zulyniak MA, Perreault M, Gerling C, Spriet LL,
Hariri AR, Manuck SB et al (2007) Long-chain Mutch DM (2013) Fish oil supplementation alters
omega-3 fatty acid intake is associated positively circulating eicosanoid concentrations in young
with corticolimbic gray matter volume in healthy healthy men. Metabolism 62(8):1107–1113
adults. Neurosci Lett 421(3):209–212 66. Zivkovic AM, Telis N, German JB, Hammock BD
52. Pottala JV, Yaffe K, Robinson JG, Espeland MA, (2011) Dietary omega-3 fatty acids aid in the modu-
Wallace R, Harris WS (2014) Higher RBC EPA + lation of inflammation and metabolic health. Calif
DHA corresponds with larger total brain and hip- Agric (Berkeley) 65(3):106–111
pocampal volumes: WHIMS-MRI study. Neurology 67. Thomas J, Thomas CJ, Radcliffe J, Itsiopoulos C
82(5):435–442 (2015) Omega-3 fatty acids in early prevention of
53. Messamore E, Almeida DM, Jandacek RJ, inflammatory neurodegenerative disease: a focus on
McNamara RK (2017) Polyunsaturated fatty acids Alzheimer’s disease. Biomed Res Int 2015:172801
112 Pei-an (Betty) Shih
68. Sears B, Ricordi C (2012) Role of fatty acids and ticals for depression: a systematic review and meta-
polyphenols in inflammatory gene transcription and analyses. Am J Psychiatry 173(6):575–587
their impact on obesity, metabolic syndrome and dia- 83. Owen MJ, O’Donovan MC, Thapar A, Craddock N
betes. Eur Rev Med Pharmacol Sci 16(9):1137–1154 (2011) Neurodevelopmental hypothesis of schizo-
69. Lee HC, Yokomizo T (2018) Applications of mass phrenia. Br J Psychiatry 198(3):173–175
spectrometry-based targeted and non-targeted 84. Khan MM, Evans DR, Gunna V, Scheffer RE,
lipidomics. Biochem Biophys Res Commun Parikh VV, Mahadik SP (2002) Reduced erythrocyte
504(3):576–581 membrane essential fatty acids and increased lipid
70. Quehenberger O, Armando AM, Brown AH, Milne peroxides in schizophrenia at the never-medicated
SB, Myers DS, Merrill AH et al (2010) Lipidomics first-episode of psychosis and after years of treat-
reveals a remarkable diversity of lipids in human ment with antipsychotics. Schizophr Res 58(1):1–10
plasma. J Lipid Res 51(11):3299–3305 85. Assies J, Lieverse R, Vreken P, Wanders RJ,
71. Veselinovic M, Vasiljevic D, Vucic V, Arsic A, Dingemans PM, Linszen DH (2001) Significantly
Petrovic S, Tomic-Lucic A et al (2017) Clinical ben- reduced docosahexaenoic and docosapentaenoic
efits of n-3 PUFA and -linolenic acid in patients with acid concentrations in erythrocyte membranes
rheumatoid arthritis. Nutrients 9(4) from schizophrenic patients compared with a
72. Magee P, Pearson S, Allen J (2008) The omega-3 carefully matched control group. Biol Psychiatry
fatty acid, eicosapentaenoic acid (EPA), prevents the 49(6):510–522
damaging effects of tumour necrosis factor (TNF)- 86. Pawelczyk T, Trafalska E, Kotlicka-Antczak M,
alpha during murine skeletal muscle cell differentia- Pawelczyk A (2016) The association between poly-
tion. Lipids Health Dis 7:24 unsaturated fatty acid consumption and the tran-
73. Orr SK, Trepanier MO, Bazinet RP (2013) n-3 sition to psychosis in ultra-high risk individuals.
Polyunsaturated fatty acids in animal models with Prostaglandins Leukot Essent Fatty Acids 108:30–37
neuroinflammation. Prostaglandins Leukot Essent 87. Shih PB, Morisseau C, Le T, Woodside B, German
Fatty Acids 88(1):97–103 JB (2017) Personalized polyunsaturated fatty acids
74. Alam R, Abdolmaleky HM, Zhou JR (2017) as a potential adjunctive treatment for anorexia ner-
Microbiome, inflammation, epigenetic altera- vosa. Prostaglandins Other Lipid Mediat 133:11–19
tions, and mental diseases. Am J Med Genet B 88. Satogami K, Tseng PT, Su KP, Takahashi S, Ukai
Neuropsychiatr Genet 174(6):651–660 S, Li DJ et al (2019) Relationship between polyun-
75. Das UN (2013) Autism as a disorder of deficiency saturated fatty acid and eating disorders: systematic
of brain-derived neurotrophic factor and altered review and meta-analysis. Prostaglandins Leukot
metabolism of polyunsaturated fatty acids. Nutrition Essent Fatty Acids 142:11–19
29(10):1175–1185 89. Song C (2013) Essential fatty acids as potential anti-
76. Das UN (2013) Polyunsaturated fatty acids and inflammatory agents in the treatment of affective dis-
their metabolites in the pathobiology of schizophre- orders. Mod Trends Pharmacopsychiatry 28:75–89
nia. Prog Neuro-Psychopharmacol Biol Psychiatry 90. Moffett JR, Arun P, Ariyannur PS, Namboodiri AM
42:122–134 (2013) N-Acetylaspartate reductions in brain injury:
77. Chiu CC, Huang SY, Su KP, Lu ML, Huang impact on post-injury neuroenergetics, lipid synthe-
MC, Chen CC et al (2003) Polyunsaturated fatty sis, and protein acetylation. Front Neuroenerg 5:11
acid deficit in patients with bipolar mania. Eur 91. Manson JE, Cook NR, Lee IM, Christen W, Bassuk
Neuropsychopharmacol 13(2):99–103 SS, Mora S et al (2019) Marine n-3 fatty acids and
78. Pomponi M, Janiri L, La Torre G, Di Stasio E, Di prevention of cardiovascular disease and cancer. N
Nicola M, Mazza M et al (2013) Plasma levels of Eng J Med 380(1):23–32
n-3 fatty acids in bipolar patients: deficit restricted 92. Group ASC, Bowman L, Mafham M, Wallendszus
to DHA. J Psychiatr Res 47(3):337–342 K, Stevens W, Buck G et al (2018) Effects of n-3
79. Scola G, Versace A, Metherel AH, Monsalve-Castro fatty acid supplements in diabetes mellitus. N Engl
LA, Phillips ML, Bazinet RP et al (2018) Alterations J Med 379(16):1540–1550
in peripheral fatty acid composition in bipolar and 93. Yang J, Schmelzer K, Georgi K, Hammock BD
unipolar depression. J Affect Disord 233:86–91 (2009) Quantitative profiling method for oxylipin
80. Sublette ME, Hibbeln JR, Galfalvy H, Oquendo MA, metabolome by liquid chromatography electrospray
Mann JJ (2006) Omega-3 polyunsaturated essential ionization tandem mass spectrometry. Anal Chem
fatty acid status as a predictor of future suicide risk. 81(19):8085–8093
Am J Psychiatry 163(6):1100–1102 94. Astarita G, Kendall AC, Dennis EA, Nicolaou A
81. Ciappolino V, Delvecchio G, Agostoni C, Mazzocchi (2015) Targeted lipidomic strategies for oxygenated
A, Altamura AC, Brambilla P (2017) The role of n-3 metabolites of polyunsaturated fatty acids. Biochim
polyunsaturated fatty acids (n-3PUFAs) in affective Biophys Acta 1851(4):456–468
disorders. J Affect Disord 224:32–47 95. Chiurchiu V, Leuti A, Maccarrone M (2018)
82. Sarris J, Murphy J, Mischoulon D, Papakostas GI, Bioactive lipids and chronic inflammation: manag-
Fava M, Berk M et al (2016) Adjunctive nutraceu- ing the fire within. Front Immunol 9:38
10 Metabolomics Biomarkers for Precision Psychiatry 113
96. Lammermann T, Afonso PV, Angermann BR, 106. Nishino S, Ueno R, Ohishi K, Sakai T, Hayaishi
Wang JM, Kastenmuller W, Parent CA et al (2013) O (1989) Salivary prostaglandin concentrations:
Neutrophil swarms require LTB4 and integrins at possible state indicators for major depression. Am
sites of cell death in vivo. Nature 498(7454):371–375 J Psychiatry 146(3):365–368
97. Buczynski MW, Dumlao DS, Dennis EA (2009) 107. Ohishi K, Ueno R, Nishino S, Sakai T, Hayaishi O
Thematic review series: proteomics. An integrated (1988) Increased level of salivary prostaglandins
omics analysis of eicosanoid biology. J Lipid Res in patients with major depression. Biol Psychiatry
50(6):1015–1038 23(4):326–334
98. Latha KS (2010) The noncompliant patient in psy- 108. Calabrese JR, Skwerer RG, Barna B, Gulledge AD,
chiatry: the case for and against covert/surreptitious Valenzuela R, Butkus A et al (1986) Depression,
medication. Mens Sana Monogr 8(1):96–121 immunocompetence, and prostaglandins of the E
99. Shih PB, Yang J, Morisseau C, Calarge C (2017) series. Psychiatry Res 17(1):41–47
American College of Neuropsychopharmacology 109. Lieb J, Karmali R, Horrobin D (1983) Elevated levels
annual meeting. Palm Springs, CA of prostaglandin E2 and thromboxane B2 in depres-
100. Morisseau C, Hammock BD (2013) Impact of sion. Prostaglandins Leukot Med 10(4):361–367
soluble epoxide hydrolase and epoxyeicosanoids 110. Vane JR (2002) Biomedicine. Back to an aspirin a
on human health. Annu Rev Pharmacol Toxicol day? Science 296(5567):474–475
53:37–58 111. Aharony D (1998) Pharmacology of leukotriene
101. Zeeland AAS-V, Bloss CS, Tewhey R, Bansal V, receptor antagonists. Am J Respir Crit Care Med
Torkamani A, Libiger O et al (2014) Evidence for 157(6 Pt 2):S214–S218. discussion S8–9, S47–8
the role of EPHX2 gene variants in anorexia ner- 112. Muller N, Ulmschneider M, Scheppach C, Schwarz
vosa. Mol Psychiatry 19(6):724–732 MJ, Ackenheil M, Moller HJ et al (2004) COX-2
102. Ren Q, Ma M, Ishima T, Morisseau C, Yang J, inhibition as a treatment approach in schizophrenia:
Wagner KM et al (2016) Gene deficiency and phar- immunological considerations and clinical effects of
macological inhibition of soluble epoxide hydrolase celecoxib add-on therapy. Eur Arch Psychiatry Clin
confers resilience to repeated social defeat stress. Neurosci 254(1):14–22
Proc Natl Acad Sci U S A 113(13):E1944–E1952 113. Muller N, Krause D, Dehning S, Musil R, Schennach-
103. Ma M, Ren Q, Fujita Y, Ishima T, Zhang JC, Wolff R, Obermeier M et al (2010) Celecoxib treat-
Hashimoto K (2013) Effects of AS2586114, a solu- ment in an early stage of schizophrenia: results of
ble epoxide hydrolase inhibitor, on hyperlocomotion a randomized, double-blind, placebo-controlled trial
and prepulse inhibition deficits in mice after admin- of celecoxib augmentation of amisulpride treatment.
istration of phencyclidine. Pharmacol Biochem Schizophr Res 121(1–3):118–124
Behav 110:98–103 114. Muller N, Riedel M, Schwarz MJ (2004)
104. Mathe AA, Sedvall G, Wiesel FA, Nyback H (1980) Psychotropic effects of COX-2 inhibitors--a possible
Increased content of immunoreactive prostaglandin new approach for the treatment of psychiatric disor-
E in cerebrospinal fluid of patients with schizophre- ders. Pharmacopsychiatry 37(6):266–269
nia. Lancet 1(8158):16–18 115. Nery FG, Monkul ES, Hatch JP, Fonseca M,
105. Kaiya H, Uematsu M, Ofuji M, Nishida A, Takeuchi Zunta-Soares GB, Frey BN et al (2008) Celecoxib
K, Nozaki M et al (1989) Elevated plasma prosta- as an adjunct in the treatment of depressive or
glandin E2 levels in schizophrenia. J Neural Transm mixed episodes of bipolar disorder: a double-
77(1):39–46 blind, randomized, placebo-controlled study. Hum
Psychopharmacol 23(2):87–94
Cytochrome P450 Eicosanoid
Signaling Pathway in Colorectal
11
Tumorigenesis
Weicang Wang, Katherine Z. Sanidad,
and Guodong Zhang
Abstract Keywords
Colorectal cancer (CRC) is the third most Colorectal cancer · Colonic inflammation ·
common cancer and the second leading cause Cytochrome P450 · Soluble epoxide hydro-
of cancer-related death in the United States. It lase · Eicosanoids
is important to discover novel cellular targets
which are crucial in the pathogenesis of CRC,
which could facilitate development of
mechanism- based strategies to reduce the 11.1 Introduction
risks of CRC. Emerging studies support that
the cytochrome P450 (CYP) monooxygenase/ Colorectal cancer (CRC) is a serious health prob-
soluble epoxide hydrolase (sEH) pathway and lem: there were ~140,250 new cases and ~50,630
their eicosanoid metabolites play critical roles deaths in the United States in 2018 [1], empha-
in colonic inflammation and CRC, and could sizing the need for discovering novel cellular tar-
be therapeutically explored for treating or pre- gets which are crucial in the pathogenesis of
venting CRC. Here in this review, we discuss CRC. Colonic inflammation is a major risk factor
recent studies about the roles of the CYP/sEH for developing CRC, therefore, targeting the
eicosanoid pathway in the pathogenesis of pathological components involved in colonic
colonic inflammation and CRC. inflammation is a promising strategy to reduce
the risks of CRC [2]. Eicosanoids, which are
W. Wang endogenous lipid signaling molecules produced
Department of Food Science, University of from enzymatic metabolism of polyunsaturated
Massachusetts, Amherst, MA, USA fatty acids (PUFAs), play essential roles in
K. Z. Sanidad inflammatory responses and were recently impli-
Molecular and Cellular Biology Graduate Program, cated in cancer [3, 4]. The most prominent CRC-
University of Massachusetts,
Amherst, MA, USA
associated eicosanoids are prostaglandins, which
are produced by the cyclooxygenase-2 (COX-2)
G. Zhang (*)
Department of Food Science, University of
enzyme that is overexpressed in most human
Massachusetts, Amherst, MA, USA CRC samples [4]. Genetic knockout of Cox-2
Molecular and Cellular Biology Graduate Program,
reduces polyp formation in azoxymethane
University of Massachusetts, Amherst, MA, USA (AOM)- or Apc mutation-induced CRC models
e-mail: [email protected] [5, 6]. Furthermore, clinical and epidemiological
studies support that pharmacological inhibitors this previously unappreciated pathway in the
of COX-2, such as nonsteroidal anti-inflammatory pathogenesis of CRC could help to develop new
drugs (NSAIDs), are effective in reducing the strategies for cancer treatment or prevention. In
risk of CRC [4]. These results support the critical this review, we will discuss the roles of the CYP/
importance of eicosanoid signaling in sEH eicosanoid pathway in the pathogenesis of
CRC. However, the gastrointestinal and cardio- colonic inflammation and CRC, as well as
vascular toxicities induced by the COX-2 inhibi- obesity-associated colonic inflammation and
tors have limited their clinical applications [7]. CRC.
Besides COX-2, the roles of other eicosanoid
pathways in colonic inflammation and CRC are
not well understood [8]. It is therefore important 11.2 Expression of CYP
to discover novel eicosanoid signaling pathways Monooxygenases and sEH
involved in the pathogenesis of CRC. in Colonic Inflammation
Besides being substrates of COX-2, PUFAs and CRC
are also substrates of cytochrome P450 (CYP)
monooxygenases (predominately the CYP2C Our recent study has shown that CYP monooxy-
and CYP2J isoforms), which convert them to genases are overexpressed in colon tumor tissues
epoxygenated fatty acids (EpFAs). EpFAs include and colon cancer cells [21]. Compared with con-
epoxyeicosatrienoic acids (EETs) produced from trol healthy mice, the expressions of mouse CYP
arachidonic acid (ARA, 20:4ω-6), monooxygenases, such as Cyp2c38, Cyp2c39,
epoxyoctadecenoic acids (EpOMEs) from lin-
Cyp2c55, Cyp2c65, Cyp2c70, Cyp2j6, Cyp2j9,
oleic acid (LA, 18:2ω-6), epoxyoctadecadienoic and Cyp2j13, are increased in colon tissues of
acids (EpODEs) from α-linolenic acid (ALA, AOM/dextran sulfate sodium (DSS)-induced
18:3ω-3), and epoxydocosapentaenoic acids CRC mice [21]. In addition, the concentrations of
(EDPs) from docosahexaenoic acid (DHA, CYP-produced EpFAs are increased in both the
22:6ω-3) [9]. EpFAs are metabolically unstable plasma and colon tissues of AOM/DSS-induced
with a half-life of several seconds in vivo, in part CRC mice [21]. Furthermore, we find that com-
because they could be rapidly metabolized by pared with normal human colon cells (CCD-
soluble epoxide hydrolase (sEH) to generate the 18co), the expression of CYP2C8, CYP2C9,
corresponding fatty acid diols [10]. Currently, the CYP2C19, and CYP2J2 is increased in human
CYP/sEH eicosanoid pathway is being explored CRC cells (HCT116 and Caco-2) [21]. These
by academic laboratories and pharmaceutical results support that the CYP monooxygenase
companies for clinical applications. For example, pathway is upregulated in mouse and cell culture
GlaxoSmithKline is conducting human clinical models.
trials to test an sEH inhibitor GSK2256294; the The expressions of CYP monooxygenases in
company has found that this drug candidate is human CRC are more complicated. We analyzed
well-tolerated and causes sustained inhibition of gene expression of CYP monooxygenases
sEH in humans [11]. Other novel classes of sEH (CYP2C8, CYP2C9, CYP2C19, and CYP2J2) in
inhibitors are also being considered for human the Cancer Genome Atlas (TCGA) database, and
trials [12]. In addition, recent studies have shown found that their expressions were not increased in
that some FDA-approved drugs are potent inhibi- colorectal adenocarcinoma [21]. However, we
tors of CYP monooxygenases [13]. must note that the expression and activity of the
Emerging research supports that the CYP/sEH CYP enzymes are regulated by multiple mecha-
pathway plays critical roles in regulating inflam- nisms, including transcription, translation, and
mation, angiogenesis, tumor growth, and tumor post-translational modification, not only the
metastasis [14–17], and could be involved in the mRNA expression levels [22]. Indeed,
pathogenesis of colonic inflammation and CRC Enayetallah et al. report that CYP2C9 is detected
[18–21]. A better understanding of the roles of in 13 out of 17 human colon tumor samples,
11 Cytochrome P450 Eicosanoid Signaling Pathway in Colorectal Tumorigenesis 117
while it is not detected in matched benign sam- mice, pharmacological inhibition of sEH reduces
ples [23]. In addition to CRC, previous studies the infiltration of macrophages and expression of
have shown that CYP monooxygenases are over- pro-inflammatory cytokines in epididymal fat
expressed in other tumor tissues, such as breast, and liver [25, 26]. Deletion of sEH also leads to
liver, and stomach tumors [16, 23]. There could reduced infiltration of neutrophils, decreased lev-
be many mechanisms by which CYP monooxy- els of pro-inflammatory cytokines and less neu-
genases are overexpressed in tumor tissues. The ronal damage in an intracerebral hemorrhage
expression of CYP monooxygenases has been mouse model [27]. Together, these studies dem-
shown to be elevated by hypoxia [10], which is a onstrate sEH is involved in many inflammation-
common feature of tumor tissues [24]. It is fea- associated diseases.
sible that the hypoxic tumor microenvironment More studies are needed to better characterize
could contribute to the increased expression of the roles of sEH in cancer. Panigrahy et al. have
CYP monooxygenases in tumor tissues. shown that pharmacological inhibition or genetic
In humans, the expression of sEH is increased deletion of sEH increases tumor growth and
in colonic dysplasia and adenocarcinomas in metastasis in xenograft models by stimulating
ulcerative colitis (UC) patients [18]. sEH has tumor angiogenesis [15]. However, this finding is
been detected in ~40% of human colon adenocar- different from the results observed in the DSS
cinomas (7 out of 17 samples) with relatively and IL-10−/− mouse models [18–20]. The differ-
high expression levels, but is not detected in ent results could be, at least in part, due to the
matched benign samples (0 out of 4 samples) differences in mouse models. The phenotypes in
[23]. More studies are needed to characterize the the DSS and IL-10−/− mouse models are strongly
expression of sEH and the concentrations of associated with inflammation, therefore, inhibi-
CYP/sEH-produced eicosanoid metabolites in tion of sEH would reduce inflammatory response
colonic inflammation and CRC. and attenuate inflammation-associated CRC [18–
20]. In other models such as xenograft models,
inhibition of sEH could upregulate angiogenesis
11.3 R
oles of CYP/sEH Eicosanoid and increase tumorigenesis [15]. However, the
Pathway in Colonic pro-angiogenic effects of sEH inhibition were
Inflammation and CRC mainly observed in mouse models [15]. A recent
human clinical trial has shown that even at 100%
Recent research has shown that sEH plays a criti- inhibition of sEH, there is no change of the
cal role in colonic inflammation and CRC. In the plasma concentration of vascular endothelial
DSS-induced mouse model, sEH−/− mice have growth factor (VEGF), which is an important
reduced colonic inflammation (as assessed by biomarker of angiogenesis [11]. Since sEH inhib-
mucosal erosion and lymphoplasmocytosis) and itors are currently being evaluated in human clin-
carcinogenesis (tumor incidence and volume) ical trials [11, 12], it is of critical importance to
compared with wild-type mice [18]. Similarly, in better understand the roles of sEH and sEH inhib-
an interleukin 10 (IL-10) deficiency-induced itors in tumorigenesis.
CRC model, sEH−/− IL-10−/− mice have reduced Recent studies also support the critical roles of
colonic expression of pro-inflammatory cyto- CYP monooxygenases in tumorigenesis. In a
kines and formation of ulcers and carcinomas xenograft tumor model, overexpression of human
compared with sEH+/+ IL-10−/− mice [19, 20]. CYP2C8 or CYP2J2 in endothelial cells led to
Together, these results support that sEH could increased xenograft tumor growth of B16F10
contribute to colonic inflammation and melanoma and T241 fibrosarcoma [15]. Using a
inflammation-associated CRC. This is largely in Lewis lung carcinoma (LLC) resection-induced
agreement with previous studies which show that tumor metastasis model, endothelial expression
inhibition of sEH reduces inflammatory responses of human CYP2C8 or CYP2J2 increases lung
in various disease models. For example, in obese metastases [15]. Together, these results support
118 W. Wang et al.
the pro-tumorigenic and pro-metastatic effect of further metabolized by sEH to form the corre-
CYP monooxygenases. sponding fatty acid diols termed dihydroxyocta-
Our recent studies also support a potential role decenoic acids (DiHOMEs) [10]. Similar to
of CYP monooxygenases in the tumorigenesis of EpOMEs, DiHOMEs have also been shown to
CRC [21]. Compared with AOM/DSS-induced induce chemotaxis, tissue injury, and cause mor-
Cyp2c+/+ mice, the AOM/DSS-induced Cyp2c+/− tality in animal models [34, 35].
mice have lower tumor numbers and total tumor The biological actions of EETs are more
burden, as well as reduced expression of CYP complicated, as they have been shown to have
monooxygenases and concentrations of CYP- anti-inflammatory effects (negatively associated
derived fatty acid epoxides in colon tissues, sup- with tumorigenesis) and pro-angiogenic effects
porting the roles of Cyp2c monooxygenases in (positively associated with tumorigenesis).
colon tumorigenesis [21]. Consistent with the Indeed, many studies have shown that EETs
results observed in transgenic mouse models, we have potent anti-inflammatory effects. In a
also find that pharmacological inhibition of CYP murine carotid artery model, treatment with
monooxygenases suppresses AOM/DSS-induced 11,12-EET decreases tumor necrosis factor-α
colon tumorigenesis in mice [21]. Together, these (TNF-α)–induced mononuclear cell adhesion to
results support that CYP monooxygenases con- the arterial endothelium [36]. Treatment with
tribute to the tumorigenesis of CRC. 14,15-EET inhibits TNF-induced degradation
of IκBα in primary human lung tissue [37] and
reduces LPS-activated IL-1β and TNF-α expres-
11.4 Effects of CYP-Produced sion in mouse macrophages [18]. Together,
Eicosanoids on Inflammation these results support the anti-inflammatory
and CRC effects of EETs. The effects of EETs on tumor
inflammation are not well characterized and
The metabolism of PUFAs by CYP monooxy- require more studies. Previous studies have
genases leads to the formation of EpFAs [9]. The shown that inhibition or deletion of sEH attenu-
major EpFAs in tissues and plasma include ates DSS- or IL-10 knockout-induced CRC [18–
EpOMEs produced from LA, EETs from ARA, 20]; these results could be, at least in part, due
and EDPs from DHA. Emerging research sup- to the anti-inflammatory effects of EETs. On the
ports that these eicosanoid metabolites have other hand, EETs have pro-angiogenic effects,
potent effects on inflammation and tumorigenesis and therefore could promote tumor growth and
with the details discussed below. metastasis. In an orthotopic cancer model, treat-
Previous studies show that EpOMEs have a ment of 14,15-EET at a dose as low as 15 μg/kg/
series of detrimental actions such as inducing day increases orthotopic PC3M-LN4 prostate
chemotaxis, inflammation, cardiovascular dis- tumor growth in SCID mice [15]. In transgenic
eases, and pulmonary injury [28–33]. In human adenocarcinoma of the mouse prostate
studies, EpOMEs, which are termed “leukotox- (TRAMP) mice, administration of 14,15-EET at
ins,” are associated with multiple organ failures a dose of 15 μg/kg/day promotes prostate tumor
and adult respiratory distress syndrome in severe growth, suggesting that EETs increase tumor
burn patients [29, 31, 34]. Notably, at a concen- growth in vivo [15]. In addition, treatment with
tration as low as 10 nM, EpOMEs enhance neu- 14,15-EET increases tumor metastasis in a LLC
trophil chemotaxis, suggesting potent resection model [15]. More studies are needed
pro-inflammatory effects of EpOMEs [33]. to further characterize the role of EETs in
Regarding CRC, our recent study shows that tumorigenesis.
treatment with 12,13-EpOME exaggerates AOM/ Opposite to the effects of EpOMEs and EETs,
DSS-induced CRC in mice [21], supporting the our studies have shown that ω-3 PUFA-produced
pro-CRC actions of EpOMEs. EpOMEs can be EDPs inhibit angiogenesis, tumor growth, and
11 Cytochrome P450 Eicosanoid Signaling Pathway in Colorectal Tumorigenesis 119
tumor metastasis [38]. 19,20-EDP inhibits tube been shown to be biologically active and could
formation, migration, and production of matrix contribute to the biological actions of CYP
metalloproteinases (MMPs) in endothelial cells, monooxygenases and sEH. Recent studies have
and suppresses VEGF- and basic fibroblast shown that dihydroxyeicosatrienoic acids
growth factor (bFGF)-induced angiogenesis in a (DHETs), which are metabolites of EETs pro-
Matrigel plug assay in mice, demonstrating its duced by sEH, have pro-inflammatory effects
anti-angiogenic effect [38]. Treatment with stabi- [10, 47]. Treatment of 5,6-DHET, 8,9-DHET,
lized 19,20-EDP also suppresses primary tumor 11,12-DHET, or 14,15-DHET at a dose of 3 μM
growth and tumor metastasis in mice [38]. stimulates primary human monocyte migration
Furthermore, our recent study showed that in vitro [50]. Similar to the pro-inflammatory
treatment with EDPs suppresses growth of MC38 effects of ARA-produced DHETs, recent studies
xenograft colon tumors, supporting the anti-CRC show the DHA-produced dihydroxydocosapen-
effects of EDPs [39]. Consistent with our results, taenoic acids (DHDPs) promote progression of
a recent study by Yanai et al. has shown that sys- retinopathy in mice [48, 49]. Treatment of
tematic treatment with EDPs inhibits pathologi- 19,20-DHDP induces retinal angiogenesis by
cal angiogenesis in a mouse model of macular increasing tip cell, sprouting, and filopodia
degeneration [14]. numbers in mice, all of which contributes to
To date, the molecular mechanisms for the proliferative retinopathy [48]. Moreover, in an
biological actions of EpFAs are not well ex vivo whole mount retina model, treatment
understood. Many eicosanoids act by binding to with 19,20-DHDP increases vascular endothe-
specific cellular targets. For example, COX-2- lial cell permeability and pericyte migration
produced prostaglandin E2 (PGE2) and lipoxy- into extravascular space, both of which are dis-
genase (LOX)-produced leukotriene B4 (LTB4) ease markers for diabetic retinopathy [49]. In
act by binding to their G-protein coupled recep- cultured murine brain microvascular endothelial
tors [40]. The direct cellular target(s) of CYP cells, treatment of 19,20-DHDP decreases junc-
monooxygenase metabolites are not well under- tion formation between cells and reduces
stood, hampering our understanding of their expression of N-cadherin, which could contrib-
mechanisms of action. Previous studies show that ute to effect of 19,20-DHDP on vascular perme-
EETs bind to cell membrane-bound proteins in a ability during diabetic retinopathy [49]. Overall,
high-affinity, specific, and saturable manner [41– these studies demonstrate that fatty acid diols
43], and some of the biological actions of EETs are bioactive eicosanoids which could contrib-
are G protein-dependent [43–46]. These results ute to the pathogenesis of many diseases.
support that the CYP monooxygenase metabo- Together, these results support that CYP-
lites also act via interactions with specific cellu- produced eicosanoid metabolites, including
lar proteins; however, the identities of the cellular EpOMEs, EETs, and EDPs, have potent effects
targets are not well understood. Beyond EETs, no on tumorigenesis. These results could help to
study has investigated the cellular targets of other establish a novel mechanistic linkage between
CYP monooxygenase metabolites (such as fatty acid intake and cancer risks. For example,
EpOMEs). Identification of their direct cellular animal experiments have shown that a high
targets could greatly enhance our understanding dietary intake of LA increases AOM-induced
for the molecular mechanisms of the CYP mono- colon tumorigenesis, suggesting its potential
oxygenase pathway. In addition, the identified adverse effect on CRC [50–54]. Here our study
cellular targets could also serve as novel molecu- about the promoting effects of LA-produced
lar targets for preventing or treating cancer and EpOMEs on CRC suggests that the formation of
other human diseases. EpOMEs could contribute to the promoting
Besides the EpFAs, their down-stream effects of LA on the risks of CRC [21].
metabolites, termed fatty acid diols, have also
120 W. Wang et al.
11.5 R
oles of CYP/sEH Eicosanoid sues of obese mice [60, 61]. In addition, pharma-
Pathway in Obesity-Induced cological inhibition or genetic ablation of sEH
Colonic Inflammation attenuated various adverse consequences of obe-
sity, including endoplasmic reticulum stress,
More than one-third of US adults (34.9% or metabolic syndrome, fatty liver, hepatic steatosis,
78.6 million) are obese [55], and obese individu- inflammation, endothelial dysfunction, and dia-
als have a 30–60% higher risk of developing betes [25, 26, 60, 62–68]. Together, these results
CRC [56, 57]. Considering the obesity epidemic support that targeting sEH could be a promising
and the potential lethal consequence of CRC, strategy to reduce the risks of CRC in obese indi-
obesity-enhanced CRC is a serious health prob- viduals. Further studies are needed to character-
lem in the US. However, the mechanism by ize the roles of sEH in obesity-enhanced CRC.
which obesity increases the risks of CRC is not In our studies, we find that the colonic expres-
well understood, and there are few effective strat- sions of CYP2C and CYP2J monooxygenases
egies to prevent obesity-enhanced CRC [58]. are not altered in HFD-treated mice [59].
Using LC-MS/MS-based metabolomics, our However, considering that the fatty acid diols are
recent research suggests that sEH could be a down-stream metabolites of CYP monooxygen-
novel therapeutic target of obesity-induced ases, it is feasible that inhibition of CYP mono-
colonic inflammation [59]. In a high fat diet oxygenases could also reduce obesity-induced
(HFD)-induced obesity model in C57BL/6 mice, colonic inflammation and associated tumorigen-
we find that the expression of sEH and the con- esis. Targeting CYP monooxygenases could be
centrations of sEH-produced fatty acid diols are an alternative approach to reduce the risks of
significantly increased in the colon tissues of obesity-enhanced CRC. This approach could
HFD-induced obese mice [59]. Furthermore, minimize potential adverse effects of sEH inhibi-
pharmacological inhibition or genetic ablation of tion on angiogenesis [15], and further studies are
sEH abolishes HFD-induced colonic inflamma- needed to better characterize the roles of the
tion in mice, with reduced expression of pro- CYP/sEH eicosanoid pathway in obesity-
inflammatory cytokines (Il-1β and Tnf-α) and/or enhanced CRC.
decreased infiltration of immune cells in colon
tissues [59]. Furthermore, we find that the inhibi-
tion or ablation of sEH attenuates HFD-induced 11.6 Conclusion
activation of Wnt signaling pathway in colon tis-
sues [59]. CRC is the third most common cancer and the
Considering the critical roles of colonic second leading cause of cancer-related death in
inflammation and Wnt signaling in the pathogen- the US [1], emphasizing the need for discovery of
esis of CRC, these results support that sEH could novel cellular targets which are crucial in the
be a potential therapeutic target of obesity- pathogenesis of colon cancer. Recent research by
enhanced CRC. This notion is supported by pre- us and others support that CYP monooxygenases,
vious studies, which show that inhibition of sEH sEH, and associated eicosanoid metabolites
has beneficial effects on colonic inflammation, (EpFAs and fatty acid diols) play critical roles in
CRC, and obesity, supporting that targeting sEH regulating angiogenesis, tumor growth, and tumor
is a promising strategy to reduce the risks of metastasis [14–17], and could be involved in the
obesity-enhanced CRC. Previous studies have pathogenesis of colonic inflammation and CRC
shown that: [1] compared with normal colon tis- [18–21] (Fig. 11.1). Currently pharmaceutical
sues, the expression of sEH is increased in human companies and academic laboratories are t argeting
CRC samples [18, 23]; [2] pharmacological inhi- the CYP/sEH eicosanoid pathway to develop
bition or genetic ablation of sEH attenuates therapeutic drugs. Further understanding about
colonic inflammation and CRC [18–20]; and [3] the roles of the CYP/sEH eicosanoid pathway in
sEH is overexpressed in the liver and adipose tis- colonic inflammation and CRC could facilitate
11 Cytochrome P450 Eicosanoid Signaling Pathway in Colorectal Tumorigenesis 121
Fig. 11.1 Biochemistry
of the CYP/sEH
eicosanoid pathway. The PLA2
metabolism of PUFAs
by cytochrome P450 COOH
COOH COOH
monooxygenases
(largely CYP2C and
CYP2J isoforms) leads LA ARA DHA
to formation of
epoxygenated fatty acids Cytochrome P450
(EpFAs), which are
further metabolized by COOH
COOH COOH
soluble epoxide
hydrolase (sEH) to form
O O O
the corresponding fatty
acid diols. Recent EpOMEs EETs EDPs
research shows that the
CYP and sEH enzymes
Soluble epoxide hydrolase
contribute to the
pathogenesis of colonic
inflammation and CRC COOH COOH COOH
HO OH HO OH
HO OH
DiHOMEs DHETs DiHDPEs
13. Veith H et al (2009) Comprehensive characterization circulatory shock and disseminated intravascular
of cytochrome P450 isozyme selectivity across chem- coagulation. Jpn J Med 30:224–228
ical libraries. Nat Biotechnol 27:1050 29. Hayakawa M et al (1990) Proposal of leukotoxin,
14. Yanai R et al (2014) Cytochrome P450-generated
9,10-epoxy-12-octadecenoate, as a burn toxin.
metabolites derived from omega-3 fatty acids attenu- Biochem Int 21:573–579
ate neovascularization. Proc Natl Acad Sci U S A 30. Hu JN et al (1988) Neutrophil-derived epoxide,
111:9603–9608 9,10-epoxy-12-octadecenoate, induces pulmonary
15. Panigrahy D et al (2012) Epoxyeicosanoids stimulate edema. Lung 166:327–337
multiorgan metastasis and tumor dormancy escape in 31. Kosaka K, Suzuki K, Hayakawa M, Sugiyama S,
mice. J Clin Invest 122:178–191 Ozawa T (1994) Leukotoxin, a linoleate epoxide: its
16. Jiang JG et al (2005) Cytochrome P450 2J2 pro-
implication in the late death of patients with extensive
motes the neoplastic phenotype of carcinoma cells burns. Mol Cell Biochem 139:141–148
and is up-regulated in human tumors. Cancer Res 32. Ozawa T et al (1988) Cytotoxic activity of leukotoxin,
65:4707–4715 a neutrophil-derived fatty acid epoxide, on cultured
17. Pozzi A et al (2010) The anti-tumorigenic properties human cells. Biochem Int 16:369–373
of peroxisomal proliferator-activated receptor alpha 33. Totani Y et al (2000) Leukotoxin and its diol induce
are arachidonic acid epoxygenase-mediated. J Biol neutrophil chemotaxis through signal transduction
Chem 285:12840–12850 different from that of fMLP. Eur Respir J 15:75–79
18. Zhang W et al (2013) Soluble epoxide hydrolase
34. Zheng J, Plopper CG, Lakritz J, Storms DH, Hammock
deficiency inhibits dextran sulfate sodium-induced BD (2001) Leukotoxin-diol: a putative toxic mediator
colitis and carcinogenesis in mice. Anticancer Res involved in acute respiratory distress syndrome. Am
33:5261–5271 J Respir Cell Mol Biol 25:434–438
19. Zhang W et al (2012) Soluble epoxide hydrolase
35. Moghaddam MF et al (1997) Bioactivation of leuko-
gene deficiency or inhibition attenuates chronic active toxins to their toxic diols by epoxide hydrolase. Nat
inflammatory bowel disease in IL-10(−/−) mice. Dig Med 3:562–566
Dis Sci 57:2580–2591 36. Node K et al (1999) Anti-inflammatory properties of
20. Zhang W et al (2013) Reduction of inflammatory cytochrome P450 epoxygenase-derived eicosanoids.
bowel disease-induced tumor development in IL-10 Science 285:1276–1279
knockout mice with soluble epoxide hydrolase gene 37. Morin C, Sirois M, Echave V, Gomes MM, Rousseau
deficiency. Mol Carcinog 52:726–738 E (2008) EET displays anti-inflammatory effects in
21. Wang W et al (2019) Targeted metabolomics identi- TNF-alpha stimulated human bronchi: putative role of
fies cytochrome P450 monooxygenase eicosanoid CPI-17. Am J Respir Cell Mol Biol 38:192–201
pathway as novel therapeutic target of colon tumori- 38. Zhang G et al (2013) Epoxy metabolites of docosa-
genesis. Cancer Res 79(8):1822–1830 hexaenoic acid (DHA) inhibit angiogenesis, tumor
22. Aguiar M, Masse R, Gibbs BF (2005) Regulation of growth, and metastasis. Proc Natl Acad Sci U S A
cytochrome P450 by posttranslational modification. 110:6530–6535
Drug Metab Rev 37:379–404 39. Wang W et al (2017) Omega-3 polyunsaturated fatty
23. Enayetallah AE, French RA, Grant DF (2006)
acids and their cytochrome P450-derived metabo-
Distribution of soluble epoxide hydrolase, cyto- lites suppress colorectal tumor development in mice.
chrome P450 2C8, 2C9 and 2J2 in human malignant J Nutr Biochem 48:29–35
neoplasms. J Mol Histol 37:133–141 40.
Funk CD (2001) Prostaglandins and leukotri-
24. Vaupel P, Mayer A (2007) Hypoxia in cancer: sig- enes: advances in eicosanoid biology. Science
nificance and impact on clinical outcome. Cancer 294:1871–1875
Metastasis Rev 26:225–239 41. Yang W et al (2008) Characterization of epoxye-
25. Liu Y et al (2012) Inhibition of soluble epoxide
icosatrienoic acid binding site in U937 membranes
hydrolase attenuates high-fat-diet-induced hepatic using a novel radiolabeled agonist, 20-125i-14,15-
steatosis by reduced systemic inflammatory status in epoxyeicosa-8(Z)-enoic acid. J Pharmacol Exp Ther
mice. PLoS One 7:e39165 324:1019–1027
26. Lopez-Vicario C et al (2015) Inhibition of soluble 42. Chen Y, Falck JR, Manthati VL, Jat JL, Campbell
epoxide hydrolase modulates inflammation and WB (2011) 20-Iodo-14,15-epoxyeicosa-8(Z)-enoyl-
autophagy in obese adipose tissue and liver: role 3-azidophenylsulfonamide: photoaffinity label-
for omega-3 epoxides. Proc Natl Acad Sci U S A ing of a 14,15-epoxyeicosatrienoic acid receptor.
112:536–541 Biochemistry 50:3840–3848
27. Wu CH et al (2017) Genetic deletion or pharmacolog- 43. Park SK et al (2018) GPR40 is a low-affinity epoxye-
ical inhibition of soluble epoxide hydrolase reduces icosatrienoic acid receptor in vascular cells. J Biol
brain damage and attenuates neuroinflammation Chem 293:10675–10691
after intracerebral hemorrhage. J Neuroinflammation 44. Liu X et al (2017) Functional screening for G protein-
14:230 coupled receptor targets of 14,15-epoxyeicosatrienoic
28. Hanaki Y et al (1991) Leukotoxin, 9, 10-epoxy-
acid. Prostaglandins Other Lipid Mediat 132:31–40
12-octadecenoate: a possible responsible factor in
11 Cytochrome P450 Eicosanoid Signaling Pathway in Colorectal Tumorigenesis 123
45. Li P et al (2015) Epoxyeicosatrienoic acids enhance 57. Ma Y et al (2013) Obesity and risk of colorectal can-
embryonic haematopoiesis and adult marrow engraft- cer: a systematic review of prospective studies. PLoS
ment. Nature 523:468–471 One 8:e53916
46. Ding Y et al (2014) The biological actions of
58. Roberts DL, Dive C, Renehan AG (2010) Biological
11,12-epoxyeicosatrienoic acid in endothelial cells mechanisms linking obesity and cancer risk: new per-
are specific to the R/S-enantiomer and require the spectives. Annu Rev Med 61:301–316
G(s) protein. J Pharmacol Exp Ther 350:14–21 59. Wang W et al (2018) Lipidomic profiling reveals
47. Kundu S et al (2013) Metabolic products of soluble soluble epoxide hydrolase as a therapeutic target of
epoxide hydrolase are essential for monocyte che- obesity-induced colonic inflammation. Proc Natl
motaxis to MCP-1 in vitro and in vivo. J Lipid Res Acad Sci U S A 115:5283–5288
54:436–447 60. Bettaieb A et al (2013) Soluble epoxide hydrolase
48. Hu J et al (2014) Muller glia cells regulate notch sig- deficiency or inhibition attenuates diet-induced endo-
naling and retinal angiogenesis via the generation of plasmic reticulum stress in liver and adipose tissue.
19,20-dihydroxydocosapentaenoic acid. J Exp Med J Biol Chem 288:14189–14199
211:281–295 61. De Taeye BM et al (2010) Expression and regula-
49. Hu J et al (2017) Inhibition of soluble epoxide
tion of soluble epoxide hydrolase in adipose tissue.
hydrolase prevents diabetic retinopathy. Nature Obesity (Silver Spring) 18:489–498
552:248–252 62. do Carmo JM et al (2012) Inhibition of soluble epox-
50. Enos RT et al (2016) High-fat diets rich in saturated fat ide hydrolase reduces food intake and increases meta-
protect against azoxymethane/dextran sulfate sodium- bolic rate in obese mice. Nutr Metab Cardiovasc Dis
induced colon cancer. Am J Physiol Gastrointest Liver 22:598–604
Physiol 310:G906–G919 63. Imig JD et al (2012) Soluble epoxide hydrolase inhi-
51. Wu B et al (2004) Dietary corn oil promotes colon bition and peroxisome proliferator activated recep-
cancer by inhibiting mitochondria-dependent apop- tor gamma agonist improve vascular function and
tosis in azoxymethane-treated rats. Exp Biol Med decrease renal injury in hypertensive obese rats. Exp
(Maywood) 229:1017–1025 Biol Med (Maywood) 237:1402–1412
52. Fujise T et al (2007) Long-term feeding of various 64. Iyer A et al (2012) Pharmacological inhibition of
fat diets modulates azoxymethane-induced colon soluble epoxide hydrolase ameliorates diet-induced
carcinogenesis through Wnt/beta-catenin signaling metabolic syndrome in rats. Exp Diabetes Res
in rats. Am J Physiol Gastrointest Liver Physiol 2012:758614
292:G1150–G1156 65. Roche C et al (2015) Soluble epoxide hydrolase inhi-
53. Reddy BS, Tanaka T, Simi B (1985) Effect of different bition improves coronary endothelial function and
levels of dietary trans fat or corn oil on azoxymethane- prevents the development of cardiac alterations in
induced colon carcinogenesis in F344 rats. J Natl obese insulin-resistant mice. Am J Physiol Heart Circ
Cancer Inst 75:791–798 Physiol 308:H1020–H1029
54. Pot GK et al (2008) Opposing associations of serum 66. Zha W et al (2014) Functional characterization of
n-3 and n-6 polyunsaturated fatty acids with colorec- cytochrome P450-derived epoxyeicosatrienoic acids
tal adenoma risk: an endoscopy-based case-control in adipogenesis and obesity. J Lipid Res 55:2124–2136
study. Int J Cancer 123:1974–1977 67. Zhang LN et al (2011) Inhibition of soluble epoxide
55. Ogden CL, Carroll MD, Fryar CD, Flegal KM (2015) hydrolase attenuates endothelial dysfunction in ani-
Prevalence of obesity among adults and youth: United mal models of diabetes, obesity and hypertension. Eur
States, 2011–2014. NCHS Data Brief:1–8 J Pharmacol 654:68–74
56. Moghaddam AA, Woodward M, Huxley R (2007) 68. Luria A et al (2011) Soluble epoxide hydrolase defi-
Obesity and risk of colorectal cancer: a meta-analysis ciency alters pancreatic islet size and improves glu-
of 31 studies with 70,000 events. Cancer Epidemiol cose homeostasis in a model of insulin resistance.
Biomark Prev 16:2533–2547 Proc Natl Acad Sci U S A 108:9038–9043
Contributions
of 12/15-Lipoxygenase to Bleeding 12
in the Brain Following Ischemic
Stroke
ischemic strokes both in the presence and absence infarct sizes (Sen group 90% reduction 72 h after
of tPA, as will be discussed in this mini-review. stroke onset, van Leyen group 40% infarct size
The liberation of polyunsaturated fatty acids reduction measured 24 h after initiation of exper-
including arachidonic acid in the brain following imental stroke). Moreover, subsequent studies
a stroke has been recognized since the early from our lab found that the mRNA encoding
1970s [1]. These give rise to a dizzying spectrum 12/15-LOX was up-regulated 2.2 fold in mice
of eicosanoids and related compounds produced 24 h after an experimental stroke [7].
by lipoxygenases, cyclooxygenases, and cyto- Immunohistochemistry showed the increased
chromes P450, including prostaglandins, leukot- 12/15-LOX signal both in neurons, and in endo-
rienes, and hydroxyeicosatetraenoic acids thelial cells [8]. Early work focused on injury to
(HETEs). Increased levels of 12-HETE were neurons, initially with the discovery that 12/15-
found along with leukotrienes C4 and D4 in ger- LOX contributes to a form of cell death termed
bil brains following an experimental stroke [2]. oxidative glutamate toxicity or oxytosis in neuro-
We have similarly seen massively increased lev- nal cells [9]. More recently, a related redox path-
els of 12-HETE specifically on the infarcted side way termed ferroptosis was introduced, which is
of the brain in mice both 12 and 24 h after onset characterized by the loss of glutathione peroxi-
of ischemia (Fig. 12.1). Therapeutically, initially dase 4 (Gpx-4) activity [10] and in which 12/15-
5-LOX was seen as the most promising target, LOX is also involved. The commonalities and
mostly due to a much better understanding of differences between these two pathways have yet
leukotriene biology compared to the much less to be clearly defined [10–12]. Glutathione as the
studied 12/15-LOX. However, two independent major intracellular antioxidant in neurons is
findings changed this perception: In 2004, a clearly important for both pathways, and gluta-
Japanese study found that Alox5 knockout mice thione levels also drop on the ischemic side of
developed the same level of ischemic injury fol- the brain following stroke, which presumably
lowing experimental stroke as matched wild type contributes to the activation of 12/15-LOX. The
mice [3]. Around the same time, the group of function of 12/15-LOX in this neuronal cell
Chandan Sen at Ohio State University [4] and death pathway is to damage mitochondria and
our group [5, 6] found that Alox15 knockout other organelles, for which the 12/15-LOX is
mice were protected against the consequences of uniquely qualified: in stress reticulocytes pro-
an experimental stroke, developing smaller duced during severe anemia, as well as when
Fig. 12.1 (a) 12-HETE was significantly increased in the identity of 12-HETE was confirmed by high-performance
infarcted ipsilateral hemisphere of mice both 12 and liquid chromatography (HPLC)/mass spectrometry analy-
24 hours after transient focal ischemia, compared to sis. The smaller peak for 15-HETE in the HPLC profile
sham-operated mice (∗p < 0.05, ∗∗∗p < 0.001; sham, n = 6 (top panel) is also a 12/15-LOX product. (Reprinted with
brains; 12 h, n = 3 brains; 24 h, n = 5 brains). (b) The permission from reference Rai et al. [24])
12 Contributions of 12/15-Lipoxygenase to Bleeding in the Brain Following Ischemic Stroke 127
incubated in vitro, the enzyme attacks mitochon- anticoagulant warfarin [22, 23]. Warfarin is a
drial membranes [13–16], priming the mitochon- vitamin K inhibitor that is frequently given to
dria for further degradation via the ubiquitin/ patients with atrial fibrillation to reduce their risk
proteasome system. This is one of three path- of blood clot formation and subsequent stroke.
ways by which reticulocytes lose their organelles While warfarin reduces the risk of stroke in these
during maturation to become functioning eryth- patients, this anticoagulant can cause excessive
rocytes, the others being degradation via autoph- bleeding leading to increased injury when a
agic vacuoles and exosome formation [17, 18]. stroke does occur. Moreover, thrombolysis with
The redundancy of these pathways may be the the clotbuster tissue plasminogen activator (tPA)
reason why no outright defects in erythropoiesis is contraindicated in effectively anticoagulated
were found in Alox15 knockout mice [19]. patients on warfarin (international normalized
Beyond causing cell death in neurons, how- ratio of coagulation time (INR) > 1.7) because
ever, it has in recent years become clear that tPA itself has bleeding as a side effect, thus elimi-
12/15-LOX also contributes to vessel injury in nating the only drug currently approved by the
rodent models of stroke. Alox15 knockout mice FDA as a treatment option. Mice pretreated with
develop 51% less edema following experimental warfarin via their drinking water for 24 h prior to
stroke than their wild type counterparts, and 30% experimental stroke develop severe hemorrhage
less immunoglobulin G (representative of blood both when the mice receive a tPA infusion fol-
proteins) extravasates into the brain parenchyma lowing removal of the filament, or in the absence
[8]. Several years later, we made the striking of tPA when the stroke is severe enough (3 h of
observation in a mouse model of thrombotic filament occlusion; Fig. 12.2) [23]. Along with
stroke that tPA infusion intended to lyse the the increased hemorrhage, we also found 25%
occluding thrombus led to massive brain hemor- higher levels of 12/15-LOX in the brains of the
rhage in these mice, which was reduced by 82% warfarin-treated mice. The increase was seen
through simultaneous administration of a 12/15- mostly in the vasculature, consistent with the
LOX inhibitor, LOXBlock-1 [20] (Fig. 12.2). We idea that the increased vessel leakage following
have expanded on these results by investigating warfarin pretreatment is due to 12/15-LOX.
the effects of 12/15-LOX inhibition on both Consistent with the idea of 12/15-LOX as con-
bleeding and infarct size in this thrombosis tributor to hemorrhage, 41% less bleeding was
model, and found that LOXBlock-1 also seen following warfarin pretreatment in Alox15
improved behavioral scores in the mice [21]. knockout mice [23]. In wild type mice, a simi-
This finding led us to systematically study larly drastic reduction in hemorrhage by 38%
mouse models of stroke where reperfusion fol- was detected when the mice were treated with the
lowing the ischemic event is associated with second generation 12/15-LOX inhibitor ML351
increased bleeding. In the classical filament [24], administered intraperitoneally at the time of
model of transient focal ischemia, a filament is reperfusion, 3 hours after onset of ischemia. The
inserted into the internal carotid artery to par- reduction in bleeding remained significant even
tially block the middle cerebral artery on one side when the results were adjusted to account for the
of the brain, which leads to a reduction of blood reduced infarct size in the ML351-treated mice,
flow and ischemia in the striatum and cortex. The confirming that there is a specific effect on hem-
filament is then removed after a pre-specified orrhage. When ML351 was administered along
time to allow for reperfusion. When the mouse is with tPA in warfarin pretreated mice, hemorrhage
sacrificed after 24 h an infarct is detected, the size was similarly reduced by 59%. Taken together,
of which is determined by the duration of the these results demonstrated that increased 12/15-
ischemia. Typically, this model does not lead to LOX in the brain vasculature can contribute to
significant bleeding, but so called hemorrhagic excessive bleeding in the brain, which is reduced
transformation of the ischemic stroke can be by treatment with a 12/15-LOX inhibitor.
induced, for example when mice are fed with the
128 Y. Zheng et al.
Fig. 12.2 Examples of stroke models associated with 24 h with the anticoagulant warfarin added to the drinking
increased hemorrhage. (a) Thrombosis was induced when water causes massive hemorrhage following a severe form
10% ferric chloride solution was topically applied to the of experimental ischemic stroke with 3 h of occlusion of
brain. An infusion of tissue plasminogen activator (tPA) the middle cerebral artery. This is again visible both on the
2 h later led to distinct hemorrhage in vehicle-treated mice surface, as well as in brain sections. Mice treated with the
after 24 h, visible both on the surface of the brain (top) 12/15-LOX inhibitor ML351 (50 mg/kg) develop far less
and in sections (below). In contrast, when mice were hemorrhage (bottom right). Quantitation graphs represent
intraperitoneally injected with the 12/15-LOX inhibitor hemorrhage area measured in brain sections and are
LOXBlock-1 (50 mg/kg), significantly less hemorrhage reprinted with permission from references [24] (top) and
was detected in the brain. (b) Pretreatment of mice for [23] (bottom)
Much work remains to be done to elucidate STAT6 are involved in up-regulating 12/15-LOX
the mechanism by which 12/15-LOX contributes under ischemic conditions [7]. Are the same
to increased hemorrhage after an ischemic stroke. STATs active here, or is a different form of regu-
Important open questions include the selective lation relevant? Finally, what happens after
up-regulation of 12/15-LOX in brain vascular 12/15-LOX is up-regulated in the endothelial
endothelial cells following warfarin administra- cells and how does this lead to vessel rupture? In
tion, both with and without subsequent tPA infu- addition to destroying endothelial cells of the
sion. Is this a direct effect of warfarin and/or tPA, brain vasculature by damaging mitochondria,
or are intermediate factors involved? Also, in there may be a second injury mechanism induced
neurons signal transducers and activators of tran- by the signaling function of 12/15-LOX via
scription (STATs), specifically STAT1 and metabolites of arachidonic acid. Both 12-HETE
12 Contributions of 12/15-Lipoxygenase to Bleeding in the Brain Following Ischemic Stroke 129
and its immediate precursor 12-HPETE are we found increased 12/15-LOX in the brains of
known as second messengers [25], activated mice 24 h after hemorrhage induction (92 ± 60
along the semaphorin pathway [26, 27]. In neu- 12/15-LOX positive cells/field vs. 2 ± 2 in sham-
rons, this can lead to axon retraction [28, 29], but operated controls, p < 0.05) [38]. In this case,
under some conditions also to cell death [30]. 12/15-LOX expression was detected mostly in
Semaphorin 3A has also been reported to increase macrophages however, rather than in neurons and
vascular permeability in experimental stroke endothelial cells; the injury mechanism may thus
models [31]. Downstream of 12-HETE and differ from that in ischemic stroke. Regardless,
12-HPETE, secretion of destabilizing matrix Alox15 knockout mice developed 72% less
metalloproteinases (MMPs) may play a role [32]. injury than wild type mice, and 12/15-LOX inhi-
Both the molecular details of this signaling path- bition also reduced injury by 55% compared to
way, and the relative contributions of both path- vehicle-treated mice in this model of hemor-
ways to vascular injury require further study. rhagic stroke.
Because 12/15-LOX contributes to both neu- In conclusion, despite different triggers – in
ronal cell death and to vessel leakage following a the presence or absence of anticoagulant, with or
stroke, 12/15-LOX inhibition appears to be a par- without tPA treatment – 12/15-LOX is activated
ticularly promising approach to stroke therapy by in various models of stroke-related hemorrhage.
targeting two separate modes of injury, killing In addition to its benefits in infarct size reduction,
two birds with one stone. Both our group [20, 23, 12/15-LOX inhibition may thus independently
24, 33, 34] and others [35, 36] have in recent reduce hemorrhagic conversion of ischemic
years focused on developing improved inhibitors strokes by protecting the vasculature.
of 12/15-LOX, and it will be exciting to see if
these novel molecules can turn the tide in the Acknowledgments Funding from the National Institutes
seemingly endless war to combat stroke. The of Health (R01 NS049430, UG3 NS106854-01 and R21
NS087165 to KvL) and the American Heart Association
finding that we can reduce bleeding subsequent (17GRNT33460100 to KvL) is gratefully acknowledged.
to an ischemic stroke in the rodent model broad-
ens the spectrum of patients that could be treated
with a 12/15-LOX inhibitor. In addition to its use References
as a stand-alone neuroprotective treatment that
could already be given in the ambulance on the 1. Bazan NG Jr (1970) Effects of ischemia and elec-
way to the hospital, 12/15-LOX inhibition could troconvulsive shock on free fatty acid pool in the
also be combined with tPA thrombolysis to make brain. Biochim Biophys Acta 218(1):1–10. Epub
1970/10/06. PubMed PMID: 5473492
the use of tPA safer. By removing the most seri- 2. Moskowitz MA, Kiwak KJ, Hekimian K, Levine L
ous side effect of tPA, this approach may lead to (1984) Synthesis of compounds with properties of
significantly more patients receiving tPA treat- leukotrienes C4 and D4 in gerbil brains after ischemia
ment. Furthermore, the recently introduced endo- and reperfusion. Science 224(4651):886–889. Epub
1984/05/25. PubMed PMID: 6719118
vascular treatment in which a stent retriever is 3. Kitagawa K, Matsumoto M, Hori M (2004) Cerebral
used to remove the obstructing blood clot could ischemia in 5-lipoxygenase knockout mice. Brain
also be rendered more effective by adding a Res 1004(1–2):198–202. Epub 2004/03/23,
12/15-LOX inhibitor, because even after throm- https://doi.org/10.1016/j.brainres.2004.01.018.
S0006899304000587 [pii], PubMed PMID: 15033436
bus removal cognitive deficits are seen in many 4. Khanna S, Roy S, Slivka A, Craft TK, Chaki
patients who could benefit from the added neuro- S, Rink C et al (2005) Neuroprotective proper-
protection[37]. Finally, besides the more com- ties of the natural vitamin E alpha-tocotrienol.
mon ischemic strokes there may also be a place Stroke 36(10):2258–2264. Epub 2005/09/17,
doi: 01.STR.0000181082.70763.22 [pii], https://
for 12/15-LOX inhibition in the treatment of doi.org/10.1161/01.STR.0000181082.70763.22.
hemorrhagic strokes. We have recently com- PubMed PMID: 16166580; PubMed Central PMCID:
pleted a study to investigate the function of PMC1829173
12/15-LOX in subarachnoid hemorrhage, where
130 Y. Zheng et al.
5. van Leyen K, Kim HY, Lee SR, Jin G, Arai K, Lo 14. Kuhn H, Banthiya S, van Leyen K (2015) Mammalian
EH (2006) Baicalein and 12/15-lipoxygenase in the lipoxygenases and their biological relevance. Biochim
ischemic brain. Stroke 37(12):3014–3018. Epub Biophys Acta. 1851(4):308–330. Epub 2014/10/16.
2006/10/21, 01.STR.0000249004.25444.a5 [pii], https://doi.org/10.1016/j.bbalip.2014.10.002.
https://doi.org/10.1161/01.STR.0000249004.25444. PubMed PMID: 25316652; PubMed Central PMCID:
a5. PubMed PMID: 17053180 PMCPMC4370320
6. van Leyen K, Lee SR, Siddiq A, Ratan RR, Lo EH 15. Grüllich C, Duvoisin RM, Wiedmann M, van Leyen
(2004) 12/15-lipoxygenase and the proteasome as K (2001) Inhibition of 15-lipoxygenase leads to
mediators of neuronal oxidative stress and stroke. delayed organelle degradation in the reticulo-
Program No 1356, 2004 abstract viewer/itinerary cyte. FEBS Lett 489(1):51–54. Epub 2001/03/07.
planner. Society for Neuroscience, Washington, DC S0014- 5793(01)02080-4 [pii]. PubMed PMID:
7. Jung JE, Karatas H, Liu Y, Yalcin A, Montaner J, 11231012.
Lo EH et al (2015) STAT-dependent upregulation of 16. van Leyen K, Duvoisin RM, Engelhardt H, Wiedmann
12/15-lipoxygenase contributes to neuronal injury M (1998) A function for lipoxygenase in programmed
after stroke. J Cereb Blood Flow Metab. 35(12):2043– organelle degradation. Nature 395(6700):392–395.
2051. Epub 2015/07/16. https://doi.org/10.1038/ Epub 1998/10/06. https://doi.org/10.1038/26500.
jcbfm.2015.169. PubMed PMID: 26174325; PubMed PubMed PMID: 9759730.
Central PMCID: PMCPMC4671126 17. Koury MJ, Sawyer ST, Brandt SJ (2002) New insights
8. Jin G, Arai K, Murata Y, Wang S, Stins MF, Lo into erythropoiesis. Curr Opin Hematol 9(2):93–100.
EH et al (2008) Protecting against cerebrovascu- Epub 2002/02/15, PubMed PMID: 11844990.
lar injury: contributions of 12/15-lipoxygenase 18. Blanc L, Barres C, Bette-Bobillo P, Vidal M (2007)
to edema formation after transient focal isch- Reticulocyte-secreted exosomes bind natural IgM
emia. Stroke 39(9):2538–2543. Epub 2008/07/19. antibodies: involvement of a ROS-activatable endo-
doi: STROKEAHA.108.514927 [pii], https://doi. somal phospholipase iPLA2. Blood 110(9):3407–
org/10.1161/STROKEAHA.108.514927. PubMed 3416. Epub 2007/08/02. https://doi.org/10.1182/
PMID: 18635843; PubMed Central PMCID: blood-2007-04-085845. PubMed PMID: 17666570.
PMC2754072 19.
Sun D, Funk CD (1996) Disruption of
9. Li Y, Maher P, Schubert D (1997) A role for 12/15-lipoxygenase expression in peritoneal macro-
12-lipoxygenase in nerve cell death caused by glu- phages. Enhanced utilization of the 5-lipoxygenase
tathione depletion. Neuron 19(2):453–463. Epub pathway and diminished oxidation of low density
1997/08/01. S0896-6273(00)80953-8 [pii]. PubMed lipoprotein. J Biol Chem 271(39):24055–24062.
PMID: 9292733. Epub 1996/09/27, PubMed PMID: 8798642
10. Galluzzi L, Vitale I, Aaronson SA, Abrams JM,
20. Yigitkanli K, Pekcec A, Karatas H, Pallast S,
Adam D, Agostinis P et al (2018) Molecular mecha- Mandeville E, Joshi N et al (2013) Inhibition of
nisms of cell death: recommendations of the nomen- 12/15-lipoxygenase as therapeutic strategy to treat
clature committee on cell death 2018 . Cell Death stroke. Ann Neurol 73(1):129–135. https://doi.
Differ. 25(3):486–541. Epub 2018/01/25. https:// org/10.1002/ana.23734. PubMed PMID: 23192915;
doi.org/10.1038/s41418-017-0012-4. PubMed PubMed Central PMCID: PMC3563836
PMID: 29362479; PubMed Central PMCID: 21. Karatas H, Eun Jung J, Lo EH, van Leyen K (2018)
PMCPMC5864239 Inhibiting 12/15-lipoxygenase to treat acute stroke
11. Maher P, van Leyen K, Dey PN, Honrath B, Dolga in permanent and tPA induced thrombolysis models.
A, Methner A. (2018) The role of Ca(2+) in cell Brain Res 1678:123–128. https://doi.org/10.1016/j.
death caused by oxidative glutamate toxicity and fer- brainres.2017.10.024. PubMed PMID: 29079502;
roptosis. Cell Calcium 70:47–55. Epub 2017/05/27. PubMed Central PMCID: PMC5714685
https://doi.org/10.1016/j.ceca.2017.05.007. PubMed 22. Pfeilschifter W, Spitzer D, Czech-Zechmeister B,
PMID: 28545724; PubMed Central PMCID: Steinmetz H, Foerch C (2011) Increased risk of hem-
PMCPMC5682235 orrhagic transformation in ischemic stroke occurring
12. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush during warfarin anticoagulation: an experimental study
AI, Conrad M, Dixon SJ et al (2017) Ferroptosis: in mice. Stroke 42(4):1116–1121. Epub 2011/02/19.
a regulated cell death nexus linking metabolism, https://doi.org/10.1161/STROKEAHA.110.604652.
redox biology, and disease. Cell. 171(2):273– PubMed PMID: 21330626.
285. Epub 2017/10/07. https://doi.org/10.1016/j. 23. Liu Y, Zheng Y, Karatas H, Wang X, Foerch C, Lo
cell.2017.09.021. PubMed PMID: 28985560; EH et al (2017) 12/15-Lipoxygenase inhibition or
PubMed Central PMCID: PMCPMC5685180 Knockout Reduces Warfarin-associated hemor-
13. Schewe T, Halangk W, Hiebsch C, Rapoport SM
rhagic transformation after experimental stroke.
(1975) A lipoxygenase in rabbit reticulocytes Stroke. 48(2):445–451. Epub 2017/01/07. https://doi.
which attacks phospholipids and intact mitochon- org/10.1161/STROKEAHA.116.014790. PubMed
dria. FEBS Lett 60(1):149–152. Epub 1975/12/01. PMID: 28057806; PubMed Central PMCID:
0014-5793(75)80439-X [pii]. PubMed PMID: 6318. PMCPMC5263178
12 Contributions of 12/15-Lipoxygenase to Bleeding in the Brain Following Ischemic Stroke 131
24. Rai G, Joshi N, Jung JE, Liu Y, Schultz L, Yasgar A Epub 2015/01/21. https://doi.org/10.1038/srep07890.
et al (2014) Potent and selective inhibitors of human PubMed PMID: 25601765; PubMed Central PMCID:
reticulocyte 12/15-lipoxygenase as anti-stroke thera- PMCPMC4298747
pies. J Med Chem 57(10):4035–4048. https://doi. 32. Dilly AK, Ekambaram P, Guo Y, Cai Y, Tucker SC,
org/10.1021/jm401915r. PubMed PMID: 24684213; Fridman R et al (2013) Platelet-type 12-lipoxygenase
PubMed Central PMCID: PMC4033661 induces MMP9 expression and cellular invasion via
25. Piomelli D, Volterra A, Dale N, Siegelbaum SA,
activation of PI3K/Akt/NF-kappaB. Int J Cancer
Kandel ER, Schwartz JH et al (1987) Lipoxygenase 133(8):1784–1791. Epub 2013/03/26. https://doi.
metabolites of arachidonic acid as second messen- org/10.1002/ijc.28165. PubMed PMID: 23526143;
gers for presynaptic inhibition of Aplysia sensory PubMed Central PMCID: PMCPMC4269488
cells. Nature 328(6125):38–43. Epub 1987/07/02. 33. Armstrong MM, Freedman CJ, Jung JE, Zheng
https://doi.org/10.1038/328038a0. PubMed PMID: Y, Kalyanaraman C, Jacobson MP et al (2016) A
2439918. potent and selective inhibitor targeting human and
26. Mikule K, Gatlin JC, de la Houssaye BA, Pfenninger murine 12/15-LOX. Bioorg Med Chem 24(6):1183–
KH (2002) Growth cone collapse induced by sema- 1190. Epub 2016/02/24. https://doi.org/10.1016/j.
phorin 3A requires 12/15-lipoxygenase. J Neurosci bmc.2016.01.042. PubMed PMID: 26899595;
22(12):4932–4941. Epub 2002/06/22. doi: 22/12/4932 PubMed Central PMCID: PMCPMC4778748
[pii] PubMed PMID: 12077190. 34. van Leyen K, Arai K, Jin G, Kenyon V, Gerstner
27. Pekcec A, Yigitkanli K, Jung JE, Pallast S, Xing
B, Rosenberg PA et al (2008) Novel lipoxygenase
C, Antipenko A et al (2013) Following experi- inhibitors as neuroprotective reagents. J Neurosci
mental stroke, the recovering brain is vulnerable Res 86(4):904–909. Epub 2007/10/27. https://doi.
to lipoxygenase- dependent semaphorin signaling. org/10.1002/jnr.21543. PubMed PMID: 17960827;
FASEB J 27(2):437–445. https://doi.org/10.1096/ PubMed Central PMCID: PMC2759176
fj.12-206896. PubMed PMID: 23070608; PubMed 35. Han H, Liang X, Ekberg M, Kritikou JS, Brunnstrom
Central PMCID: PMC3545523 A, Pelcman B et al (2017) Human 15-lipoxygenase-1
28. He Z, Wang KC, Koprivica V, Ming G, Song HJ is a regulator of dendritic-cell spreading and podo-
(2002) Knowing how to navigate: mechanisms of some formation. FASEB J 31(2):491–504. Epub
semaphorin signaling in the nervous system. Sci 2016/11/09. https://doi.org/10.1096/fj.201600679RR.
STKE 2002(119):re1. Epub 2002/02/14. https:// PubMed PMID: 27825104.
doi.org/10.1126/stke.2002.119.re1. PubMed PMID: 36. Lapchak PA, Boitano PD, Bombien R, Cook DJ,
11842242. Doyan S, Lara JM et al (2018) CNB-001, a pleiotropic
29. Kolodkin AL, Tessier-Lavigne M (2011) Mechanisms drug is efficacious in embolized agyrencephalic New
and molecules of neuronal wiring: a primer. Cold Zealand white rabbits and ischemic gyrencephalic
Spring Harb Perspect Biol. 3(6). Epub 2010/12/03. cynomolgus monkeys. Exp Neurol 313:98–108.
https://doi.org/10.1101/cshperspect.a001727. Epub 2018/12/07. https://doi.org/10.1016/j.expneu-
PubMed PMID: 21123392; PubMed Central PMCID: rol.2018.11.010. PubMed PMID: 30521790
PMCPMC3098670 37. Berkhemer OA, Fransen PS, Beumer D, van den Berg
30. Jiang SX, Whitehead S, Aylsworth A, Slinn J,
LA, Lingsma HF, Yoo AJ et al (2015) A random-
Zurakowski B, Chan K et al (2010) Neuropilin 1 ized trial of intraarterial treatment for acute ischemic
directly interacts with Fer kinase to mediate sema- stroke. N Engl J Med 372(1):11–20. Epub 2014/12/18.
phorin 3A-induced death of cortical neurons. J Biol https://doi.org/10.1056/NEJMoa1411587. PubMed
Chem 285(13):9908-9918. Epub 2010/02/06. PMID: 25517348
https://doi.org/10.1074/jbc.M109.080689. PubMed 38. Gaberel T, Gakuba C, Zheng Y, Lepine M, Lo EH,
PMID: 20133938; PubMed Central PMCID: van Leyen K (2019) Impact of 12/15-Lipoxygenase
PMCPMC2843238 on Brain Injury After Subarachnoid Hemorrhage.
31. Hou ST, Nilchi L, Li X, Gangaraju S, Jiang SX,
Stroke 50(2): 520–523. STROKEAHA118022325.
Aylsworth A et al (2015) Semaphorin3A elevates https://doi.org/10.1161/STROKEAHA.118.022325.
vascular permeability and contributes to cerebral Epub 2019/01/04. PubMed PMID: 30602353.
ischemia-induced brain damage. Sci Rep 5:7890.
Systematic Understanding
of Bioactive Lipids 13
in Neuro-Immune Interactions:
Lessons from an Animal Model
of Multiple Sclerosis
Yasuyuki Kihara
Y. Kihara (*)
Sanford Burnham Prebys Medical Discovery
Institute, La Jolla, CA, USA
e-mail: kihara-[email protected]
13.2.1 Phospholipase A2s (PLA2s) progression [44, 47–50]. These results suggest
that eicosanoids, which are produced down-
PLA2s including cytosolic PLA2s (cPLA2), stream of PLA2, should have unique roles in EAE
secreted PLA2s (sPLA2) and calcium-independent and MS.
PLA2s (iPLA2), hydrolyze the sn-2 position of
membrane glycerophospholipids to liberate fatty
acyls including AA [43]. Among them, group 13.2.2 Cyclooxygenases (COXs)
IVA cPLA2 (cPLA2α; gene name, Pla2g4a) is a
key enzyme to produce eicosanoids, since its COXs (COX-1 and COX-2; gene names, Ptgs1
deficiency lacks the ability of producing eico- and Ptgs2, respectively) insert two molecular
sanoids, resulting in resistance to various disease oxygen into AA to produce an unstable endoper-
models [43]. Several lines of evidence, such as oxide intermediate, PGH2, which in turn metabo-
(1) up-regulation of Ca2+-dependent PLA2 activ- lizes to PGD2, PGE2, PGF2α, PGI2, and TXA2 by
ity and PLA2 mRNAs (cPLA2α, cPLA2β, cPLA2ζ, isomerases and terminal synthases [51].
and sPLA2-V) in EAE spinal cords (SCs) [38], Acetylsalicylic acid (Aspirin®) is the first non-
(2) cPLA2α expression in brain endothelium and steroidal anti-inflammatory drug (NSAID) syn-
infiltrated immune cells in EAE mice [44], and thesized by Bayer in 1897 [52]. The MOA of
(3) increased sPLA2 activity in both MS and EAE aspirin is suppression of prostaglandin synthesis
urine [45], suggest active involvement of these by inhibiting COXs, which was revealed by John
enzymes in EAE. Using cPLA2α-KO mice [46], Vane in 1971 [53].
Marusic et al. revealed the requirement of both There were no beneficial effects of aspirin in
peripheral and neuronal cPLA2α in EAE develop- guinea pig EAE [54, 55], while a high dose
ment and progression [47]. No other PLA2-KO sodium salicylate, a major metabolite of aspirin,
mice have been tested for EAE. Pharmacological showed efficacy in guinea pig EAE [56]. Two
inhibition of PLA2s (Pan inhibitors, AACOCF3 clinical trials in the early 1960’s examined aspi-
and FKGK2; cPLA2α inhibitor, WAY-196025; rin’s efficacy in MS failed [57, 58]. However,
sPLA2 inhibitor, CHEC-9; and iPLA2 inhibitor, mouse EAE studies tested other NSAIDs includ-
FKGK11) also blocked EAE development and ing indomethacin and naproxen (non-selective
13 Systematic Understanding of Bioactive Lipids in Neuro-Immune Interactions: Lessons from an Animal… 137
COX inhibitors) that clearly demonstrated the lesions [37]. mPGES-1 deficiency almost com-
involvement of COXs in EAE [50, 59]. Miyamoto pletely inhibited PGE2 production in EAE SCs,
et al. reported that COX-2-KO mice showed while doubling PGI2 production. mPEGS-1-KO
equivalent disease course with controls [59]. The mice showed EAE amelioration and impairment
study also demonstrated that a COX-2 selective of TH1/TH17 differentiation [37]. Later, another
inhibitor, celecoxib, prevented EAE in mice, group reported essentially the same results using
while COX-2-KO mice were not rescued by cele- the same mPGES-1-KO mice. They proposed
coxib. Thus, the MOA of celecoxib in EAE sup- that mPGES-1 in vascular endothelial cells con-
pression is independent from COX-2 inhibition trols IL-1β signaling in both CD4+ T cells and
[59]. Recently, the efficacy of low-dose aspirin (1endothelial cells, resulting in EAE exacerbation
~ 3 mg/kg), which is commonly used for lower- [65–67]. EAE phenomics using 8 PG receptor
ing cardiovascular risks [60], on the EAE pheno- KO mice was reported [68], which identified a
type was reported [61]. The study revealed that significant suppression of EAE only in PGE2
low-dose aspirin prevented loss of regulatory T receptor 4 (EP4; gene name, Ptger4)-KO mice on
cells (Treg) by stimulating IL-11 production, and the mixed background of C57BL/6 and 129/Ola.
also suppressed TH1/TH17 d ifferentiation, result- EAE phenotypes in other PGE2 receptors (EP1,
ing in EAE amelioration [61]. However, since EP2, and EP3) were equivalent to their matching
this MOA of low-dose aspirin is not tested in controls [68]. Prophylactic administration of the
COX-1/2-KO mice and eicosanoid levels were EP4 antagonist (ONO-AE3-208) prevented mice
not quantified, it remains unclear if the efficacy from developing EAE, while the therapeutic par-
of low-dose aspirin is related to COX-1/2 inhibi- adigm showed little effect on EAE development
tion. Considering the results from cPLA2α-KO and severity [68]. The efficacy of ONO-AE3-208
and COX-2-KO studies, COX-1 may play some was greater in EP2-KO mice than wild-type (WT)
role in EAE, but EAE has not been tested in mice, suggesting a redundancy of EP4 and EP2
COX-1-KO mice to date. [68]. Indeed, PGE2 facilitated TH1 differentiation
and IL-23-induced TH17 expansion through both
EP2 and EP4 [69]. These results clearly demon-
13.2.3 Prostaglandins (PGs) strate that the mPGES-1-PGE2-EP2/4 axis is an
exacerbating pathway in EAE and MS.
PGs are produced downstream of COX-1/2 and Although the EAE phenomics study con-
are functionally coupled with terminal PG syn- cluded no aggravation or facilitation of EAE in
thases to produce each PG efficiently [62–64], any PG receptor KO mice except for EP4-KO
followed by binding to their specific receptors (a mice, the clinical score of PGI2 receptor (IP; gene
member of the superfamily of G-protein coupled name, Ptgir)-KO mice appeared to show a right
receptors (GPCRs)), to elicit a variety of biologi- shift (delayed onset and/or milder EAE) as com-
cal responses [42]. pared to controls [68]. Indeed, another group
We applied targeted lipidomics using liquid reported a similar trend with significant differ-
chromatography tandem mass spectrometry (LC- ences between IP-KO mice vs. controls [70].
MS/MS) in combination with transcriptomics to They also demonstrated that PGI2-IP signaling
identify a key eicosanoid pathway that is involved induces IL-17A production in naïve CD4+ T cells
in EAE [37]. Lipidomics identified a metabolic and promotes TH17 differentiation [70]. The
shift from the PGD2 pathway in naïve spinal decreased PGI2 in EAE SCs could be explained
cords (SCs) to the PGE2 pathway in EAE SCs by down-regulation of PGI synthase (PGIS; gene
[37]. Among the three PGE2 synthases, micro- name, Ptgis) mRNA that is abundant in endothe-
somal PGE synthase 1 (mPGES-1; gene name, lial cells [37], suggesting a direct neuroprotective
Ptges) mRNA was uniquely up-regulated in EAE role of PGI2 in the CNS. Muramatsu et al. reported
SCs. mPGES-1 protein was expressed in that knockdown of PGIS and IP by siRNA deliv-
microglia/macrophages in both EAE and MS ered directly into the CNS, delayed recovery
138 Y. Kihara
metabolized to LTD4 followed by LTE4 [88, 89]. trial using ginkgolide B, a PAFR antagonist
Levels of these 5-LO metabolites in naïve SCs extracted from Ginkgo biloba, showed no effi-
were very low as compared to COX metabolites, cacy for the treatment of acute exacerbations of
which were further declined in EAE SCs [37]. MS [95], several lines of evidence support an
Therefore, LTs may not play pivotal roles in involvement of PAF in MS/EAE to certain
EAE. However, mRNAs of the LTB4 receptor 1 extent, including (1) elevated PAF levels in cere-
(BLT1; gene name, Ltb4r1) and the cys-LT recep- brospinal fluids (CSF) and plasma of RRMS
tor 1 (CysLT1; gene name, Cysltr1) were highly patients [96], (2) up-regulation of PAFR mRNA
up-regulated in EAE SCs from disease onset in chronic MS plaques [97], (3) a relationship
through the acute phase [37, 39], implying accu- between PAFR gene missense mutation vs. the
mulation of immune cells expressing these recep- susceptibility for MS [98], and (4) an efficacy of
tors into the CNS. The BLT1 antagonist, PAFR antagonist (BN52021) on EAE [99]. We
CP-105,696, prevented EAE and suppressed found increased activity of PAF producing
eosinophil infiltration into the CNS in a enzymes (PLA2 and LysoPAFAT) and unchanged
dose-dependent manner [90]. We reported that PAFAH activity in EAE SCs as compared to
BLT1-KO mice showed delayed onset, less severe controls, resulting in a significant elevation of
EAE, and reduced TH1/TH17 responses than con- PAF levels [36, 38]. We also tested EAE in
trols [39]. Moreover, BLT1-KO SCs from asymp- PAFR-KO mice that showed a lower incidence
tomatic EAE mice showed no T cell, neutrophil, and less severe disease than controls [36]. PAF-
or macrophage infiltrations, whereas those cells PAFR signaling promoted cytokine and chemo-
were found in asymptomatic WT EAE controls kine production in SCs before disease onset [36].
[39]. These results suggest that BLT1 is respon- In addition, PAF accelerated phagocytotic activ-
sible for immune cell recruitment into the CNS ity and its associated TNF-α production in mac-
in the early phase of the disease. EAE studies rophages via PAFR [36]. Another group
using KO mice of LTA4H, LTC4S, BLT2 (gene independently confirmed these results using the
name, Ltb4r2), CysLT1, 2, 3 (gene name, Cysltr1, same PAFR-KO mice [100]. These results sug-
Cysltr2, Cyltr3/Oxgr1/Gpr99) are not available gest a dual role of PAF-PAFR signaling in EAE
at this time. development and progression.
Fig. 13.2 EAE phenotypes in LP signaling-KO mice. (a) LPA pathway. (b) S1P pathway
13.4.1 Lysophosphatidic Acid (LPA) carrying TCR specific for MOG92-106 in the con-
text of I-As in SJL/J genetic background; TCR1640
LPA is produced by three distinct pathways: (1) mice [115]), unsaturated LPAs in plasma and SCs
de novo synthesis from glycerol-3-phosphate were highly elevated during the first remission,
(G3P) by G3P acyltransferase (GPAT; gene followed by suppression in the second relapse
names, Gpam, Gpat2, Gpat3 and Gpat4) [93, [114]. Receptor expression profiles in spleen
104], (2) head group removal from LPs by auto- cells of EAE mice identified a suppression of
taxin (ATX; gene name, Enpp2) [105, 106], and LPA2 protein expression on CD4+ T cells and
(3) hydrolysis of phosphatidic acid (PA) by myeloid cells [114]. LPA1 mRNA in spleen, LPA2
PA-specific PLA1 (PA-PLA1α; gene name, Liph) and LPA3 mRNA in white blood cells, and LPA2,
[105, 107]. LPA levels in blood (0.1 μM in plasma LPA3, LPA4 and LPA5 mRNA in SCs were up-
~ 10 μM in serum) are of sufficiently high con- regulated in EAE mice [114]. In LPA2-KO mice
centration to activate six cognate GPCRs (LPA1-6; on a C57BL6 x Sv129 mixed background, about
gene names, Lpar1-6) [101, 108–111], in which 25% developed EAE with increased T cell and
LPA1 dissociation constants (Kd) for LPA approx- myeloid cell infiltration into the SCs, while their
imated subnanomolar range [112]. Although LPA matching WT controls showed very mild disease
was isolated and identified from soy beans in course because Sv129 mice are resistant to EAE
1978 [113], it has not been well-studied in MS [114]. LPA2 agonist, GRI-977,143, effectively
and EAE until recently. In 2017, Schmitz et al. ameliorated EAE in SJL mice [114].
reported a meticulous investigation of the rela- Controversial, small cohort studies were also
tionship between LPA and MS and EAE [114]. reported that LPA levels increased in serum of
LPA concentrations quantified by LC-MS/MS RRMS patients, while they were determined by
were significantly reduced in the serum of MS ELISA or phosphate quantification [116, 117].
patients, regardless of disease type, and EAE ATX activity was elevated in the CSF and serum
mice on an SJL/J background as compared to of RRMS patients [118]. A naturally occurring
their controls [114]. Moreover, unsaturated LPA LPA analog, cyclic phosphatidic acid (cPA,
levels in serum were responsive to some DMTs 1-acyl-sn-glycero-2,3-cyclic phosphate), is
(fingolimod and natalizumab), which were also reported to activate LPA receptors [119].
observed in EAE mice on an SJL/J background Treatment using a metabolically stable analog of
[114]. In a spontaneous relapsing-remitting EAE cPA (2ccPA) in both a prophylactic and therapeu-
model (T cell receptor (TCR) transgenic mice tic manner ameliorated EAE and reduced inflam-
13 Systematic Understanding of Bioactive Lipids in Neuro-Immune Interactions: Lessons from an Animal… 141
mation in the SCs [120], which may be mediated administration of fingolimod. Moreover, fingoli-
via LPA2. The EAE course of LPA4-KO mice was mod-P was identified as a high affinity agonist to
equivalent to controls (unpublished observation). four out of the five known S1P receptors (S1P1,
The EAE disease course in LPA1, LPA3, LPA5, S1P3, S1P4, and S1P5). Brinkman et al. also
and LPA6 deficiency, as well as LPA-producing reported the efficacy of fingolimod in EAE by a
and degrading enzyme deficiency, need to be prophylactic treatment paradigm [127]. In 2003,
studied to characterize their roles in EAE. Sphk2 was reported to show a greater effect of
fingolimod phosphorylation as compared to
Sphk1 [129]. In 2004, Jason Cyster’s group
13.4.2 Sphingosine 1-phosphate revised the MOA of fingolimod from agonism to
(S1P) functional antagonism based upon findings that
(1) S1P1 was required for lymphocyte egress
S1P is produced from sphingosine (2-amino-4- from lymphoid organs to circulation, and (2) fin-
octadecene-1,3-diol) by the action of sphingosine golimod treatment down-regulated S1P1 cell sur-
kinases (Sphks; gene name, Sphk1, Sphk2) [121]. face expression [130, 131]. These studies
S1P is enriched in blood plasma (low micromolar provided a basis for the biological significance of
range), where S1P binds to albumin (gene name, the S1P gradient in lymphocyte trafficking
Alb) and apolipoprotein M (ApoM; gene name, between circulatory and lymph systems, which
Apom) tethered to high-density lipoprotein are supported by other studies using S1P-
(HDL) at a ratio of approximately three to seven generating (Sphks) and -degrading (S1P lyase;
[122]. S1P levels in lymph and tissues are lower gene name, Sgpl1) enzyme KO mice [132, 133].
than in blood (nanomolar range), which gener- Additional studies provided more evidence of
ates an S1P gradient between the circulatory sys- S1P1 involvement in MS and EAE. Proteomics
tem and tissues [122]. The biological significance analysis on MS brain tissue identified S1P1 phos-
of this gradient was recognized about 10 years phorylation on a C-terminal serine residue that is
after the discovery of fingolimod (trade name, crucial for receptor internalization [134]. A
Gilenya™), originally called FTY720. knock-in mouse harboring alanine mutations on
In 1995, fingolimod was first synthesized S1P1 C-terminal serine residues (S1P1-S5A mice)
from an immunosuppressive natural product, [135] developed more severe EAE than controls
myriocin (ISP-I), which was isolated from cul- with an enhanced TH17 immunity. S1P1 floxed
ture broths of Isaria sinclairii [123–125]. Chiba mice (S1P1flox/flox) were used to study cell-specific
et al. reported that fingolimod strikingly reduced roles of S1P1 in EAE, since global S1P1-KO mice
the number of circulating lymphocytes [125], were embryonically lethal due to impairment of
while the molecular basis of fingolimod activity vascular maturation [136]. TH17-specific
remained unclear at that time. This immunosup- S1P1-KO mice (IL-17A-Cre: S1P1flox/flox mice)
pressive effect of fingolimod was clinically tested were completely resistant to EAE development
for preventing organ graft rejection, while phase [137]. On the other hand, Treg-specific S1P1-KO
III clinical trials did not support a superior effi- mice (Foxp3-Cre: S1P1flox/flox mice) developed
cacy of fingolimod in renal transplantation as systemic autoimmunity, since Treg cells were
compared to the prior standard treatment [126]. retained in lymphoid organs and reduced in
During 1998–2000, five S1P receptors (S1P1-5: peripheral tissues. Also, S1P1 deletion, as well as
gene names, S1pr1-5) that belong to rhodopsin fingolimod treatment, promoted to effector Treg
family GPCRs were identified [101, 108]. In cell conversion from central Treg cell. Tamoxifen-
2002, the MOA of fingolimod was independently inducible Treg-specific S1P1-KO mice (Foxp3-
proposed by two pharmaceutical companies, CreERT2: S1P1flox/flox mice) developed severe EAE
Merck and Novartis [127, 128]. Both groups [137]. The immunological roles of ApoM-bound
clearly demonstrated that phosphorylated fingoli- S1P were investigated using ApoM-KO mice,
mod (fingolimod-P) was generated in vivo after which revealed that ApoM-S1P suppresses
142 Y. Kihara
LPA1, S1P5, and GPR17) that are highly expressed 6. Filiano AJ, Xu Y, Tustison NJ, Marsh RL, Baker
W, Smirnov I, Overall CC, Gadani SP, Turner SD,
in oligodendrocytes and anti-inflammatory/pro- Weng Z, Peerzade SN, Chen H, Lee KS, Scott
resolving lipid mediators (EETs and SPMs), are MM, Beenhakker MP, Litvak V, Kipnis J (2016)
attractive neuroprotective targets to study in Unexpected role of interferon-gamma in regulating
EAE/MS. neuronal connectivity and social behaviour. Nature
535:425–429
7. Hickey WF (1999) The pathology of multiple scle-
Acknowledgements Thanks to Prof. Takao Shimizu and rosis: a historical perspective. J Neuroimmunol
all the members in his laboratory (The University of 98:37–44
Tokyo), and Prof. Takehiko Yokomizo (Juntendo 8. Murray TJ (2009) The history of multiple sclerosis:
University) and Prof. Satoshi Ishii (Akita University) for the changing frame of the disease over the centuries.
their support to complete my Ph.D. work, Profs. K. Frank J Neurol Sci 277(Suppl 1):S3–S8
Austen and Yoshihide Kanaoka (Harvard University) for 9. Young IR, Hall AS, Pallis CA, Legg NJ, Bydder
providing me an opportunity to study in the U.S.A., Profs. GM, Steiner RE (1981) Nuclear magnetic resonance
Edward A. Dennis and Shankar Subramaniam for accep- imaging of the brain in multiple sclerosis. Lancet
tance to join LIPID MAPS®, Prof. Jerold Chun (Sanford 2:1063–1066
Burnham Prebys Medical Discovery Institute (SBP)) for 10. Mcdonald WI, Compston A, Edan G, Goodkin D,
his continuous support and encouragement, Dr. Deepa Hartung HP, Lublin FD, Mcfarland HF, Paty DW,
Jonnalagadda (SBP) for helpful discussions, and Ms. Polman CH, Reingold SC, Sandberg-Wollheim
Danielle Jones (SBP) for editorial assistance. This work M, Sibley W, Thompson A, Van Den Noort S,
was supported by a grant from NIH/NINDS R01NS103940 Weinshenker BY, Wolinsky JS (2001) Recommended
and fellowships from Japan Society for the Promotion of diagnostic criteria for multiple sclerosis: guidelines
Science and Human Frontier Science Program. from the International Panel on the diagnosis of mul-
tiple sclerosis. Ann Neurol 50:121–127
Conflict of Interest Y.K. declares no competing financial 11. Thompson AJ, Banwell BL, Barkhof F, Carroll WM,
interests. Coetzee T, Comi G, Correale J, Fazekas F, Filippi
M, Freedman MS, Fujihara K, Galetta SL, Hartung
HP, Kappos L, Lublin FD, Marrie RA, Miller AE,
Miller DH, Montalban X, Mowry EM, Sorensen PS,
References Tintore M, Traboulsee AL, Trojano M, Uitdehaag
BMJ, Vukusic S, Waubant E, Weinshenker BG,
1. Carson MJ, Doose JM, Melchior B, Schmid CD, Reingold SC, Cohen JA (2018) Diagnosis of mul-
Ploix CC (2006) CNS immune privilege: hiding in tiple sclerosis: 2017 revisions of the McDonald cri-
plain sight. Immunol Rev 213:48–65 teria. Lancet Neurol 17:162–173
2. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani 12. Kurtzke JF (1983) Rating neurologic impairment
SJ, Peske JD, Derecki NC, Castle D, Mandell JW, in multiple sclerosis: an expanded disability status
Lee KS, Harris TH, Kipnis J (2015) Structural and scale (EDSS). Neurology 33:1444–1452
functional features of central nervous system lym- 13. Kurtzke JF (2015) On the origin of EDSS. Mult
phatic vessels. Nature 523:337–341 Scler Relat Disord 4:95–103
3. Herisson F, Frodermann V, Courties G, Rohde D, 14. Medaer R (1979) Does the history of multiple scle-
Sun Y, Vandoorne K, Wojtkiewicz GR, Masson GS, rosis go back as far as the 14th century? Acta Neurol
Vinegoni C, Kim J, Kim DE, Weissleder R, Swirski Scand 60:189–192
FK, Moskowitz MA, Nahrendorf M (2018) Direct 15. Leray E, Moreau T, Fromont A, Edan G (2016)
vascular channels connect skull bone marrow and Epidemiology of multiple sclerosis. Rev Neurol
the brain surface enabling myeloid cell migration. (Paris) 172:3–13
Nat Neurosci 21:1209–1217 16. Lancet T (2018) End of the road for daclizumab in
4. Arima Y, Harada M, Kamimura D, Park JH, multiple sclerosis. Lancet 391:1000
Kawano F, Yull FE, Kawamoto T, Iwakura Y, Betz 17. Derfuss T, Kuhle J, Lindberg R, Kappos L (2013)
UA, Marquez G, Blackwell TS, Ohira Y, Hirano Natalizumab therapy for multiple sclerosis. Semin
T, Murakami M (2012) Regional neural activation Neurol 33:26–36
defines a gateway for autoreactive T cells to cross the 18. Willis MD, Robertson NP (2016) Alemtuzumab for
blood-brain barrier. Cell 148:447–457 multiple sclerosis. Curr Neurol Neurosci Rep 16:84
5. Da Mesquita S, Louveau A, Vaccari A, Smirnov 19. Hauser SL, Bar-Or A, Comi G, Giovannoni G,
I, Cornelison RC, Kingsmore KM, Contarino C, Hartung HP, Hemmer B, Lublin F, Montalban X,
Onengut-Gumuscu S, Farber E, Raper D, Viar KE, Rammohan KW, Selmaj K, Traboulsee A, Wolinsky
Powell RD, Baker W, Dabhi N, Bai R, Cao R, Hu S, JS, Arnold DL, Klingelschmitt G, Masterman D,
Rich SS, Munson JM, Lopes MB, Overall CC, Acton Fontoura P, Belachew S, Chin P, Mairon N, Garren
ST, Kipnis J (2018) Functional aspects of menin- H, Kappos L, Opera I, Investigators OIC (2017)
geal lymphatics in ageing and Alzheimer’s disease. Ocrelizumab versus interferon Beta-1a in relapsing
Nature 560:185–191 multiple sclerosis. N Engl J Med 376:221–234
144 Y. Kihara
20. Jakimovski D, Kolb C, Ramanathan M, Zivadinov 34. Langrish CL, Chen Y, Blumenschein WM, Mattson
R, Weinstock-Guttman B (2018) Interferon beta for J, Basham B, Sedgwick JD, Mcclanahan T, Kastelein
multiple sclerosis. Cold Spring Harb Perspect Med RA, Cua DJ (2005) IL-23 drives a pathogenic T cell
8:a032003 population that induces autoimmune inflammation.
21. Chun J, Brinkmann V (2011) A mechanistically J Exp Med 201:233–240
novel, first oral therapy for multiple sclerosis: the 35. Yu CY, Whitacre CC (2004) Sex, MHC and comple-
development of fingolimod (FTY720, Gilenya). ment C4 in autoimmune diseases. Trends Immunol
Discov Med 12:213–228 25:694–699
22. Cohen JA, Chun J (2011) Mechanisms of fingoli- 36. Kihara Y, Ishii S, Kita Y, Toda A, Shimada A,
mod’s efficacy and adverse effects in multiple scle- Shimizu T (2005) Dual phase regulation of experi-
rosis. Ann Neurol 69:759–777 mental allergic encephalomyelitis by platelet-
23. Kihara Y, Groves A, Rivera RR, Chun J (2015a) activating factor. J Exp Med 202:853–863
Dimethyl fumarate inhibits integrin alpha4 expres- 37. Kihara Y, Matsushita T, Kita Y, Uematsu S, Akira S,
sion in multiple sclerosis models. Ann Clin Transl Kira J, Ishii S, Shimizu T (2009) Targeted lipido-
Neurol 2:978–983 mics reveals mPGES-1-PGE2 as a therapeutic tar-
24. Von Glehn F, Dias-Carneiro RPC, Moraes AS, Farias get for multiple sclerosis. Proc Natl Acad Sci U S A
AS, Silva V, Oliveira FTM, Silva C, De Carvalho 106:21807–21812
F, Rahal E, Baecher-Allan C, Santos LMB (2018) 38. Kihara Y, Yanagida K, Masago K, Kita Y, Hishikawa
Dimethyl fumarate downregulates the immune D, Shindou H, Ishii S, Shimizu T (2008) Platelet-
response through the HCA2/GPR109A pathway: activating factor production in the spinal cord of
implications for the treatment of multiple sclerosis. experimental allergic encephalomyelitis mice via the
Mult Scler Relat Disord 23:46–50 group IVA cytosolic phospholipase A2-lyso-PAFAT
25. Groves A, Kihara Y, Chun J (2013) Fingolimod: axis. J Immunol 181:5008–5014
direct CNS effects of sphingosine 1-phosphate (S1P) 39. Kihara Y, Yokomizo T, Kunita A, Morishita Y,
receptor modulation and implications in multiple Fukayama M, Ishii S, Shimizu T (2010) The leukot-
sclerosis therapy. J Neurol Sci 328:9–18 riene B4 receptor, BLT1, is required for the induc-
26. Rothhammer V, Mascanfroni ID, Bunse L, Takenaka tion of experimental autoimmune encephalomyelitis.
MC, Kenison JE, Mayo L, Chao CC, Patel B, Yan R, Biochem Biophys Res Commun 394:673–678
Blain M, Alvarez JI, Kebir H, Anandasabapathy N, 40. Terry RL, Ifergan I, Miller SD (2016) Experimental
Izquierdo G, Jung S, Obholzer N, Pochet N, Clish autoimmune encephalomyelitis in Mice. Methods
CB, Prinz M, Prat A, Antel J, Quintana FJ (2016) Mol Biol 1304:145–160
Type I interferons and microbial metabolites of tryp- 41. Groves A, Kihara Y, Jonnalagadda D, Rivera R,
tophan modulate astrocyte activity and central ner- Kennedy G, Mayford M, Chun J (2018) A function-
vous system inflammation via the aryl hydrocarbon ally defined in vivo astrocyte population identified
receptor. Nat Med 22:586–597 by c-Fos activation in a mouse model of multiple
27. Al-Jaderi Z, Maghazachi AA (2016) Utilization of sclerosis modulated by S1P signaling: immediate-
Dimethyl Fumarate and related molecules for treat- early Astrocytes (ieAstrocytes). eNeuro 5
ment of multiple sclerosis, cancer, and other dis- 42. Shimizu T (2009) Lipid mediators in health and dis-
eases. Front Immunol 7:278 ease: enzymes and receptors as therapeutic targets
28. Rivers TM, Sprunt DH, Berry GP (1933) for the regulation of immunity and inflammation.
Observations on Attempts to Produce Acute Annu Rev Pharmacol Toxicol 49:123–150
Disseminated Encephalomyelitis in Monkeys. J Exp 43. Kita Y, Ohto T, Uozumi N, Shimizu T (2006)
Med 58:39–53 Biochemical properties and pathophysiological roles
29. Paterson PY (1960) Transfer of allergic encephalo- of cytosolic phospholipase A2s. Biochim Biophys
myelitis in rats by means of lymph node cells. J Exp Acta 1761:1317–1322
Med 111:119–136 44. Kalyvas A, David S (2004) Cytosolic phospholipase
30. Ben-Nun A, Wekerle H, Cohen IR (1981) A2 plays a key role in the pathogenesis of multiple
Vaccination against autoimmune encephalomyelitis sclerosis-like disease. Neuron 41:323–335
with T-lymphocyte line cells reactive against myelin 45. Cunningham TJ, Yao L, Oetinger M, Cort L,
basic protein. Nature 292:60–61 Blankenhorn EP, Greenstein JI (2006) Secreted
31. Zamvil SS, Mitchell DJ, Moore AC, Kitamura K, phospholipase A2 activity in experimental autoim-
Steinman L, Rothbard JB (1986) T-cell epitope of mune encephalomyelitis and multiple sclerosis.
the autoantigen myelin basic protein that induces J Neuroinflammation 3:26
encephalomyelitis. Nature 324:258–260 46. Uozumi N, Kume K, Nagase T, Nakatani N, Ishii
32. Rangachari M, Kuchroo VK (2013) Using EAE to S, Tashiro F, Komagata Y, Maki K, Ikuta K, Ouchi
better understand principles of immune function and Y, Miyazaki J, Shimizu T (1997) Role of cytosolic
autoimmune pathology. J Autoimmun 45:31–39 phospholipase A2 in allergic response and parturi-
33. Windhagen A, Nicholson LB, Weiner HL, Kuchroo tion. Nature 390:618–622
VK, Hafler DA (1996) Role of Th1 and Th2 cells in 47. Marusic S, Leach MW, Pelker JW, Azoitei ML,
neurologic disorders. Chem Immunol 63:171–186 Uozumi N, Cui J, Shen MW, Declercq CM,
13 Systematic Understanding of Bioactive Lipids in Neuro-Immune Interactions: Lessons from an Animal… 145
Miyashiro JS, Carito BA, Thakker P, Simmons DL, mune encephalomyelitis through interleukin-11-
Leonard JP, Shimizu T, Clark JD (2005) Cytosolic mediated protection of regulatory T cells. Sci Signal
phospholipase A2 alpha-deficient mice are resistant 11:eaar8278
to experimental autoimmune encephalomyelitis. 62. Kihara Y, Gupta S, Maurya MR, Armando A,
J Exp Med 202:841–851 Shah I, Quehenberger O, Glass CK, Dennis EA,
48. Kalyvas A, Baskakis C, Magrioti V, Constantinou- Subramaniam S (2014a) Modeling of eicosanoid
Kokotou V, Stephens D, Lopez-Vales R, Lu JQ, fluxes reveals functional coupling between cyclo-
Yong VW, Dennis EA, Kokotos G, David S (2009) oxygenases and terminal synthases. Biophys
Differing roles for members of the phospholipase A2 J 106:966–975
superfamily in experimental autoimmune encepha- 63. Murakami M, Kambe-Ohkura T, Kudo I (2002)
lomyelitis. Brain 132:1221–1235 Functional coupling between phospholipase A2S
49. Marusic S, Thakker P, Pelker JW, Stedman NL, Lee and cyclooxygenases in immediate and delayed
KL, Mckew JC, Han L, Xu X, Wolf SF, Borey AJ, prostanoid biosynthetic pathways. Adv Exp Med
Cui J, Shen MW, Donahue F, Hassan-Zahraee M, Biol 507:15–19
Leach MW, Shimizu T, Clark JD (2008) Blockade 64. Ueno N, Takegoshi Y, Kamei D, Kudo I, Murakami
of cytosolic phospholipase A2 alpha prevents M (2005) Coupling between cyclooxygenases and
experimental autoimmune encephalomyelitis and terminal prostanoid synthases. Biochem Biophys
diminishes development of Th1 and Th17 responses. Res Commun 338:70–76
J Neuroimmunol 204:29–37 65. Takemiya T, Kawakami M, Takeuchi C (2018)
50. Thakker P, Marusic S, Stedman NL, Lee KL, Mckew Endothelial microsomal prostaglandin E
JC, Wood A, Goldman SJ, Leach MW, Collins M, Synthetase-1 Upregulates vascularity and endothe-
Kuchroo VK, Wolf SF, Clark JD, Hassan-Zahraee M lial interleukin-1beta in deteriorative progression
(2011) Cytosolic phospholipase A2alpha blockade of experimental autoimmune encephalomyelitis. Int
abrogates disease during the tissue-damage effector J Mol Sci 19:3647
phase of experimental autoimmune encephalomyeli- 66. Takemiya T, Takeuchi C, Kawakami M (2017)
tis by its action on APCs. J Immunol 187:1986–1997 Microsomal prostaglandin E Synthase-1 facilitates
51. Smith WL, Urade Y, Jakobsson PJ (2011) Enzymes an intercellular interaction between CD4(+) T cells
of the cyclooxygenase pathways of prostanoid bio- through IL-1beta Autocrine function in experimen-
synthesis. Chem Rev 111:5821–5865 tal autoimmune encephalomyelitis. Int J Mol Sci
52. Vane JR, Botting RM (2003) The mechanism of 18:2758
action of aspirin. Thromb Res 110:255–258 67. Takeuchi C, Matsumoto Y, Kohyama K, Uematsu
53. Vane JR (1971) Inhibition of prostaglandin synthesis S, Akira S, Yamagata K, Takemiya T (2013)
as a mechanism of action for aspirin-like drugs. Nat Microsomal prostaglandin E synthase-1 aggravates
New Biol 231:232–235 inflammation and demyelination in a mouse model
54. Kolb LC, Karlson AG, Sayre GP (1952) Prevention of multiple sclerosis. Neurochem Int 62:271–280
of experimental allergic encephalomyelitis by vari- 68. Esaki Y, Li Y, Sakata D, Yao C, Segi-Nishida E,
ous agents. Trans Am Neurol Assoc 56:117–121 Matsuoka T, Fukuda K, Narumiya S (2010) Dual
55. Tsau S, Emerson MR, Lynch SG, Levine SM (2015) roles of PGE2-EP4 signaling in mouse experimental
Aspirin and multiple sclerosis. BMC Med 13:153 autoimmune encephalomyelitis. Proc Natl Acad Sci
56. Good RA, Campbell B, Good TA (1949) Prophylactic U S A 107:12233–12238
and therapeutic effect of para-aminobenzoic acid and 69. Yao C, Sakata D, Esaki Y, Li Y, Matsuoka T, Kuroiwa
sodium salicylate on experimental allergic encepha- K, Sugimoto Y, Narumiya S (2009) Prostaglandin
lomyelitis. Proc Soc Exp Biol Med 72:341–347 E2-EP4 signaling promotes immune inflamma-
57. Miller H, Newell DJ, Ridley A (1961) Multiple scle- tion through Th1 cell differentiation and Th17 cell
rosis. Trials of maintenance treatment with predniso- expansion. Nat Med 15:633–640
lone and soluble aspirin. Lancet 1:127–129 70. Zhou W, Dowell DR, Huckabee MM, Newcomb DC,
58. Miller HG, Foster JB, Newell DJ, Barwick DD, Boswell MG, Goleniewska K, Lotz MT, Toki S, Yin
Brewis RA (1963) Multiple sclerosis: therapeutic H, Yao S, Natarajan C, Wu P, Sriram S, Breyer RM,
trials of chloroquine, soluble aspirin, and gamma- Fitzgerald GA, Peebles RS Jr (2012) Prostaglandin
globulin. Br Med J 2:1436–1439 I2 signaling drives Th17 differentiation and exacer-
59. Miyamoto K, Miyake S, Mizuno M, Oka N, bates experimental autoimmune encephalomyelitis.
Kusunoki S, Yamamura T (2006) Selective COX-2 PLoS One 7:e33518
inhibitor celecoxib prevents experimental auto- 71. Muramatsu R, Takahashi C, Miyake S, Fujimura
immune encephalomyelitis through COX-2- H, Mochizuki H, Yamashita T (2012) Angiogenesis
independent pathway. Brain 129:1984–1992 induced by CNS inflammation promotes neuronal
60. Patrono C, Garcia Rodriguez LA, Landolfi R, Baigent remodeling through vessel-derived prostacyclin. Nat
C (2005) Low-dose aspirin for the prevention of ath- Med 18:1658–1664
erothrombosis. N Engl J Med 353:2373–2383 72. Narumiya S, Ogorochi T, Nakao K, Hayaishi O
61. Mondal S, Jana M, Dasarathi S, Roy A, Pahan K (1982) Prostaglandin D2 in rat brain, spinal cord and
(2018) Aspirin ameliorates experimental autoim-
146 Y. Kihara
pituitary: basal level and regional distribution. Life 85. Whitney LW, Ludwin SK, Mcfarland HF, Biddison
Sci 31:2093–2103 WE (2001) Microarray analysis of gene expres-
73. Huang YC, Lyu RK, Tseng MY, Chang HS, Hsu sion in multiple sclerosis and EAE identifies
WC, Kuo HC, Chu CC, Wu YR, Ro LS, Huang CC, 5-lipoxygenase as a component of inflammatory
Chen CM (2009) Decreased intrathecal synthesis of lesions. J Neuroimmunol 121:40–48
prostaglandin D2 synthase in the cerebrospinal fluid 86. Emerson MR, Levine SM (2004) Experimental
of patients with acute inflammatory demyelinating allergic encephalomyelitis is exacerbated in mice
polyneuropathy. J Neuroimmunol 206:100–105 deficient for 12/15-lipoxygenase or 5-lipoxygenase.
74. Kagitani-Shimono K, Mohri I, Oda H, Ozono K, Brain Res 1021:140–145
Suzuki K, Urade Y, Taniike M (2006) Lipocalin-type 87. Yokomizo T, Nakamura M, Shimizu T (2018)
prostaglandin D synthase (beta-trace) is upregulated Leukotriene receptors as potential therapeutic tar-
in the alphaB-crystallin-positive oligodendrocytes gets. J Clin Invest 128:2691–2701
and astrocytes in the chronic multiple sclerosis. 88. Kanaoka Y, Boyce JA (2014) Cysteinyl leukotrienes
Neuropathol Appl Neurobiol 32:64–73 and their receptors; emerging concepts. Allergy
75. Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, Zhang Asthma Immunol Res 6:288–295
X, Yin KJ, Gao Y, Bennett MVL, Leak RK, Chen 89. NationalMSSociety.org (2018) National Multiple
J (2018) Peroxisome proliferator-activated receptor Sclerosis Society [Online]. Available: https://www.
gamma (PPARgamma): a master gatekeeper in CNS nationalmssociety.org/. Accessed 2018
injury and repair. Prog Neurobiol 163–164:27–58 90. Gladue RP, Carroll LA, Milici AJ, Scampoli DN,
76. Natarajan C, Muthian G, Barak Y, Evans RM, Stukenbrok HA, Pettipher ER, Salter ED, Contillo
Bright JJ (2003) Peroxisome proliferator-activated L, Showell HJ (1996) Inhibition of leukotriene
receptor-gamma-deficient heterozygous mice B4-receptor interaction suppresses eosinophil infil-
develop an exacerbated neural antigen-induced Th1 tration and disease pathology in a murine model of
response and experimental allergic encephalomyeli- experimental allergic encephalomyelitis. J Exp Med
tis. J Immunol 171:5743–5750 183:1893–1898
77. Raikwar HP, Muthian G, Rajasingh J, Johnson C, 91. Honda Z, Ishii S, Shimizu T (2002) Platelet-
Bright JJ (2005) PPARgamma antagonists exacer- activating factor receptor. J Biochem 131:773–779
bate neural antigen-specific Th1 response and exper- 92. Honda Z, Nakamura M, Miki I, Minami M, Watanabe
imental allergic encephalomyelitis. J Neuroimmunol T, Seyama Y, Okado H, Toh H, Ito K, Miyamoto T,
167:99–107 Et AL (1991) Cloning by functional expression of
78. Diab A, Deng C, Smith JD, Hussain RZ, Phanavanh platelet-activating factor receptor from guinea-pig
B, Lovett-Racke AE, Drew PD, Racke MK (2002) lung. Nature 349:342–346
Peroxisome proliferator-activated receptor-gamma 93. Shindou H, Shimizu T (2009) Acyl-
agonist 15-deoxy-Delta(12,14)-prostaglandin J(2) CoA:lysophospholipid acyltransferases. J Biol
ameliorates experimental autoimmune encephalo- Chem 284:1–5
myelitis. J Immunol 168:2508–2515 94. Kono N, Arai H (2019) Platelet-activating factor
79. Niino M, Iwabuchi K, Kikuchi S, Ato M, acetylhydrolases: an overview and update. Biochim
Morohashi T, Ogata A, Tashiro K, Onoe K (2001) Biophys Acta Mol Cell Biol Lipids 1864(6):922–931
Amelioration of experimental autoimmune encepha- 95. Brochet B, Guinot P, Orgogozo JM, Confavreux C,
lomyelitis in C57BL/6 mice by an agonist of per- Rumbach L, Lavergne V (1995) Double blind pla-
oxisome proliferator- activated receptor-gamma. cebo controlled multicentre study of ginkgolide B in
J Neuroimmunol 116:40–48 treatment of acute exacerbations of multiple sclero-
80. Radmark O, Werz O, Steinhilber D, Samuelsson B sis. The Ginkgolide Study Group in multiple sclero-
(2015) 5-Lipoxygenase, a key enzyme for leukot- sis. J Neurol Neurosurg Psychiatry 58:360–362
riene biosynthesis in health and disease. Biochim 96. Callea L, Arese M, Orlandini A, Bargnani C, Priori
Biophys Acta 1851:331–339 A, Bussolino F (1999) Platelet activating factor
81. Singh NK, Rao GN (2018) Emerging role of is elevated in cerebral spinal fluid and plasma of
12/15-Lipoxygenase (ALOX15) in human patholo- patients with relapsing-remitting multiple sclerosis.
gies. Prog Lipid Res 73:28–45 J Neuroimmunol 94:212–221
82. Serhan CN (2017) Treating inflammation and infec- 97. Lock C, Hermans G, Pedotti R, Brendolan A, Schadt
tion in the 21st century: new hints from decoding E, Garren H, Langer-Gould A, Strober S, Cannella
resolution mediators and mechanisms. FASEB B, Allard J, Klonowski P, Austin A, Lad N, Kaminski
J 31:1273–1288 N, Galli SJ, Oksenberg JR, Raine CS, Heller R,
83. Serhan CN, Chiang N, Dalli J (2018) New pro- Steinman L (2002) Gene-microarray analysis of
resolving n-3 mediators bridge resolution of infec- multiple sclerosis lesions yields new targets vali-
tious inflammation to tissue regeneration. Mol Asp dated in autoimmune encephalomyelitis. Nat Med
Med 64:1–17 8:500–508
84. Serhan CN, Levy BD (2018) Resolvins in inflamma- 98. Osoegawa M, Miyagishi R, Ochi H, Nakamura I,
tion: emergence of the pro-resolving superfamily of Niino M, Kikuchi S, Murai H, Fukazawa T, Minohara
mediators. J Clin Invest 128:2657–2669 M, Tashiro K, Kira J (2005) Platelet-activating
13 Systematic Understanding of Bioactive Lipids in Neuro-Immune Interactions: Lessons from an Animal… 147
factor receptor gene polymorphism in Japanese 113. Tokumura A, Fukuzawa K, Akamatsu Y, Yamada
patients with multiple sclerosis. J Neuroimmunol S, Suzuki T, Tsukatani H (1978) Identification of
161:195–198 vasopressor phospholipid in crude soybean lecithin.
99. Bellizzi MJ, Geathers JS, Allan KC, Gelbard HA Lipids 13:468–472
(2016) Platelet-activating factor receptors mediate 114. Schmitz K, Brunkhorst R, De Bruin N, Mayer CA,
excitatory postsynaptic hippocampal injury in exper- Haussler A, Ferreiros N, Schiffmann S, Parnham
imental autoimmune Encephalomyelitis. J Neurosci MJ, Tunaru S, Chun J, Offermanns S, Foerch C,
36:1336–1346 Scholich K, Vogt J, Wicker S, Lotsch J, Geisslinger
100. Rodrigues DH, Lacerda-Queiroz N, De Miranda AS, G, Tegeder I (2017) Dysregulation of lysophospha-
Fagundes CT, Campos RD, Arantes RE, Vilela Mde tidic acids in multiple sclerosis and autoimmune
C, Rachid MA, Teixeira MM, Teixeira AL (2011) encephalomyelitis. Acta Neuropathol Commun 5:42
Absence of PAF receptor alters cellular infiltrate 115. Pollinger B, Krishnamoorthy G, Berer K, Lassmann
but not rolling and adhesion of leukocytes in experi- H, Bosl MR, Dunn R, Domingues HS, Holz A,
mental autoimmune encephalomyelitis. Brain Res Kurschus FC, Wekerle H (2009) Spontaneous
1385:298–306 relapsing-remitting EAE in the SJL/J mouse: MOG-
101. Kihara Y, Maceyka M, Spiegel S, Chun J (2014b) reactive transgenic T cells recruit endogenous MOG-
Lysophospholipid receptor nomenclature review: specific B cells. J Exp Med 206:1303–1316
IUPHAR review 8. Br J Pharmacol 171:3575–3594 116. Balood M, Zahednasab H, Siroos B, Mesbah-Namin
102. Fahy E, Sud M, Cotter D, Subramaniam S (2007) SA, Torbati S, Harirchian MH (2014) Elevated
LIPID MAPS online tools for lipid research. Nucleic serum levels of lysophosphatidic acid in patients
Acids Res 35:W606–W612 with multiple sclerosis. Hum Immunol 75:411–413
103. Alexander SP, Kelly E, Marrion NV, Peters JA, 117. Jiang D, Ju W, Wu X, Zhan X (2018) Elevated
Faccenda E, Harding SD, Pawson AJ, Sharman lysophosphatidic acid levels in the serum and cere-
JL, Southan C, Buneman OP, Cidlowski JA, brospinal fluid in patients with multiple sclerosis:
Christopoulos A, Davenport AP, Fabbro D, Spedding therapeutic response and clinical implication. Neurol
M, Striessnig J, Davies JA, Collaborators C (2017) Res 40:335–339
The concise guide to pharmacology 2017/18: over- 118. Zahednasab H, Balood M, Harirchian MH,
view. Br J Pharmacol 174(Suppl 1):S1–S16 Mesbah-Namin SA, Rahimian N, Siroos B (2014)
104. Shindou H, Hishikawa D, Harayama T, Yuki K, Increased autotaxin activity in multiple sclerosis.
Shimizu T (2009) Recent progress on acyl CoA: J Neuroimmunol 273:120–123
lysophospholipid acyltransferase research. J Lipid 119. Fujiwara Y (2008) Cyclic phosphatidic acid - a
Res 50(Suppl):S46–S51 unique bioactive phospholipid. Biochim Biophys
105. Aikawa S, Hashimoto T, Kano K, Aoki J (2015) Acta 1781:519–524
Lysophosphatidic acid as a lipid mediator with mul- 120. Yamamoto S, Yamashina K, Ishikawa M, Gotoh M,
tiple biological actions. J Biochem 157:81–89 Yagishita S, Iwasa K, Maruyama K, Murakami-
106. Nishimasu H, Ishitani R, Aoki J, Nureki O (2012) Murofushi K, Yoshikawa K (2017) Protective and
A 3D view of autotaxin. Trends Pharmacol Sci therapeutic role of 2-carba-cyclic phosphatidic acid in
33:138–145 demyelinating disease. J Neuroinflammation 14:142
107. Aoki J, Nagai Y, Hosono H, Inoue K, Arai H (2002) 121. Maceyka M, Harikumar KB, Milstien S, Spiegel S
Structure and function of phosphatidylserine- (2012) Sphingosine-1-phosphate signaling and its
specific phospholipase A1. Biochim Biophys Acta role in disease. Trends Cell Biol 22:50–60
1582:26–32 122. Proia RL, Hla T (2015) Emerging biology of
108. Kihara Y, Mizuno H, Chun J (2015b) sphingosine- 1-phosphate: its role in pathogenesis
Lysophospholipid receptors in drug discovery. Exp and therapy. J Clin Invest 125:1379–1387
Cell Res 333:171–177 123. Chiba K (2005) FTY720, a new class of immuno-
109. Sheng X, Yung YC, Chen A, Chun J (2015) modulator, inhibits lymphocyte egress from sec-
Lysophosphatidic acid signalling in development. ondary lymphoid tissues and thymus by agonistic
Development 142:1390–1395 activity at sphingosine 1-phosphate receptors.
110. Yung YC, Stoddard NC, Chun J (2014) LPA recep- Pharmacol Ther 108:308–319
tor signaling: pharmacology, physiology, and patho- 124. Chiba K, Adachi K (2012) Discovery of fingolimod,
physiology. J Lipid Res 55:1192–1214 the sphingosine 1-phosphate receptor modulator and
111. Yung YC, Stoddard NC, Mirendil H, Chun J (2015) its application for the therapy of multiple sclerosis.
Lysophosphatidic acid signaling in the nervous sys- Future Med Chem 4:771–781
tem. Neuron 85:669–682 125. Chiba K, Hoshino Y, Suzuki C, Masubuchi Y,
112. Mizuno H, Kihara Y, Kussrow A, Chen A, Yanagawa Y, Ohtsuki M, Sasaki S, Fujita T (1996)
Ray M, Rivera R, Bornhop D, Chun J (2018) FTY720, a novel immunosuppressant possessing
Lysophospholipid G protein-coupled receptor bind- unique mechanisms. I. Prolongation of skin allograft
ing parameters as determined by backscattering survival and synergistic effect in combination with
interferometry (BSI). J Lipid Res 60:212–217 cyclosporine in rats. Transplant Proc 28:1056–1059
148 Y. Kihara
126. Mansoor M, Melendez AJ (2008) Recent trials for lymphocytes determines lymphocyte egress kinetics.
FTY720 (fingolimod): a new generation of immuno- J Exp Med 207:1475–1483
modulators structurally similar to sphingosine. Rev
136. Liu Y, Wada R, Yamashita T, Mi Y, Deng CX,
Recent Clin Trials 3:62–69 Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee
127. Brinkmann V, Davis MD, Heise CE, Albert R, MJ, Liu CH, Hla T, Spiegel S, Proia RL (2000) Edg-
Cottens S, Hof R, Bruns C, Prieschl E, Baumruker 1, the G protein-coupled receptor for sphingosine-
T, Hiestand P, Foster CA, Zollinger M, Lynch KR 1-phosphate, is essential for vascular maturation.
(2002) The immune modulator FTY720 targets J Clin Invest 106:951–961
sphingosine 1-phosphate receptors. J Biol Chem 137. Eken A, Duhen R, Singh AK, Fry M, Buckner JH,
277:21453–21457 Kita M, Bettelli E, Oukka M (2017) S1P1 deletion
128. Mandala S, Hajdu R, Bergstrom J, Quackenbush differentially affects TH17 and Regulatory T cells.
E, Xie J, Milligan J, Thornton R, Shei GJ, Card Sci Rep 7:12905
D, Keohane C, Rosenbach M, Hale J, Lynch CL, 138. Blaho VA, Galvani S, Engelbrecht E, Liu C,
Rupprecht K, Parsons W, Rosen H (2002) Alteration Swendeman SL, Kono M, Proia RL, Steinman L,
of lymphocyte trafficking by sphingosine-1- Han MH, Hla T (2015) HDL-bound sphingosine-1-
phosphate receptor agonists. Science 296:346–349 phosphate restrains lymphopoiesis and neuroinflam-
129. Paugh SW, Payne SG, Barbour SE, Milstien S, mation. Nature 523:342–346
Spiegel S (2003) The immunosuppressant FTY720 139. Foster CA, Howard LM, Schweitzer A, Persohn
is phosphorylated by sphingosine kinase type 2. E, Hiestand PC, Balatoni B, Reuschel R, Beerli C,
FEBS Lett 554:189–193 Schwartz M, Billich A (2007) Brain penetration of
130. Lo CG, Xu Y, Proia RL, Cyster JG (2005) Cyclical the oral immunomodulatory drug FTY720 and its
modulation of sphingosine-1-phosphate receptor 1 phosphorylation in the central nervous system dur-
surface expression during lymphocyte recirculation ing experimental autoimmune encephalomyelitis:
and relationship to lymphoid organ transit. J Exp consequences for mode of action in multiple sclero-
Med 201:291–301 sis. J Pharmacol Exp Ther 323:469–475
131. Matloubian M, Lo CG, Cinamon G, Lesneski MJ, 140. Choi JW, Gardell SE, Herr DR, Rivera R, Lee CW,
Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster Noguchi K, Teo ST, Yung YC, Lu M, Kennedy G,
JG (2004) Lymphocyte egress from thymus and Chun J (2011) FTY720 (fingolimod) efficacy in an
peripheral lymphoid organs is dependent on S1P animal model of multiple sclerosis requires astrocyte
receptor 1. Nature 427:355–360 sphingosine 1-phosphate receptor 1 (S1P1) modula-
132. Pappu R, Schwab SR, Cornelissen I, Pereira JP, tion. Proc Natl Acad Sci U S A 108:751–756
Regard JB, Xu Y, Camerer E, Zheng YW, Huang 141. Kappos L, Bar-Or A, Cree BAC, Fox RJ,
Y, Cyster JG, Coughlin SR (2007) Promotion of Giovannoni G, Gold R, Vermersch P, Arnold DL,
lymphocyte egress into blood and lymph by dis- Arnould S, Scherz T, Wolf C, Wallstrom E, Dahlke
tinct sources of sphingosine-1-phosphate. Science F, Investigators EC (2018) Siponimod versus pla-
316:295–298 cebo in secondary progressive multiple sclerosis
133. Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang (EXPAND): a double-blind, randomised, phase 3
Y, Cyster JG (2005) Lymphocyte sequestration study. Lancet 391:1263–1273
through S1P lyase inhibition and disruption of S1P 142. Cruz-Orengo L, Daniels BP, Dorsey D, Basak
gradients. Science 309:1735–1739 SA, Grajales-Reyes JG, Mccandless EE, Piccio
134. Garris CS, Wu L, Acharya S, Arac A, Blaho VA, L, Schmidt RE, Cross AH, Crosby SD, Klein RS
Huang Y, Moon BS, Axtell RC, Ho PP, Steinberg (2014) Enhanced sphingosine-1-phosphate receptor
GK, Lewis DB, Sobel RA, Han DK, Steinman L, 2 expression underlies female CNS autoimmunity
Snyder MP, Hla T, Han MH (2013) Defective sphin- susceptibility. J Clin Invest 124:2571–2584
gosine 1-phosphate receptor 1 (S1P1) phosphory- 143. Seyedsadr MS, Weinmann O, Amorim A, Ineichen
lation exacerbates TH17-mediated autoimmune BV, Egger M, Mirnajafi-Zadeh J, Becher B, Javan
neuroinflammation. Nat Immunol 14:1166–1172 M, Schwab ME (2018) Inactivation of sphingosine-
135. Thangada S, Khanna KM, Blaho VA, Oo ML, Im 1-phosphate receptor 2 (S1PR2) decreases demyelin-
DS, Guo C, Lefrancois L, Hla T (2010) Cell-surface ation and enhances remyelination in animal models
residence of sphingosine 1-phosphate receptor 1 on of multiple sclerosis. Neurobiol Dis 124:189–201
Role of Bioactive Sphingolipids
in Inflammation and Eye Diseases 14
Koushik Mondal and Nawajes Mandal
K. Mondal
Department of Ophthalmology, University of
Tennessee Health Science Center, UTHSC,
Memphis, TN, USA 14.1 Introduction
N. Mandal (*)
Department of Ophthalmology, University of Inflammation is a defensive mechanism of a host
Tennessee Health Science Center, UTHSC, organism against infectious agents and injury.
Memphis, TN, USA Inflammatory mechanisms represent a network
Anatomy and Neurobiology, University of Tennessee of complex processes requiring the involvement
Health Science Center, UTHSC, Memphis, TN, USA of different metabolic and signaling pathways to
e-mail: [email protected]
resolve damage to tissue or to fight against infec- cant roles in immune surveillance and
tion. However, inflammation may also be detri- inflammatory processes [10]. During
mental if it progresses out of control. Host complement-mediated targeted cell lysis, there is
organisms have evolved different signaling an initiation of strong opsonization of the foreign
mechanisms to respond appropriately against a pathogen or apoptotic cell/cellular compartment,
range of threats by utilizing specialized immune followed by induction of proinflammatory sig-
cells such as neutrophils, resident and recruited naling by anaphylatoxins, which lead to recruit-
macrophages, dendritic cells, and lymphocytes ment of macrophages and eventually phagocytosis
[57]. Host immune machinery is activated against of the pathogen through the formation of the
microbial pathogens and recognizes molecular membrane attack complex (MAC) [129]. Thus,
structures found in pathogens, known as the inflammasome-complement pathway elimi-
Pathogen-Associated Molecular Patterns nates the pathogen and clears and eliminates
(PAMPs) [80], whereas signals released from potential mediators of damage and injury. Acute
stressed and damaged host cells are known as and chronic inflammation can influence vascular
Damage Associated Molecular Patterns (DAMPs) permeability of the cell. Activation of proinflam-
[175]. Both PAMPs and DAMPs are recognized matory cytokines up-regulates selectins (e.g.,
by molecular structures on immune cells, known P-selectin) and integrin ligands, e.g., Vascular
as Pattern recognition receptors (PRR). The Toll- cell adhesion molecule 1 (VCAM-1) and
like receptors (TLR) are important class of PRR Intercellular adhesion molecule 1 (ICAM-1), on
[82] that recognize bacterial (pathogen) mem- the lumen of endothelial cells. These are sensed
brane lipopolysaccharides and viral RNA as well by selectin ligands, e.g., P-selectin glycoprotein
as endogenous molecules that are secreted from ligand 1 (PSGL1) and integrins, e.g., Lymphocyte
damaged or dying cells [126]. After activation, function-associated antigen 1 (LFA1) on the sur-
TLRs recruit downstream signal adaptor pro- face of leukocytes, and promote loosening of
teins, including Myeloid differentiation primary tight junctions between endothelial cells while
response 88 (MyD88) and TIR-domain contain- permitting transfer of solutes to peripheral tissues
ing adapter inducing interferon β (TRIF), which and leukocyte infiltration through the blood-brain
leads to activation of kinases, such as Inhibitor of barrier [127]. Inflammasome activation can also
kappa B (IkB) and Mitogen activated protein regulate synthesis of various eicosanoids, such as
kinase (MAPK), downstream transcription fac- prostaglandins (PGs), thromboxane, hydroxye-
tors, such as Nuclear factor kappa B (NF-kB), icosatetraenoic acid (HETEs) and leukotrienes
Activator protein-1 (AP-1), and interferon regu- [128]. In addition to inflammasome-mediated
latory factor family proteins. These factors can canonical activation of caspase-1-dependent mat-
stimulate transcription of several amplifiers and uration of proinflammatory cytokines IL-1β and
effectors [155]. Different types of cytokines, IL-18, caspase-1 independently activates cyto-
such as Tumor necrosis factor (TNF)-α, solic phospholipase A2 (cPLA2) that stimulates
Interleukin (IL)-1β and IL-6, and chemokines, eicosanoid synthesis. In this mechanism, there is
e.g., C-C motif chemokine ligand 2 (CCL2), formation of a membrane pore, which drives
C-X-C motif chemokine ligand 1 (CXCL1), and rapid Ca2+ influx. The influx of Ca2+ then acti-
C-X-C motif chemokine ligand 10 (CXCL10) act vates cPLA2 and generates arachidonic acid
as amplifiers and effector molecules in this mech- (AA) from membrane phospholipids. This ara-
anism. Another member of the innate immune chidonic acid is further converted to prostaglan-
sensor is the NOD-like receptor (NLR), which is dins and thromboxanes by cyclooxygenases- 1
a component of the inflammasome multiprotein (COX-1) and COX-2, and leukotrienes and
complex [125]. The third type of pattern recogni- HETEs are converted by lipoxygenases [163].
tion receptor is Retinoic acid inducible gene 1 The generation of eicosanoids is responsible for
(RIG1) [87]. Cooperative interactions of inflam- increasing vascular permeability and leucocyte
masomes and complement cascades play signifi- recruitment during diverse homeostatic and path-
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 151
ological processes [30]. Thus, during injury or research that reveals involvement of sphingolipid
disease, the immune cells become reactive and metabolites in inflammation and their role in ocu-
their PRRs are activated, which leads to genera- lar diseases.
tion of innate inflammatory mediators including
complement pathway, chemokines and cytokines,
and inflammatory enzymes. These proinflamma- 14.2.1 Ceramides
tory mediators stimulate immune cells to prolif-
erate, migrate and induce expression of adhesion Sphingolipid metabolism is regulated by cas-
molecules on endothelial cells, which promote cades of enzyme activation within different cel-
loosening of tight junctions and eventually infil- lular compartments wherein Cer occupies a
tration of immune cells leading to recovery from central position. [56, 109] Cer plays a structural
injury or infection from pathogens or otherwise role by regulating membrane properties and its
pathological changes in diseased state. permeability [161] leading to promotion of raft
fusion [46]. Cer- enriched platforms facilitate
clustering of receptor molecules and their ligands
14.2 Sphingolipid Metabolites [65]. This in turn helps the induction of apoptosis
and Inflammation by clustering of CD95/Fas death receptor ligand
[47]. In addition to its structural role, Cer acts as
Sphingolipids serve both structural and regula- a second messenger by activating a diverse set of
tory roles in eukaryotic cells [31, 56]. The sphin- kinases and phosphatases [132]. The de novo Cer
golipid metabolites Ceramide (Cer), biosynthesis pathway is an anabolic pathway,
Sphingosine-1-phosphate (S1P), Ceramide-1 which begins with condensation of serine and
phosphate (C1P), and Lactosylceramide (LacCer) palmitoyl-CoA catalyzed by Serine palmitoyl-
are the major signaling molecules regulating key transferase (SPT) in the endoplasmic reticulum
physiological functions and a variety of patho- (ER). The catabolic pathway of Cer generation
logical processes, mainly related to inflammatory occurs in the plasma membrane and lysosome via
responses or inflammation-associated diseases degradation of sphingomyelin (SM) to Cer and
[55]. Cer acts as a potent pro-inflammatory agent, phosphorylcholine by sphingomyelinase. The
whereas C1P and S1P can regulate either inflam- third pathway is the lysosomal salvage pathway
mation or participate in anti-inflammatory func- involving complex sphingolipids. Ceramide syn-
tions. LacCer plays a role as a signaling molecule thases play a significant role in the salvage path-
in inflammation-induced proliferation or angio- way, thereby bypassing the formation of
genesis. Immune cell mediated secretion of pro- dihydroceramide. The fourth pathway of Cer bio-
inflammatory cytokines stimulate inflammatory synthesis occurs in liver mitochondria. The first
sphingolipid metabolic enzymes to convert report of the involvement of Cer in the inflamma-
sphingomyelin (SM) into Cer. Cer is then con- tory process demonstrated intracellular induction
verted into either S1P or C1P or glycosphingo- of a proinflammatory cytokine, TNF-α, which
lipid. The schematic diagram of sphingolipid induced sphingomyelinase and, in turn, elevated
metabolism and the biosynthetic enzymes Cer [75, 94]. Sphingolipidomics and transcrip-
involved in this process has been presented in tomics studies revealed that lipopolysaccharide
Fig. 14.1. The conversion of different sphingo- (LPS) induces inflammation through TLR4 in
lipid metabolites varies with cell type. These macrophage cell lines by inducing an increase in
inflammatory sphingolipid mediators then induce Cer [29]. In macrophages, the LPS-induced
different types of inflammatory transcription fac- TLR4-mediated increase in de novo Cer biosyn-
tors (e.g., NFkB) or they may activate cyclooxy- thesis is necessary for autophagosome formation,
genase -2, leading to production of which could play a role in innate immunity [140].
pro-inflammatory prostaglandins. In this review, Production of Cer subsequently activates a
we provide an overview of the significant body of proinflammatory transcription factor, NFkB, the
152 K. Mondal and N. Mandal
Fig. 14.1 Sphingolipid Metabolites in Ocular Diseases. Sphingosine kinase isoenzymes (SPHKs) and for reverse
The name of the disorders are presented in red and reaction Sphingosine-1-phosphate phosphatase (S1PP)
enzymes responsible for these metabolites are presented plays its role. Addition of glucose moiety with Cer is
in blue. The de-novo Ceramide (Cer) biosynthesis is mediated by Glucosylceramide synthase (GCS).
mediated by Serine palmitoyltransferase (SPT). In sal- Lactosylceramide synthase (LCS) converts
vage pathway Ceramide synthase isoenzymes (CerSs) Glucosylceramide (GlcCer) to Lactosylceramide (LacCer)
plays role in formation of Cer from Sphingosine. and Lactosylceramidase (LCdase) converts LacCer to
Sphingomyelin phosphodiesterase isoenzymes (SMPDs) GlcCer.GM3 ganglioside formation is mediated by GM3
converts Sphingomyelin (SM) to Cer. Sphingomyelin syn- synthase. Complex Gangliosides are generated by other
thase (SmS) is the enzyme that converts Cer to carbohydrate moiety adding enzymes, which we dis-
SM. Ceramide kinase (CerK) and Ceramide-1-phosphate cussed in our earlier review. AMD Age-related macular
phosphatase (C1PP), respectively converts Cer to degeneration, DR Diabetic Retinopathy, DTS
Ceramide-1-phosphate or vice versa. Conversion of Dysfunctional Tear Syndrome, MS Multiple Sclerosis, RP
Sphingosine-1-phosphate (S1P) is mediated by Retinitis Pigmentosa
Interestingly, C1P can have a dual regulatory role peripheral blood mononuclear cells [54]. The
by serving as an intracellular second messenger bimodal behavior of C1P in pro- and anti-
to regulate cell survival and/or as an extracellular inflammation could provide a novel therapeutic
receptor ligand to stimulate chemotaxis [43]. In strategy for modulating inflammation associated
inflammation, CIP also behaves in a promiscuous with different diseases.
manner, either as a pro-inflammatory or anti-
inflammatory agent. The biosynthesis of C1P
takes place in the trans-Golgi network through 14.2.3 Sphingosine-1-P
phosphorylation of Cer by Ceramide kinase
(CerK). The role of C1P in inflammation was first To date, Sphingosine-1-phosphate (S1P) is the
reported in A549 lung adenocarcinoma cells, best-described mediator within the sphingolipid
where it stimulated release of arachidonic acid pathway. Unlike Cer, which promotes apoptosis,
(AA) and subsequent production of proinflam- S1P is responsible for suppressing apoptosis
matory eicosanoids [120]. C1P-mediated inflam- [146]. As an extracellular and intracellular mes-
mation is directly regulated by activation of senger, S1P plays diversified roles in inflamma-
cytosolic phospholipase-A2α (cPLA2α), an tion, cancer, atherosclerosis, autoimmunity, and
enzyme that releases AA from membrane phos- angiogenesis [153]. The diversified actions of
pholipids [121]. Further studies demonstrated S1P are mainly regulated by five widely expressed
that C1P-mediated activation of group IV cPLA2 S1P G-protein-coupled receptors, S1PR(1–5)
proinflammatory enzyme is chain length- [147]. S1P is produced by phosphorylation of
specific; C1P bearing an acyl chain of 6 carbon or free sphingosine by Sphingosine kinase isoen-
higher, activated cPLA2α in vitro, whereas CIP zymes 1 and 2 (SphK1 and SphK2). Topologically,
with a shorter acyl chain length (C2-C1P) was SphK1 is a cytosolic protein and may also be
unable to activate this proinflammatory enzyme localized at the plasma membrane and endocytic
[150, 166]. Interestingly, prostaglandin produc- membrane trafficking network, whereas SphK2
tion is a coordinated function of C1P and S1P, mainly resides in the nucleus, mitochondria or in
where S1P stimulates COX-2 activity and then the ER [53]. It has been well documented that
cPLA2α functions in synthesis of AA to produce S1P gradient in circulation plays major role in
prostaglandins [122]. The interaction of C1P in lymphocyte migration and trafficking [12].
proinflammation has also been proposed in some Usually, in the blood and lymph S1P level is
studies. It has been shown that, compared to wild higher than in tissues, which is important for
type mice, CerK knock-out mice generate maintaining vascular integrity. The maintenance
decreased level of proinflammatory cytokines, of circulating lymphocytes is balanced through
IL-6 and TNF-α in high fat diets and show nor- S1P; lymphocytes are attached through S1P with
mal insulin signaling [98]. These knock out ani- their receptors in the lymphoid organ to prevent
mals show higher expression of insulin receptors their egression into blood. In inflamed tissues,
and glucose transporter GLUT4 and decreased there is production of S1P by SphK1 in endothe-
signaling of MCP-1 in infiltrating macrophages lial cells. Simultaneously, S1PR1 is also acti-
of adipose tissue. However, the role of C1P in vated by inflammatory signals, which in turn lead
proinflammation is complex, as other studies to disbalance in circulating lymphocytes [133].
have demonstrated that C1P may also reduce The synthetic S1P analog FTY720 (Fingolimod)
inflammation. C1P was shown to inhibit tumor prevents egression of lymphocytes in circulation
necrosis factor (TNF)-converting enzyme or and accumulation in the lymph nodes [131].
TACE, which is the major metalloprotease for Thus, FTY720 acts as an immunosuppressive
cleaving pro-TNF to its active form that plays a drug in inflammation and autoimmune diseases.
role in inflammation [79]. Also, exogenous use of The role of S1P in inflammation varies with cell
C1P was shown to suppress production of IL-6, type. It plays an important role during activation
IL-8, TNF, and IL-1β in LPS treated human of mast cells and the subsequent development of
154 K. Mondal and N. Mandal
Krabbe’s disease [152]. Niemann-Pick is a lyso- Uveitis. In Intermediate Uveitis there is chronic
somal storage disease caused by a mutation in the inflammation, which affects the vitreous. It
acid sphingomyelinase gene leading to dysfunc- includes Pars planitis, Posterior cyclitis and
tion of sphingolipid signaling [81]. In Neimann- Hyalitis. During Posterior Uveitis inflammation
Pick Type A, ocular abnormalities range from affects retina, choroid and optic nerve. It could be
corneal opacification to retinal opacification with chronic and recurrent in nature. The disease
a macular cherry red spot [164]. Whereas in the named Chorioretinitis, Retinochoroiditis,
case of Neimann-Pick Type B, the ocular mani- Retinitis and Neuroretinitis are under the cate-
festation is mainly retinal with pathological fea- gory of Posterior Uveitis. Pathologically,
tures including macular halos and cherry-red Posterior Uveitis involves breakdown of blood-
maculae [96]. Optic nerve pallor and perimacular retinal barrier (BRB), whereas in other forms of
gray discoloration in Neimann-Pick Type C have uveitis do not [37]. Like Posterior uveitis pathol-
been observed, both clinically and histologically. ogy, in Experimental Autoimmune Uveitis
In knock-out mouse models of Neimann-Pick (EAU), there is an extensive breakdown of BRB
disease, there is enhancement of microglial activ- and release of retinal autoantigen [49]. The EAU
ity and upregulation of IL-1β from astrocytes follows classical example of organ-specific auto-
[15]. In Farber disease, there is accumulation of immune disease that resembles Posterior Uveitis
ceramide in the joints, liver, throat, CNS and ret- in humans [22]. In case of Panuveitis, the inflam-
ina due to deficiency of ceramidase [41]. The mation affects entire uvea. The inflammation
pathological changes are observed in retinal gan- associated with uveitis is due to infiltration of
glion cells with gross distention and inclusions both innate and adaptive immune cells [61].
[172]. Fabry disease is a deficiency of Using a murine model of uveitis, it has been con-
α-galactosidase A which leads to accumulation firmed that T cells are involved and that Th17 and
of globotriosylceramide. The most common ocu- Th1 play a significant role in the inflammatory
lar condition arising from this is cornea verticil- mechanism. Th17 and Th1 recruit different innate
late [144]. Overall, these observations suggest a effector molecules: Th17 recruits neutrophils and
role for sphingolipids in ocular inflammatory Th1 recruits monocytes; both cause tissue
diseases. destruction with independent mechanisms of
pathology, with proinflammatory cytokines play-
ing a major role [119]. Interestingly, FTY720, a
14.3.3 Sphingolipids structural analog of Sphingosine (Sph) and an
and Autoimmune Eye FDA-approved therapeutic drug for Multiple
Diseases sclerosis (MS), has been found to be effective in
a rat model of experimental autoimmune uveitis
Uveitis is an autoimmune eye disease where the [26]. The exact mechanism of FTY720 is still
uvea is pathologically affected. There is inflam- unknown, but it acts in a complex way on sphin-
mation of the uvea, which composes the middle golipid metabolism. Sphk2 phosphorylates
layer of eye including the iris, ciliary body and FTY720 to FTY720-P, which is a mimetic of S1P
choroid. There are different types of uveitis, and inactivates S1P receptor mediated signaling
which is classified according to International [91]. It also inhibits de novo ceramide synthesis
Uveitis Study Group (IUSG) Classification, and also acts to inhibit ceramide synthase
which includes Anterior Uveitis, Intermediate enzymes [23]. This same drug was earlier used in
Uveitis, Posterior Uveitis and Panuveitis [66]. experimental treatment of Vogt-Koyanagi-Harada
Anterior Uveitis is acute type and it is most com- (VKH) uveitis patients to suppress production of
mon kind of uveitis where anterior chamber is granulocyte monocyte colony stimulating factor
inflamed. It mainly affects the iris and is often by T cells [134]. The T cell clones (TCC) from
called as Iritis. Iridocyclitis and Anterior cyclitis aqueous humor (AH) or peripheral blood mono-
are also included in this category of Anterior nuclear cells (PBMCs) from VKH patients pro-
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 157
duced significantly higher level of duction of ceramide has been linked with types of
proinflammatory cytokines IL-6, IL-8 and IFN-γ neuroinflammation other than MS, including
in comparison with healthy donors. This finding those that are connected to NFkB regulated path-
suggests a role for sphingolipid in inflammation ways that cause blood brain barrier disruption,
and lymphocyte migration in uveitis. Recently, in vascular leakage, and lymphocyte migration with
a Wister rat model of endotoxin-induced uveitis upregulation of ICAM1, VCAM1 and selectin
(EIV), increased levels of proinflammatory cyto- [67]. Although Fingolimod is currently used in
kines IL-6 and TNF-α were noted in the aqueous the treatment of MS, one of the common side
humor [165]. Increased levels of ceramides effects of this drug is Fingolimod-associated
C24:0 and C24:1, and sphingomyelin C24:0 were macular edema (FAME) [90]. Retinal hemor-
also reported in the aqueous humor. In the retina, rhages and retinal vein occlusion can also occur
similar length carbon chain species of ceramide in Fingolimod treated patients. Although infor-
also have been noticed in EIV rats. Increased lev- mation pertaining to molecular mechanisms
els of proinflammatory transcription factor NFkB associated with FAME is still lacking, a possible
were also observed in the retina of EIV rats. mechanism could be disruption of cell-to-cell
These observations suggest that infiltration of and cell-to-matrix adhesion complexes in retinal
innate and adaptive immune cells induces inflam- vessels resulting in stress in vascular permeabil-
mation, which could be mediated through modu- ity and subsequent macular edema [97, 113].
lation of sphingolipid metabolites. Thus, While our current understanding of MS is incom-
sphingolipids may play a major role in uveitis plete, there appears to be a strong correlation
pathology. between MS related retinal degeneration and
Multiple Sclerosis is an autoimmune disease. ceramide-related inflammatory pathways.
Inflammation-related retinal atrophy is one of the
pathological features related to MS. Significant
loss of retinal ganglion cells and the presence of 14.3.4 Sphingolipids
human leukocyte antigen-DR positive cells in the and Degenerative Retinal
retina, with activation of microglia are character- Diseases
istic abnormalities associated with MS [48, 160].
In the central nervous system (CNS), oligoden- Progressive damage to the retina and death of
drocytes are the myelin forming cells, which are photoreceptors is a hallmark for degenerative
affected during MS by activation of glial cells retinal diseases, including Age-related macular
and infiltration of lymphocytes and macrophages, degeneration (AMD) and Retinitis Pigmentosa
leading to apoptosis of oligodendrocytes. (RP). AMD is associated with several pathologi-
Sphingolipids are the major component of myelin cal disorders, ranging from inflammation, mal-
sheath and there are multiple pathophysiological functioning of autophagy and chronic oxidative
roles of sphingolipids in MS. In MS patients, stress leading to degeneration of retinal pigment
increased levels of ceramide have been reported epithelium (RPE) and ultimately photoreceptor
in oligodendrocytes [141] in association with an death with vision loss [99, 123]. RPE is the pig-
increase in sphingosine in white matter [102]. In mented cell layer, which is attached to underlying
addition to NSMase- Ceramide upregulation in choroid and provides nourishment to overlying
MS, sphingosine kinase 1- S1P receptor signal- retinal visual cells. It also functions in phagocy-
ing regulates astroglial proliferation and gliosis tosis, secretion and immune modulation.
[168]. As S1P- S1P receptor1 is a main pathway Photoreceptor cells function in visual phototrans-
of lymphocyte egression in MS, application of duction and visual signal generation. There are
Fingolimod or FTY720, an immunosuppressive two types of photoreceptor cells in mammalian
S1P receptor agonist drug reduces lesion forma- retinas, rods and cones, along with second and
tion in MS patients [73]. Neutral third order neurons, bipolar and ganglion cells,
Sphingomyelinase (NSMase) activation and pro- respectively. During early stage of AMD there is
158 K. Mondal and N. Mandal
accumulation of extracellular deposits called dru- [151]. On the other hand, overexpression of
sen in the retina, between RPE and Bruch’s mem- Sphingomyelin phosphodiesterase 3 (SMPD3)
brane. Drusen formation is linked to chronic enhances Cer production, which in turn leads to
low-level inflammation and complement activa- enhancement of RPE cell death by increasing
tion during initial stages in the pathogenesis of inflammatory factors and stress [174]. In mouse
AMD [7, 69]. Activation and secretion of various models, it has been reported that cholesterol
cytokines and chemokines, e.g., IL-1β, IL-6, mediates activation of acid sphingomyelinase,
TNF-α, CXCL8 play a significant role in initia- which disrupts autophagy in RPE and leads to
tion of inflammation [83]. The later stage of dis- early onset macular degeneration [159]. Increases
ease progression is classified as either ‘Dry in Cer eventually promote inflammatory factors
AMD’ or ‘Wet AMD’. Dry AMD is limited to and oxidative stress, which prevent proper endo-
damage of the macula region of the retina caused somal recycling of complement regulatory pro-
by atrophy whereas wet AMD also includes both teins after complement attack and disrupt
macular atrophy as well as choroidal neovascu- endosomal biogenesis [156]. Aberrant endosomal
larization (CNV). Inflammation mediated by biogenesis mediates complement activation in
complement factor plays an important role in the RPE cells in murine model of macular degen-
AMD. In addition to this, genetic mutations asso- eration [74]. In Rd10 mouse models, inhibition
ciated with complement factor gene is among the of de novo Cer biosynthesis by myriocin lowers
major risk factors in AMD pathogenesis. One retinal Cer levels and restricts photoreceptor
such factor in AMD is the inheritable genetic death in RP [149]. Accumulation of POS
mutation, Y402H in complement factor H (CFH) increases oxidative stress and activates CFB,
[52, 76]. Other variants are present in C3, CFB, leading to AMD associated neovascularization
C2 genes, associated with susceptibility to AMD [157]. The inflammatory factor also activates
[8]. These mutations are associated with a reduc- complement factor C3 and aggravates AMD
tion of anti-inflammatory iC3b component and pathogenesis [105]. Ceramide induces retinal
an increase of proinflammatory cytokines TNF-α degeneration, whereas choroidal neovasculariza-
and IFN-γ [24]. RPE also plays role in autophagy tion (CNV) is promoted by administration of
by autophagic degradation of photoreceptor outer alpha-galactosylceramide into the vitreous cavity
segments (POS) in the process called heteroph- of C57BL/6 mice [58]. Similarly, S1P2 receptor
agy [70]. In aging, the function of RPE declines deficient mice do not develop neovascularization
and results in accumulation of POS, which even- in the murine model of ischemia driven retinopa-
tually forms lipofuscin in lysosomes leading to thy [142]. The blockage of S1P by sonepcizumab,
malfunctioning of lysosomes, generation of oxi- a humanized monoclonal antibody, also signifi-
dative stress and retinal inflammation [36]. cantly reduces CNV in mouse models [170]. In
Oxidative stress-induced Cer biosynthesis genes summary, sphingolipids appear to play a signifi-
are involved in photoreceptor cell death [13]. cant role in retinal degenerative diseases by
Increased Cer levels in RPE cells raises the level increasing inflammation, generating oxidative
of inflammatory factor and ROS, which leads to stress and deregulating lysosomal function in
mitochondrial permeabilization and activation of RPE and triggering photoreceptor cell death and/
caspase-3, followed by apoptosis [72]. Use of or neovascularization.
desipramine protects photoreceptor death by
reducing inflammatory factors and oxidative
stress augmented by Cer, as desipramine inhibits 14.3.5 Sphingolipids and Diabetic
sphingomyelinase’s ability to convert sphingo- Retinopathy
myelin to Cer [136]. Similarly, overexpression of
Acid ceramidase (ASAH1) in ARPE19 cells Diabetic retinopathy is a microvascular disease
(Human retinal pigment epithelial cell line) pro- affecting retinal vascular degeneration and defec-
tects from oxidative stress by reducing Cer level tive repair of retinal endothelial cells with persis-
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 159
14.3.7 Sphingolipids and Dry Eye bomian gland tear film as compare to the good
Syndrome quality individuals [118]. The sphingolipid
metabolites in meibomian gland tear film could
Dysfunctional tear syndrome (DTS), commonly serve as clinical signature of different types of
known as dry eye disease, is caused by tear defi- eye diseases.
ciency or excessive evaporation [19]. In addition
to this, ocular surface inflammation due to
increase in tear osmolarity plays a major role in 14.4 Summary and Conclusion
DTS [78]. The tear film performs diversified
functions, ranging from maintaining light refrac- This chapter summarizes our current understand-
tion, supplying the cornea with nutrients and ing of inflammation and its correlation with
oxygen, lubrication of the cornea and conjunc- sphingolipid metabolites in eye diseases.
tiva, and ocular surface protection against foreign Although ocular immune privilege protects the
materials [110]. There are three different layers eye and retina from inflammation, the modula-
in tear film composition: the carbohydrate-rich tion and accumulation of different sphingolipid
glycocalyx layer, the intermediate aqueous layer, metabolites can perturb the ocular anti-
and the superficial tear film lipid layer (TFLL) inflammatory environment and lead to ocular
[28]. Meibomian glands are the major source of pathology in different lysosomal storage disor-
TFLL lipids. Meibomian Gland Dysfunction ders, autoimmune diseases, age related macular
(MGD) is a complex multifactorial disorder arise degeneration and diabetic retinopathy, suggest-
from combination of five different pathophysio- ing an involvement of sphingolipid metabolites
logical mechanisms; these are eyelid inflamma- in maintaining homeostasis of the eye. Fig. 14.1
tion, conjunctival inflammation, corneal damage, shows a schematic diagram of sphingolipid
microbiological changes and dry eye disease metabolism and involvement of different sphin-
resulting from tear film instability [14]. Mass golipid metabolites in ocular diseases. The bioac-
spectrometry (MS) data from dry eye disease tive sphingolipid ceramide acts as a
patient reveals the role of sphingolipid in main- proinflammatory lipid, whereas C1P and S1P
taining TFLL integrity [78]. In patient meibo- have both pro- and anti-inflammatory functions.
mian samples, significant increases of SM levels LacCer, on the other hand, acts as an angiogenic
were observed compared to normal subjects. The sphingolipid and induces neovascularization. In
individual short chain GlcCer species were sig- ocular diseases, ceramides are found to be inflam-
nificantly increased in patient meibomian sam- matory factors for stimulating proinflammatory
ples. Whereas in case of meibomian cytokines and in some cases, proinflammatory
keratoconjunctivitis (MKC), increased levels of cytokines induce the formation of ceramide that
Cer were reported due to abnormal hyperkerati- may be ultimately responsible for retinal cell
nization [93]. Similarly, increased Cer levels dis- death. GlcCer, LacCer, and S1P have been found
rupt stability and elasticity of TFLL [11]. to be associated with the cross talk between
However, patients with chronic blepharitis had immune cells and endothelial cells that eventu-
been reported with decreased amount of cerebro- ally develop neovascularization in ‘wet AMD’
sides in their meibomian gland [106]. In animal and diabetic retinopathy. The loss of homeostasis
models, very long chain ceramides (acylce- in diseased conditions leads to stress in the endo-
ramide) in TFLL prevent dry eye disease, as plasmic reticulum, mitochondria, lysosomes and
transgene ELOVL1 mice developed corneal ultimately to activation of different proinflamma-
opacity with vascular invasion, accompanied by tory factors. A more complete understanding of
epidermalization of the cornea due to lower level sphingolipid metabolites and their role in inflam-
of acylceramide in epidermis and in the meibo- mation will help in our understanding of the eti-
mian gland [137]. Recently, our lab has reported ology and pathobiology of various eye diseases
increase in Cer/S1P ratio from poor quality mei- that have inflammatory links.
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 161
Acknowledgements This work was supported by 11. Arciniega JC, Uchiyama E, Butovich IA (2013)
National Eye Institute grants [EY022071, EY025256, Disruption and destabilization of meibomian lipid
EY021725], and grants from Foundation Fighting films caused by increasing amounts of ceramides
Blindness Inc., USA and Research to Prevent Blindness and cholesterol. Invest Ophthalmol Vis Sci 54:1352–
Inc., USA. The authors gratefully acknowledge the edito- 1360. https://doi.org/10.1167/iovs.12-10662
rial help received from Dr. Dianna A. Johnson, Emeritus 12. Baeyens A, Fang V, Chen C, Schwab SR (2015)
Professor, UTHSC and Richard C. Grambergs, UTHSC, Exit Strategies: S1P Signaling and T Cell
Memphis, TN. Migration. Trends Immunol 36:778–787. https://doi.
org/10.1016/j.it.2015.10.005
13. Barak A, Morse LS, Goldkorn T (2001) Ceramide:
a potential mediator of apoptosis in human retinal
References pigment epithelial cells. Invest Ophthalmol Vis Sci
42:247–254
1. Abcouwer SF (2013) Angiogenic factors and cyto- 14. Baudouin C et al (2016) Revisiting the vicious
kines in diabetic retinopathy. J Clin Cell Immunol circle of dry eye disease: a focus on the patho-
Suppl 1. https://doi.org/10.4172/2155-9899 physiology of meibomian gland dysfunction. Br
2. Adada MM, Orr-Gandy KA, Snider AJ, Canals J Ophthalmol 100:300–306. https://doi.org/10.1136/
D, Hannun YA, Obeid LM, Clarke CJ (2013) bjophthalmol-2015-307415
Sphingosine kinase 1 regulates tumor necrosis 15. Baudry M, Yao Y, Simmons D, Liu J, Bi X (2003)
factor-mediated RANTES induction through p38 Postnatal development of inflammation in a murine
mitogen-activated protein kinase but independently model of Niemann-Pick type C disease: immunohis-
of nuclear factor kappaB activation. J Biol Chem tochemical observations of microglia and astroglia.
288:27667–27679. https://doi.org/10.1074/jbc. Exp Neurol 184:887–903. https://doi.org/10.1016/
M113.489443 S0014-4886(03)00345-5
3. Aerts JM et al (2007) Pharmacological inhibition 16. Bhunia AK, Han H, Snowden A, Chatterjee S (1996)
of glucosylceramide synthase enhances insulin Lactosylceramide stimulates Ras-GTP loading,
sensitivity. Diabetes 56:1341–1349. https://doi. kinases (MEK, Raf), p44 mitogen-activated protein
org/10.2337/db06-1619 kinase, and c-fos expression in human aortic smooth
4. Aljohani AJ, Edwards G, Guerra Y, Dubovy S, muscle cells. J Biol Chem 271:10660–10666
Miller D, Lee RK, Bhattacharya SK (2014) Human 17. Bhunia AK, Han H, Snowden A, Chatterjee S (1997)
trabecular meshwork sphingolipid and ceramide Redox-regulated signaling by lactosylceramide in
profiles and potential latent fungal commensalism. the proliferation of human aortic smooth muscle
Invest Ophthalmol Vis Sci 55:3413–3422. https:// cells. J Biol Chem 272:15642–15649
doi.org/10.1167/iovs.13-13570 18. Blaho VA et al (2015) HDL-bound sphingosine-
5. Aljohani AJ, Munguba GC, Guerra Y, Lee RK, 1-phosphate restrains lymphopoiesis and neuro-
Bhattacharya SK (2013) Sphingolipids and cerami- inflammation. Nature 523:342–346. https://doi.
des in human aqueous humor. Mol Vis 19:1966–1984 org/10.1038/nature14462
6. Alvarez SE et al (2010) Sphingosine-1-phosphate is a 19. Bron AJ et al (2014) Rethinking dry eye dis-
missing cofactor for the E3 ubiquitin ligase TRAF2. ease: a perspective on clinical implications.
Nature 465:1084–1088. https://doi.org/10.1038/ Ocul Surf 12:S1–S31. https://doi.org/10.1016/j.
nature09128 jtos.2014.02.002
7. Anderson DH, Mullins RF, Hageman GS, Johnson 20. Busik JV, Mohr S, Grant MB (2008) Hyperglycemia-
LV (2002) A role for local inflammation in the for- induced reactive oxygen species toxicity to endo-
mation of drusen in the aging eye. Am J Ophthalmol thelial cells is dependent on paracrine mediators.
134:411–431 Diabetes 57:1952–1965. https://doi.org/10.2337/
8. Anderson DH et al (2010) The pivotal role of the db07-1520
complement system in aging and age-related macu- 21. Caspi RR (2010) A look at autoimmunity and
lar degeneration: hypothesis re-visited. Prog Retin inflammation in the eye. J Clin Invest 120:3073–
Eye Res 29:95–112. https://doi.org/10.1016/j. 3083. https://doi.org/10.1172/JCI42440
preteyeres.2009.11.003 22. Caspi RR (2011) Understanding autoimmune uveitis
9. Arai T, Bhunia AK, Chatterjee S, Bulkley GB (1998) through animal models. The Friedenwald Lecture.
Lactosylceramide stimulates human neutrophils to Invest Ophthalmol Vis Sci 52:1872–1879. https://
upregulate Mac-1, adhere to endothelium, and gen- doi.org/10.1167/iovs.10-6909
erate reactive oxygen metabolites in vitro. Circ Res 23. Chen H, Chan AY, Stone DU, Mandal NA (2014)
82:540–547 Beyond the cherry-red spot: Ocular manifestations
10. Arbore G, Kemper C (2016) A novel “complement- of sphingolipid-mediated neurodegenerative and
metabolism- inflammasome axis” as a key regu- inflammatory disorders. Surv Ophthalmol 59:64–76.
lator of immune cell effector function. Eur https://doi.org/10.1016/j.survophthal.2013.02.005
J Immunol 46:1563–1573. https://doi.org/10.1002/ 24. Chen M, Muckersie E, Robertson M, Forrester JV,
eji.201546131 Xu H (2008) Up-regulation of complement factor
162 K. Mondal and N. Mandal
B in retinal pigment epithelial cells is accompa- cell death in diabetic retinopathy. Diabetes 55:3573–
nied by complement activation in the aged retina. 3580. https://doi.org/10.2337/db06-0539
Exp Eye Res 87:543–550. https://doi.org/10.1016/j. 39. Fox TE et al (2007) Ceramide recruits and activates
exer.2008.09.005 protein kinase C zeta (PKC zeta) within structured
25. Cho YH, Lee CH, Kim SG (2003) Potentiation of membrane microdomains. J Biol Chem 282:12450–
lipopolysaccharide-inducible cyclooxygenase 2 12457. https://doi.org/10.1074/jbc.M700082200
expression by C2-ceramide via c-Jun N-terminal 40. Fujiwaki T, Yamaguchi S, Tasaka M, Takayanagi M,
kinase-mediated activation of CCAAT/enhancer Isobe M, Taketomi T (2004) Evaluation of sphingo-
binding protein beta in macrophages. Mol Pharmacol lipids in vitreous bodies from a patient with Gaucher
63:512–523 disease, using delayed extraction matrix-assisted
26. Commodaro AG, Peron JP, Lopes CT, Arslanian C, laser desorption ionization time-of-flight mass spec-
Belfort R Jr, Rizzo LV, Bueno V (2010) Evaluation trometry. J Chromatogr B Analyt Technol Biomed
of experimental autoimmune uveitis in mice treated Life Sci 806:47–51. https://doi.org/10.1016/j.
with FTY720. Invest Ophthalmol Vis Sci 51:2568– jchromb.2004.02.027
2574. https://doi.org/10.1167/iovs.09-4769 41. Gangoiti P et al (2010) Control of metabolism and
27. Cousins SW, Espinosa-Heidmann DG, Csaky KG signaling of simple bioactive sphingolipids: implica-
(2004) Monocyte activation in patients with age- tions in disease. Prog Lipid Res 49:316–334. https://
related macular degeneration: a biomarker of risk doi.org/10.1016/j.plipres.2010.02.004
for choroidal neovascularization? Arch Ophthalmol 42. Giltiay NV, Karakashian AA, Alimov AP, Ligthle
122:1013–1018. https://doi.org/10.1001/ S, Nikolova-Karakashian MN (2005) Ceramide-
archopht.122.7.1013 and ERK-dependent pathway for the activation of
28. Cwiklik L (2016) Tear film lipid layer: a molecular CCAAT/enhancer binding protein by interleukin-
level view. Biochim Biophys Acta 1858:2421–2430. 1beta in hepatocytes. J Lipid Res 46:2497–2505.
https://doi.org/10.1016/j.bbamem.2016.02.020 https://doi.org/10.1194/jlr.M500337-JLR200
29. Dennis EA et al (2010) A mouse macrophage lipi- 43. Gomez-Munoz A, Gangoiti P, Arana L, Ouro A,
dome. J Biol Chem 285:39976–39985. https://doi. Rivera IG, Ordonez M, Trueba M (2013) New
org/10.1074/jbc.M110.182915 insights on the role of ceramide 1-phosphate in
30. Dennis EA, Norris PC (2015) Eicosanoid storm inflammation. Biochim Biophys Acta 1831:1060–
in infection and inflammation. Nat Rev Immunol 1066. https://doi.org/10.1016/j.bbalip.2013.02.001
15:511–523. https://doi.org/10.1038/nri3859 44. Gomez-Munoz A, Presa N, Gomez-Larrauri A,
31. Dressler KA, Mathias S, Kolesnick RN (1992) Rivera IG, Trueba M, Ordonez M (2016) Control
Tumor necrosis factor-alpha activates the sphingo- of inflammatory responses by ceramide, sphingo-
myelin signal transduction pathway in a cell-free sine 1-phosphate and ceramide 1-phosphate. Prog
system. Science 255:1715–1718 Lipid Res 61:51–62. https://doi.org/10.1016/j.
32. Edwards AO, Ritter R 3rd, Abel KJ, Manning A, plipres.2015.09.002
Panhuysen C, Farrer LA (2005) Complement factor 45. Gong N, Wei H, Chowdhury SH, Chatterjee S (2004)
H polymorphism and age-related macular degenera- Lactosylceramide recruits PKCalpha/epsilon and
tion. Science 308:421–424. https://doi.org/10.1126/ phospholipase A2 to stimulate PECAM-1 expres-
science.1110189 sion in human monocytes and adhesion to endothe-
33. Edwards G, Aribindi K, Guerra Y, Bhattacharya SK lial cells. Proc Natl Acad Sci USA 101:6490–6495.
(2014) Sphingolipids and ceramides of mouse aque- https://doi.org/10.1073/pnas.0308684101
ous humor: comparative profiles from normotensive 46. Goni FM, Alonso A (2009) Effects of ceramide and
and hypertensive DBA/2J mice. Biochimie 105:99– other simple sphingolipids on membrane lateral
109. https://doi.org/10.1016/j.biochi.2014.06.019 structure. Biochim Biophys Acta 1788:169–177.
34. Emery JM, Green WR, Huff DS (1972) Krabbe’s https://doi.org/10.1016/j.bbamem.2008.09.002
disease. Histopathology and ultrastructure of the 47. Grassme H et al (2001) CD95 signaling via ceramide-
eye. Am J Ophthalmol 74:400–406 rich membrane rafts. J Biol Chem 276:20589–20596.
35. Erikson A, Wahlberg I (1985) Gaucher disease– https://doi.org/10.1074/jbc.M101207200
Norrbottnian type. Ocular abnormalities. Acta 48. Green AJ, McQuaid S, Hauser SL, Allen IV, Lyness
Ophthalmol (Copenh) 63:221–225 R (2010) Ocular pathology in multiple sclerosis:
36. Feeney-Burns L, Berman ER, Rothman H (1980) retinal atrophy and inflammation irrespective of
Lipofuscin of human retinal pigment epithelium. disease duration. Brain 133:1591–1601. https://doi.
Am J Ophthalmol 90:783–791 org/10.1093/brain/awq080
37. Forrester JV, Kuffova L, Dick AD (2018) 49. Greenwood J (1992) The blood-retinal barrier in
Autoimmunity, autoinflammation, and infection in experimental autoimmune uveoretinitis (EAU): a
uveitis. Am J Ophthalmol 189:77–85. https://doi. review. Curr Eye Res 11(Suppl):25–32
org/10.1016/j.ajo.2018.02.019 50. Gregory MS et al (2011) Opposing roles for mem-
38. Fox TE et al (2006) Diabetes alters sphingolipid brane bound and soluble Fas ligand in glaucoma-
metabolism in the retina: a potential mechanism of associated retinal ganglion cell death. PLoS
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 163
One 6:e17659. https://doi.org/10.1371/journal. 64. Howell GR, Walton DO, King BL, Libby RT, John
pone.0017659 SW (2011b) Datgan, a reusable software system for
51. Guemes A, Kosmorsky GS, Moodie DS, Clark B, facile interrogation and visualization of complex
Meisler D, Traboulsi EI (1998) Corneal opacities in transcription profiling data. BMC Genomics 12:429.
Gaucher disease. Am J Ophthalmol 126:833–835 https://doi.org/10.1186/1471-2164-12-429
52. Hageman GS et al (2005) A common haplotype in 65. Hueber AO, Bernard AM, Herincs Z, Couzinet A,
the complement regulatory gene factor H (HF1/ He HT (2002) An essential role for membrane rafts
CFH) predisposes individuals to age-related macular in the initiation of Fas/CD95-triggered cell death in
degeneration. Proc Natl Acad Sci USA 102:7227– mouse thymocytes. EMBO Rep 3:190–196. https://
7232. https://doi.org/10.1073/pnas.0501536102 doi.org/10.1093/embo-reports/kvf022
53. Hait NC, Maiti A (2017) The role of sphingosine- 66. Jabs DA, Nussenblatt RB, Rosenbaum JT,
1-phosphate and ceramide-1-phosphate in inflam- Standardization of Uveitis Nomenclature Working
mation and cancer. Mediat Inflamm 2017:4806541. G (2005) Standardization of uveitis nomencla-
https://doi.org/10.1155/2017/4806541 ture for reporting clinical data. Results of the
54. Hankins JL, Fox TE, Barth BM, Unrath KA, Kester First International Workshop. Am J Ophthalmol
M (2011) Exogenous ceramide-1-phosphate reduces 140:509–516
lipopolysaccharide (LPS)-mediated cytokine expres- 67. Jana A, Pahan K (2010) Sphingolipids in multiple
sion. J Biol Chem 286:44357–44366. https://doi. sclerosis. NeuroMolecular Med 12:351–361. https://
org/10.1074/jbc.M111.264010 doi.org/10.1007/s12017-010-8128-4
55. Hannun YA (1996) Functions of ceramide in 68. Jeyakumar M et al (2003) Central nervous system
coordinating cellular responses to stress. Science inflammation is a hallmark of pathogenesis in mouse
274:1855–1859 models of GM1 and GM2 gangliosidosis. Brain
56. Hannun YA, Obeid LM (2008) Principles of bio- 126:974–987
active lipid signalling: lessons from sphingolip- 69. Johnson LV, Leitner WP, Staples MK, Anderson DH
ids. Nat Rev Mol Cell Biol 9:139–150. https://doi. (2001) Complement activation and inflammatory
org/10.1038/nrm2329 processes in Drusen formation and age related mac-
57. Heneka MT, McManus RM, Latz E (2018) ular degeneration. Exp Eye Res 73:887–896. https://
Inflammasome signalling in brain function and neu- doi.org/10.1006/exer.2001.1094
rodegenerative disease. Nat Rev Neurosci 19:610– 70. Kaarniranta K et al (2013) Autophagy and heteroph-
621. https://doi.org/10.1038/s41583-018-0055-7 agy dysregulation leads to retinal pigment epithelium
58. Hijioka K, Sonoda KH, Tsutsumi-Miyahara C, dysfunction and development of age-related macular
Fujimoto T, Oshima Y, Taniguchi M, Ishibashi T degeneration. Autophagy 9:973–984. https://doi.
(2008) Investigation of the role of CD1d-restricted org/10.4161/auto.24546
invariant NKT cells in experimental choroi- 71. Kady NM et al (2018) ELOVL4-mediated produc-
dal neovascularization. Biochem Biophys Res tion of very long-chain ceramides stabilizes tight
Commun 374:38–43. https://doi.org/10.1016/j. junctions and prevents diabetes-induced retinal vas-
bbrc.2008.06.080 cular permeability. Diabetes 67:769–781. https://doi.
59. Hinze A, Stolzing A (2011) Differentiation of mouse org/10.2337/db17-1034
bone marrow derived stem cells toward microglia- 72. Kannan R, Jin M, Gamulescu MA, Hinton DR
like cells. BMC Cell Biol 12:35. https://doi. (2004) Ceramide-induced apoptosis: role of cata-
org/10.1186/1471-2121-12-35 lase and hepatocyte growth factor. Free Radic
60. Holland WL et al (2011) Lipid-induced insulin Biol Med 37:166–175. https://doi.org/10.1016/j.
resistance mediated by the proinflammatory receptor freeradbiomed.2004.04.011
TLR4 requires saturated fatty acid-induced ceramide 73. Kappos L et al (2006) Oral fingolimod (FTY720) for
biosynthesis in mice. J Clin Invest 121:1858–1870. relapsing multiple sclerosis. N Engl J Med 355:1124–
https://doi.org/10.1172/JCI43378 1140. https://doi.org/10.1056/NEJMoa052643
61. Horai R et al (2013) Breakdown of immune privi- 74. Kaur G et al (2018) Aberrant early endosome bio-
lege and spontaneous autoimmunity in mice express- genesis mediates complement activation in the
ing a transgenic T cell receptor specific for a retinal retinal pigment epithelium in models of macular
autoantigen. J Autoimmun 44:21–33. https://doi. degeneration. Proc Natl Acad Sci USA 115:9014–
org/10.1016/j.jaut.2013.06.003 9019. https://doi.org/10.1073/pnas.1805039115
62. Howell GR et al (2011a) Molecular clustering iden- 75. Kim MY, Linardic C, Obeid L, Hannun Y (1991)
tifies complement and endothelin induction as early Identification of sphingomyelin turnover as an effec-
events in a mouse model of glaucoma. J Clin Invest tor mechanism for the action of tumor necrosis fac-
121:1429–1444. https://doi.org/10.1172/JCI44646 tor alpha and gamma-interferon. Specific role in cell
63. Howell GR et al (2012) Radiation treatment inhibits differentiation. J Biol Chem 266:484–489
monocyte entry into the optic nerve head and pre- 76. Klein R, Peto T, Bird A, Vannewkirk MR (2004)
vents neuronal damage in a mouse model of glau- The epidemiology of age-related macular degen-
coma. J Clin Invest 122:1246–1261. https://doi. eration. Am J Ophthalmol 137:486–495. https://doi.
org/10.1172/JCI61135 org/10.1016/j.ajo.2003.11.069
164 K. Mondal and N. Mandal
77. Kolmakova A, Rajesh M, Zang D, Pili R, Chatterjee 90. Mandal P, Gupta A, Fusi-Rubiano W, Keane PA,
S (2009) VEGF recruits lactosylceramide to Yang Y (2017) Fingolimod: therapeutic mechanisms
induce endothelial cell adhesion molecule expres- and ocular adverse effects. Eye (Lond) 31:232–240.
sion and angiogenesis in vitro and in vivo. https://doi.org/10.1038/eye.2016.258
Glycoconj J 26:547–558. https://doi.org/10.1007/ 91. Mann H (2010) Oral cladribine and fingolimod
s10719-008-9206-9 for relapsing multiple sclerosis. N Engl J Med
78. Lam SM, Tong L, Yong SS, Li B, Chaurasia SS, Shui 362:1738.; author reply 1739–1740. https://doi.
G, Wenk MR (2011) Meibum lipid composition in org/10.1056/NEJMc1002550
Asians with dry eye disease. PLoS One 6:e24339. 92. Marathe S, Schissel SL, Yellin MJ, Beatini N,
https://doi.org/10.1371/journal.pone.0024339 Mintzer R, Williams KJ, Tabas I (1998) Human
79. Lamour NF, Wijesinghe DS, Mietla JA, Ward KE, vascular endothelial cells are a rich and regulatable
Stahelin RV, Chalfant CE (2011) Ceramide kinase source of secretory sphingomyelinase. Implications
regulates the production of tumor necrosis fac- for early atherogenesis and ceramide-mediated cell
tor alpha (TNFalpha) via inhibition of TNFalpha- signaling. J Biol Chem 273:4081–4088
converting enzyme. J Biol Chem 286:42808–42817. 93. Mathers WD, Lane JA (1998) Meibomian gland
https://doi.org/10.1074/jbc.M111.310169 lipids, evaporation, and tear film stability. Adv Exp
80. Lampron A, Elali A, Rivest S (2013) Innate immu- Med Biol 438:349–360
nity in the CNS: redefining the relationship between 94. Mathias S, Dressler KA, Kolesnick RN (1991)
the CNS and Its environment. Neuron 78:214–232. Characterization of a ceramide-activated protein
https://doi.org/10.1016/j.neuron.2013.04.005 kinase: stimulation by tumor necrosis factor alpha.
81. Ledesma MD, Prinetti A, Sonnino S, Schuchman Proc Natl Acad Sci USA 88:10009–10013
EH (2011) Brain pathology in Niemann Pick disease 95. Matsushima GK, Taniike M, Glimcher LH, Grusby
type A: insights from the acid sphingomyelinase MJ, Frelinger JA, Suzuki K, Ting JP (1994) Absence
knockout mice. J Neurochem 116:779–788. https:// of MHC class II molecules reduces CNS demy-
doi.org/10.1111/j.1471-4159.2010.07034.x elination, microglial/macrophage infiltration, and
82. Lehnardt S (2010) Innate immunity and neuro- twitching in murine globoid cell leukodystrophy.
inflammation in the CNS: the role of microglia in Cell 78:645–656
Toll-like receptor-mediated neuronal injury. Glia 96. McGovern MM, Wasserstein MP, Aron A, Desnick
58:253–263. https://doi.org/10.1002/glia.20928 RJ, Schuchman EH, Brodie SE (2004) Ocular
83. Lentsch AB, Ward PA (2000) Regulation manifestations of Niemann-Pick disease type
of inflammatory vascular damage. J Pathol B. Ophthalmology 111:1424–1427. https://doi.
190:343–348. https://doi.org/10.1002/ org/10.1016/j.ophtha.2003.10.034
(SICI)1096-9896(200002)190:3<343::AID- 97. McVerry BJ, Garcia JG (2004) Endothelial cell bar-
PATH522>3.0.CO;2-M rier regulation by sphingosine 1-phosphate. J Cell
84. LeVine SM, Brown DC (1997) IL-6 and TNFalpha Biochem 92:1075–1085. https://doi.org/10.1002/
expression in brains of twitcher, quaking and normal jcb.20088
mice. J Neuroimmunol 73:47–56 98. Mitsutake S, Date T, Yokota H, Sugiura M, Kohama
85. Li G, Veenstra AA, Talahalli RR, Wang X, Gubitosi- T, Igarashi Y (2012) Ceramide kinase deficiency
Klug RA, Sheibani N, Kern TS (2012) Marrow- improves diet-induced obesity and insulin resis-
derived cells regulate the development of early tance. FEBS Lett 586:1300–1305. https://doi.
diabetic retinopathy and tactile allodynia in mice. org/10.1016/j.febslet.2012.03.032
Diabetes 61:3294–3303. https://doi.org/10.2337/ 99. Mitter SK et al (2014) Dysregulated autophagy in
db11-1249 the RPE is associated with increased susceptibility
86. Liang J et al (2013) Sphingosine-1-phosphate to oxidative stress and AMD. Autophagy 10:1989–
links persistent STAT3 activation, chronic intes- 2005. https://doi.org/10.4161/auto.36184
tinal inflammation, and development of colitis- 100. Mohr S (2004) Potential new strategies to prevent
associated cancer. Cancer Cell 23:107–120. https:// the development of diabetic retinopathy. Expert
doi.org/10.1016/j.ccr.2012.11.013 Opin Investig Drugs 13:189–198. https://doi.
87. Loo YM, Gale M Jr (2011) Immune signaling by org/10.1517/13543784.13.3.189
RIG-I-like receptors. Immunity 34:680–692. https:// 101. Mohri I et al (2006) Prostaglandin D2-mediated
doi.org/10.1016/j.immuni.2011.05.003 microglia/astrocyte interaction enhances astro-
88. Luo C, Yang X, Kain AD, Powell DW, Kuehn MH, gliosis and demyelination in twitcher. J Neurosci
Tezel G (2010) Glaucomatous tissue stress and 26:4383–4393. https://doi.org/10.1523/
the regulation of immune response through glial JNEUROSCI.4531-05.2006
Toll-
like receptor signaling. Invest Ophthalmol 102. Moscatelli EA, Isaacson E (1969) Gas liquid chro-
Vis Sci 51:5697–5707. https://doi.org/10.1167/ matographic analysis of sphingosine bases in sphin-
iovs.10-5407 golipids of human normal and multiple sclerosis
89. Maceyka M, Spiegel S (2014) Sphingolipid metabo- cerebral white matter. Lipids 4:550–555
lites in inflammatory disease. Nature 510:58–67. 103. Nakamura H, Moriyama Y, Makiyama T, Emori
https://doi.org/10.1038/nature13475 S, Yamashita H, Yamazaki R, Murayama T (2013)
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 165
Lactosylceramide interacts with and activates astrocytes. Implications for astrogliosis follow-
cytosolic phospholipase A2alpha. J Biol Chem ing neurotrauma. J Biol Chem 280:13742–13751.
288:23264–23272. https://doi.org/10.1074/jbc. https://doi.org/10.1074/jbc.M411959200
M113.491431 117. Pannu R, Won JS, Khan M, Singh AK, Singh I (2004)
104. Nakaya-Onishi M, Suzuki A, Okamoto N, Fukada M A novel role of lactosylceramide in the regulation
(2000) Observations on time course changes of the of lipopolysaccharide/interferon-gamma-mediated
cherry red spot in a patient with Tay-Sachs disease. inducible nitric oxide synthase gene expression:
Br J Ophthalmol 84:1320–1321 implications for neuroinflammatory diseases.
105. Natoli R et al (2017) Retinal macrophages synthesize J Neurosci 24:5942–5954. https://doi.org/10.1523/
C3 and activate complement in AMD and in mod- JNEUROSCI.1271-04.2004
els of focal retinal degeneration. Invest Ophthalmol 118. Paranjpe V, Tan J, Nguyen J, Lee J, Allegood J,
Vis Sci 58:2977–2990. https://doi.org/10.1167/ Galor A, Mandal N (2018) Clinical signs of meibo-
iovs.17-21672 mian gland dysfunction (MGD) are associated with
106. Nicolaides N, Kaitaranta JK, Rawdah TN, Macy JI, changes in meibum sphingolipid composition. Ocul
Boswell FM 3rd, Smith RE (1981) Meibomian gland Surf. https://doi.org/10.1016/j.jtos.2018.12.006
studies: comparison of steer and human lipids. Invest 119. Perez VL, Caspi RR (2015) Immune mechanisms in
Ophthalmol Vis Sci 20:522–536 inflammatory and degenerative eye disease. Trends
107. Novgorodov SA et al (2016) Lactosylceramide con- Immunol 36:354–363. https://doi.org/10.1016/j.
tributes to mitochondrial dysfunction in diabetes. it.2015.04.003
J Lipid Res 57:546–562. https://doi.org/10.1194/jlr. 120. Pettus BJ, Bielawska A, Spiegel S, Roddy P, Hannun
M060061 YA, Chalfant CE (2003) Ceramide kinase mediates
108. Nussenblatt RB, Liu B, Wei L, Sen HN (2013) The cytokine- and calcium ionophore-induced arachi-
immunological basis of degenerative diseases of the donic acid release. J Biol Chem 278:38206–38213.
eye. Int Rev Immunol 32:97–112. https://doi.org/10. https://doi.org/10.1074/jbc.M304816200
3109/08830185.2012.740536 121. Pettus BJ et al (2004) Ceramide 1-phosphate is a
109. Ogretmen B (2018) Sphingolipid metabolism in can- direct activator of cytosolic phospholipase A2. J Biol
cer signalling and therapy. Nat Rev Cancer 18:33– Chem 279:11320–11326. https://doi.org/10.1074/
50. https://doi.org/10.1038/nrc.2017.96 jbc.M309262200
110. Ohashi Y, Dogru M, Tsubota K (2006) Laboratory 122. Pettus BJ et al (2005) The coordination of prosta-
findings in tear fluid analysis. Clin Chim Acta 369:17– glandin E2 production by sphingosine-1-phosphate
28. https://doi.org/10.1016/j.cca.2005.12.035 and ceramide-1-phosphate. Mol Pharmacol 68:330–
111. Olivera A (2008) Unraveling the complexi- 335. https://doi.org/10.1124/mol.104.008722
ties of sphingosine-1-phosphate function: the 123. Piippo N et al (2014) Decline in cellular clearance
mast cell model. Prostaglandins Other Lipid systems induces inflammasome signaling in human
Mediat 86:1–11. https://doi.org/10.1016/j. ARPE-19 cells. Biochim Biophys Acta 1843:3038–
prostaglandins.2008.02.005 3046. https://doi.org/10.1016/j.bbamcr.2014.09.015
112. Olivera A, Mizugishi K, Tikhonova A, Ciaccia 124. Rajesh M, Kolmakova A, Chatterjee S (2005) Novel
L, Odom S, Proia RL, Rivera J (2007) The sphin- role of lactosylceramide in vascular endothelial
gosine kinase-sphingosine-1-phosphate axis is a growth factor-mediated angiogenesis in human
determinant of mast cell function and anaphylaxis. endothelial cells. Circ Res 97:796–804. https://doi.
Immunity 26:287–297. https://doi.org/10.1016/j. org/10.1161/01.RES.0000185327.45463.A8
immuni.2007.02.008 125. Ransohoff RM, Brown MA (2012) Innate immu-
113. Oo ML et al (2011) Engagement of S1P(1)- nity in the central nervous system. J Clin Invest
degradative mechanisms leads to vascular leak in 122:1164–1171. https://doi.org/10.1172/JCI58644
mice. J Clin Invest 121:2290–2300. https://doi. 126. Ransohoff RM, Engelhardt B (2012) The anatomical
org/10.1172/JCI45403 and cellular basis of immune surveillance in the cen-
114. Opreanu M, Lydic TA, Reid GE, McSorley KM, tral nervous system. Nat Rev Immunol 12:623–635.
Esselman WJ, Busik JV (2010) Inhibition of cyto- https://doi.org/10.1038/nri3265
kine signaling in human retinal endothelial cells 127. Ransohoff RM, Kivisakk P, Kidd G (2003) Three
through downregulation of sphingomyelinases by or more routes for leukocyte migration into the cen-
docosahexaenoic acid. Invest Ophthalmol Vis Sci tral nervous system. Nat Rev Immunol 3:569–581.
51:3253–3263. https://doi.org/10.1167/iovs.09-4731 https://doi.org/10.1038/nri1130
115. Opreanu M et al (2011) The unconventional role of 128. Rathinam VA, Fitzgerald KA (2016) Inflammasome
acid sphingomyelinase in regulation of retinal micro- complexes: emerging mechanisms and effector func-
angiopathy in diabetic human and animal models. tions. Cell 165:792–800. https://doi.org/10.1016/j.
Diabetes 60:2370–2378. https://doi.org/10.2337/ cell.2016.03.046
db10-0550 129. Ricklin D, Hajishengallis G, Yang K, Lambris JD
116. Pannu R, Singh AK, Singh I (2005) A novel role of (2010) Complement: a key system for immune sur-
lactosylceramide in the regulation of tumor necrosis veillance and homeostasis. Nat Immunol 11:785–
factor alpha-mediated proliferation of rat primary 797. https://doi.org/10.1038/ni.1923
166 K. Mondal and N. Mandal
130. Rivera J, Proia RL, Olivera A (2008) The alliance 144. Sodi A, Ioannidis AS, Mehta A, Davey C, Beck
of sphingosine-1-phosphate and its receptors in M, Pitz S (2007) Ocular manifestations of Fabry’s
immunity. Nat Rev Immunol 8:753–763. https://doi. disease: data from the Fabry Outcome Survey. Br
org/10.1038/nri2400 J Ophthalmol 91:210–214. https://doi.org/10.1136/
131. Rosen H, Sanna G, Alfonso C (2003) Egress: a bjo.2006.100602
receptor-regulated step in lymphocyte trafficking. 145. Soulet D, Rivest S (2008) Bone-marrow-
Immunol Rev 195:160–177 derived microglia: myth or reality? Curr Opin
132. Ruvolo PP (2003) Intracellular signal transduction Pharmacol 8:508–518. https://doi.org/10.1016/j.
pathways activated by ceramide and its metabolites. coph.2008.04.002
Pharmacol Res 47:383–392 146. Spiegel S, Milstien S (2002) Sphingosine
133. Saba JD (2015) A B cell-dependent mechanism 1-phosphate, a key cell signaling molecule. J Biol
restrains T cell transendothelial migration. Nat Med Chem 277:25851–25854. https://doi.org/10.1074/
21:424–426. https://doi.org/10.1038/nm.3858 jbc.R200007200
134. Sakaguchi M, Sugita S, Sagawa K, Itoh K, Mochizuki 147. Spiegel S, Milstien S (2003) Sphingosine-1-
M (1998) Cytokine production by T cells infiltrat- phosphate: an enigmatic signalling lipid. Nat Rev
ing in the eye of uveitis patients. Jpn J Ophthalmol Mol Cell Biol 4:397–407. https://doi.org/10.1038/
42:262–268 nrm1103
135. Sandhoff K, Harzer K (2013) Gangliosides and gan- 148. Streilein JW (2003) Ocular immune privilege: the
gliosidoses: principles of molecular and metabolic eye takes a dim but practical view of immunity and
pathogenesis. J Neurosci 33:10195–10208. https:// inflammation. J Leukoc Biol 74:179–185
doi.org/10.1523/JNEUROSCI.0822-13.2013 149. Strettoi E, Gargini C, Novelli E, Sala G, Piano I,
136. Sanvicens N, Cotter TG (2006) Ceramide is the key Gasco P, Ghidoni R (2010) Inhibition of ceramide
mediator of oxidative stress-induced apoptosis in reti- biosynthesis preserves photoreceptor structure and
nal photoreceptor cells. J Neurochem 98:1432–1444. function in a mouse model of retinitis pigmentosa.
https://doi.org/10.1111/j.1471-4159.2006.03977.x Proc Natl Acad Sci USA 107:18706–18711. https://
137. Sassa T, Tadaki M, Kiyonari H, Kihara A (2018) Very doi.org/10.1073/pnas.1007644107
long-chain tear film lipids produced by fatty acid 150. Subramanian P, Stahelin RV, Szulc Z, Bielawska A,
elongase ELOVL1 prevent dry eye disease in mice. Cho W, Chalfant CE (2005) Ceramide 1-phosphate
FASEB J 32:2966–2978. https://doi.org/10.1096/ acts as a positive allosteric activator of group IVA
fj.201700947R cytosolic phospholipase A2 alpha and enhances the
138. Schroder S, Palinski W, Schmid-Schonbein GW interaction of the enzyme with phosphatidylcho-
(1991) Activated monocytes and granulocytes, cap- line. J Biol Chem 280:17601–17607. https://doi.
illary nonperfusion, and neovascularization in dia- org/10.1074/jbc.M414173200
betic retinopathy. Am J Pathol 139:81–100 151. Sugano E et al (2018) Overexpression of acid-
139. Schutze S, Potthoff K, Machleidt T, Berkovic D, ceramidase (ASAH1) protects retinal cells
Wiegmann K, Kronke M (1992) TNF activates (ARPE19) from oxidative stress. J Lipid Res. https://
NF-kappa B by phosphatidylcholine-specific phos- doi.org/10.1194/jlr.M082198
pholipase C-induced “acidic” sphingomyelin break- 152. Suzuki K (2003) Globoid cell leukodystrophy
down. Cell 71:765–776 (Krabbe’s disease): update. J Child Neurol 18:595–
140. Sims K et al (2010) Kdo2-lipid A, a TLR4-specific 603. https://doi.org/10.1177/0883073803018009020
agonist, induces de novo sphingolipid biosynthesis 1
in RAW264.7 macrophages, which is essential for 153. Takabe K, Paugh SW, Milstien S, Spiegel S (2008)
induction of autophagy. J Biol Chem 285:38568– “Inside-out” signaling of sphingosine-1-phosphate:
38579. https://doi.org/10.1074/jbc.M110.170621 therapeutic targets. Pharmacol Rev 60:181–195.
141. Singh I, Pahan K, Khan M, Singh AK (1998) https://doi.org/10.1124/pr.107.07113
Cytokine-mediated induction of ceramide produc- 154. Takeshita H et al (2012) Sphingosine 1-phosphate
tion is redox-sensitive. Implications to proinflamma- (S1P)/S1P receptor 1 signaling regulates receptor
tory cytokine-mediated apoptosis in demyelinating activator of NF-kappaB ligand (RANKL) e xpression
diseases. J Biol Chem 273:20354–20362 in rheumatoid arthritis. Biochem Biophys Res
142. Skoura A, Sanchez T, Claffey K, Mandala SM, Proia Commun 419:154–159. https://doi.org/10.1016/j.
RL, Hla T (2007) Essential role of sphingosine bbrc.2012.01.103
1-phosphate receptor 2 in pathological angiogenesis 155. Takeuchi O, Akira S (2010) Pattern recognition
of the mouse retina. J Clin Invest 117:2506–2516. receptors and inflammation. Cell 140:805–820.
https://doi.org/10.1172/JCI31123 https://doi.org/10.1016/j.cell.2010.01.022
143. Snider AJ, Kawamori T, Bradshaw SG, Orr KA, 156.
Tan LX, Toops KA, Lakkaraju A (2016)
Gilkeson GS, Hannun YA, Obeid LM (2009) A role Protective responses to sublytic complement in
for sphingosine kinase 1 in dextran sulfate sodium- the retinal pigment epithelium. Proc Natl Acad
induced colitis. FASEB J 23:143–152. https://doi. Sci USA 113:8789–8794. https://doi.org/10.1073/
org/10.1096/fj.08-118109 pnas.1523061113
14 Role of Bioactive Sphingolipids in Inflammation and Eye Diseases 167
157. Tanaka K, Nakayama T, Mori R, Sato N, Kawamura dodecane delivery system to determine lipid-specific
A, Yuzawa M (2014) Associations of comple- effects. J Lipid Res 50:1986–1995. https://doi.
ment factor B and complement component 2 org/10.1194/jlr.M800367-JLR200
genotypes with subtypes of polypoidal choroidal 167. Wu YP, McMahon EJ, Matsuda J, Suzuki K,
vasculopathy. BMC Ophthalmol 14:83. https://doi. Matsushima GK, Suzuki K (2001) Expression of
org/10.1186/1471-2415-14-83 immune-related molecules is downregulated in
158. Tikhonenko M et al (2010) Remodeling of reti- twitcher mice following bone marrow transplanta-
nal Fatty acids in an animal model of diabetes: a tion. J Neuropathol Exp Neurol 60:1062–1074
decrease in long-chain polyunsaturated fatty acids 168. Wu YP, Mizugishi K, Bektas M, Sandhoff R, Proia
is associated with a decrease in fatty acid elongases RL (2008) Sphingosine kinase 1/S1P receptor sig-
Elovl2 and Elovl4. Diabetes 59:219–227. https://doi. naling axis controls glial proliferation in mice with
org/10.2337/db09-0728 Sandhoff disease. Hum Mol Genet 17:2257–2264.
159. Toops KA, Tan LX, Jiang Z, Radu RA, Lakkaraju https://doi.org/10.1093/hmg/ddn126
A (2015) Cholesterol-mediated activation of acid 169. Xiao Y, Zhong Y, Su H, Zhou Z, Chiao P, Zhong G
sphingomyelinase disrupts autophagy in the retinal (2005) NF-kappa B activation is not required for
pigment epithelium. Mol Biol Cell 26:1–14. https:// Chlamydia trachomatis inhibition of host epithelial
doi.org/10.1091/mbc.E14-05-1028 cell apoptosis. J Immunol 174:1701–1708
160. Trip SA et al (2005) Retinal nerve fiber layer axo- 170. Xie B, Shen J, Dong A, Rashid A, Stoller G,
nal loss and visual dysfunction in optic neuritis. Campochiaro PA (2009) Blockade of sphingosine-
Ann Neurol 58:383–391. https://doi.org/10.1002/ 1-phosphate reduces macrophage influx and
ana.20575 retinal and choroidal neovascularization. J Cell
161. van Blitterswijk WJ, van der Luit AH, Veldman RJ, Physiol 218:192–198. https://doi.org/10.1002/
Verheij M, Borst J (2003) Ceramide: second mes- jcp.21588
senger or modulator of membrane structure and 171. Yu RK, Tsai YT, Ariga T, Yanagisawa M (2011)
dynamics? Biochem J 369:199–211. https://doi. Structures, biosynthesis, and functions of ganglio-
org/10.1042/BJ20021528 sides–an overview. J Oleo Sci 60:537–544
162. Vandanmagsar B et al (2011) The NLRP3 inflamma- 172. Zarbin MA, Green WR, Moser HW, Morton
some instigates obesity-induced inflammation and SJ (1985) Farber’s disease. Light and electron
insulin resistance. Nat Med 17:179–188. https://doi. microscopic study of the eye. Arch Ophthalmol
org/10.1038/nm.2279 103:73–80
163. von Moltke J et al (2012) Rapid induction of inflam- 173. Zeng XX, Ng YK, Ling EA (2000) Neuronal and
matory lipid mediators by the inflammasome in vivo. microglial response in the retina of streptozotocin-
Nature 490:107–111. https://doi.org/10.1038/ induced diabetic rats. Vis Neurosci 17:463–471
nature11351 174. Zhu D, Sreekumar PG, Hinton DR, Kannan R
164. Walton DS, Robb RM, Crocker AC (1978) Ocular (2010) Expression and regulation of enzymes in
manifestations of group A Niemann-Pick disease. the ceramide metabolic pathway in human retinal
Am J Ophthalmol 85:174–180 pigment epithelial cells and their relevance to reti-
165. Wang HY, Wang Y, Zhang Y, Wang J, Xiong SY, Sun nal degeneration. Vis Res 50:643–651. https://doi.
Q (2018) Crosslink between lipids and acute uveitis: org/10.1016/j.visres.2009.09.002
a lipidomic analysis. Int J Ophthalmol 11:736–746. 175. Zhu H et al (2011) An efficient delivery of DAMPs
https://doi.org/10.18240/ijo.2018.05.05 on the cell surface by the unconventional secretion
166. Wijesinghe DS et al (2009) Chain length specific- pathway. Biochem Biophys Res Commun 404:790–
ity for activation of cPLA2alpha by C1P: use of the 795. https://doi.org/10.1016/j.bbrc.2010.12.061
Roles of Ceramides and Other
Sphingolipids in Immune Cell 15
Function and Inflammation
Sabrin Albeituni and Johnny Stiban
the SL recycling or salvage pathway. In this path- has been recently reported by Tidhar et al., that
way, complex SLs are catabolized to So which is 11 key amino acid residues might be critical in
subsequently N-acylated to ceramide. A more determining the acyl chain length specificity of
complex network of enzymes involving SMases, CerS2, CerS4, and CerS5 [21].
ceramidases, and CerS are implicated in the path- Functional CerS are important players for
way [13]. the wellbeing of cells. Loss of CerS has led to
In mammals CerS family consists of 6 abnormalities in mouse models (Table 15.1):
enzyme isoforms, named CerS1-6 [9, 14, 15]. CerS1 knockout mice suffered from reduced
Different CerS are distributed differentially in ganglioside levels and Purkinje cell loss leading
cells and tissues in the body (Fig. 15.1) [14, 16]. to impaired behavioral and motor development
All CerS transfer acyl-CoA of variable lengths [17, 22]. CerS2 knockout mice developed hepa-
to the amine group of a sphingoid base [2]. Each tocarcinoma and cerebral degeneration [28–30].
CerS has a higher specificity towards the transfer In addition, CerS3 loss resulted in disruption of
of acyl-CoA of a certain length [9]. CerS1 shows the skin barrier and spermatogenic arrest [18,
specificity towards the transfer of C18-CoA [17]; 31]. While CerS4 deficient mice developed
CerS2 is specific to C20–26 [16]; CerS3 acylates alopecia [32] due to destabilization in epider-
sphingoid bases with very long-chain fatty acyl mal stem/progenitor cell homeostasis [35].
CoAs (>26) [18]. CerS4 is specific for the trans- Interestingly, CerS5 loss led to improved adi-
fer of C18–22 acyl CoAs [19], whereas the pre- pose tissue health and function after consump-
ferred substrates for CerS5 and CerS6 are C14–18 tion of high fat diet [20]. Deficiency of CerS6
CoAs [20], and C14–16 CoAs [16], respectively. led to behavioral abnormalities and abnormal
Even though the specificity of CerS towards dif- clasping of the hind limbs in mice [34].
ferent CoAs has been well established, little is Ceramides are the simplest SLs composed of
known about the CerS regions that determine two hydrophobic tails and a simple rather than
such specificity, because the crystal structure of complex hydrophilic head, consisting of a
different CerS has not been solved. However, it hydroxyl group [36]. Because ceramides are
Fig. 15.1 Tissue distribution of CerS isoforms in mice. Tissue distribution of all CerS isoforms based on mRNA levels
[14, 16]. The percentage next to organ pictures represents the relative amount of each CerS isoform in that organ
172 S. Albeituni and J. Stiban
Table 15.1 Cellular abnormalities resulting from CerS [41]). In all, ceramides are not mere structural
deficiency
components in membranes. Along with So,
Deficient sphingosine 1-phosphate (S1P), C1P and lyso-
CerS
isoforom Abnormalitya Reference(s)
sphingomyelin, ceramides have been implicated
CerS1 Purkinje cell loss; motor Ginkel et al. as bioactive lipids [42] that act as second mes-
development impairment [17] and Zhao sengers and regulate cellular functions including
et al. [22] apoptosis and stress responses [43], tumor cell
CerS2 Delayed liver Jin et al. [23] death and metabolism [44], and cytokine signal-
regeneration after partial
ing and inflammation [45, 46]. Interestingly,
hepatectomy
Increased intestinal Chen et al. numerous evidences point to the ability of
permeability [24] ceramides to self-assemble into protein-
Pheochromocytoma Park et al. [25] permeable channels [47] in artificial membranes
Impaired neutrophil Barthelmes [48, 49], mitochondria [50–53] and lysosomes
migration et al. [26] [54] that are upstream to caspase-dependent
Enhanced liver Chen et al.
apoptotic cell death.
tumorigenesis [27]
Cerebral degeneration, Imgrund et al.
Despite the enormous progress in understand-
myelin sheath defects [28] and ing the effects of ceramides in regulating key
Pewzner-Jung events in cellular biology, their role in regulating
et al. [29, 30] immune cell function and inflammatory diseases
CerS3 Skin barrier deformation Jennemann
has only gained momentum in the last two
et al. [18]
Blocked spermatogenesis Rabionet et al.
decades. Herein, we discuss the main research
[31] findings describing the roles of ceramides in reg-
CerS4 Altered lipid Ebel et al. [32] ulating the function of various immune cells,
composition in skin; including dendritic cells (DC) and neutrophils,
alopecia and the modulation of T cell function and
CerS5 Improved glucose Gosejacob
homeostasis and adipose et al. [20]
macrophages in different disease settings. In
tissue health following addition, we will address the main findings high-
high fat dieta lighting ceramide function in various inflamma-
CerS6 Protection against the Scheffel et al. tory diseases.
development of colitisa [33]
Behavioral problems Ebel et al. [34]
a
Abnormality may not necessarily be detrimental to cells,
it may be beneficial, but still it is a change from normal
15.2 C
eramides and Other SLs
conditions in Immune Cell Function
in regulating DC function was initially associ- The study of ceramides in DC gained further
ated with induction of apoptosis by C2-ceramide attention due to the structural resemblance
[57]. Similarly, induction of DC apoptosis was between the toxic component of LPS, lipid A,
linked to increased accumulation of ceramides and ceramide (Fig. 15.2) [62]. It was suggested
in DC cultured with tumor supernatants, subse- that LPS mediates its function by mimicking
quently leading to down regulation of the fol- ceramides since, similar to LPS inducing DC
lowing survival signaling pathways, maturation, C2-ceramide reduces micropinocyto-
phosphatidylinositol 3 kinase (PI3K), Akt sis and antigen presentation to T cells by DC
kinase, Bcl-xL, and NF-κB [58]. However, [60]. However, while both LPS and C2-ceramide
induction of ceramide accumulation in DC by induce c-Jun N-terminal kinase (JNK), only LPS
factors that cause DC maturation including, activates extracellular signal-regulated kinases
CD40L, interleukin 1β (IL-1β), tumor necrosis (ERKs) and NF-κB. In addition, LPS stimulates
factor alpha (TNFα), and the gram-negative bac- the production of ceramides regardless of whether
terial endotoxin lipopolysaccharide (LPS) did macrophages are genetically responsive or unre-
not induce DC apoptosis [59, 60]. These contro- sponsive to LPS [63]. Therefore, this further sup-
versial findings were later reconciled by Franchi ported that LPS exerts its function by inducing
and colleagues demonstrating that ceramide- ceramide accumulation and not by interacting
induced cell death in DC is only exacerbated in with ceramide-producing enzymes or as a struc-
the absence of serum, while in the presence of tural mimic of ceramide.
serum and LPS, DC survival was achieved by Moreover, ceramides play important roles in
the action of cellular ceramidases that deacylate DC during viral infections. GalCer on epithelial
ceramides to So, thus, preventing ceramide cells binds the human immunodeficiency virus
accumulation and DC apoptosis [61]. (HIV)-1 envelope glycoproteins gp120 [64, 65]
Fig. 15.2 Structural similarity between Lipid A of structure to ceramides. The 2D structures were
LPS and ceramides. Two representative ceramides are obtained from PubChem and were redrawn to increase
included for comparison, the most abundant C16- resolution. PubChem CID: 9877306 (Lipid A),
ceramide and the phosphorylated C12-ceramide 1-phos- 71314646 (C16-ceramide) and 5283580 (C12-ceramide
phate. The blue triangle identifies the comparable 1-phosphate).
174 S. Albeituni and J. Stiban
and gp41 [66, 67]. In addition, GalCer is essen- neutrophil extracellular traps (NETs) in
tial in the formation of membrane lipid rafts that response to inflammatory stimuli are crucial for
allow for the internalization of HIV-1 through mounting an effective frontline immune
endocytosis and transcytosis [68]. Interestingly, response against invading microorganisms,
GalCer is present in monocyte-derived immature especially extracellular pathogens [72]. Since
DC and human primary DC isolated from the isolation of the free long-chain base So from
mucosa, suggesting that DC mediates HIV-1 human neutrophils [73], the role of ceramides in
transfer to its target cells through GalCer [69]. mediating and regulating neutrophil function
Ceramides also stabilized the membrane for has gained considerable traction. This was par-
measles virus entry in DC, as Dendritic Cell- ticularly due to early studies linking TNFα sig-
specific intercellular adhesion molecule-3-grab- naling, a potent inducer of superoxide and
bing non-integrin ligation on DC results in apoptosis in neutrophils [74], to ceramide func-
sphingomyelinase activation and subsequent tion [75]. In HL-60 human promyelocytic leu-
ceramide accumulation in DC exposed to either kemia cells, TNFα causes early sphingomyelin
mannan or measles virus [70]. hydrolysis and ceramide production [76]. In
The role of ceramides was further extended addition, in a cell-free system both sphingomy-
beyond viral entry in DC. It has been demon- elin content and ceramide concentration were
strated that local administration of a ceramide reported to be increased in response to TNFα
analog C8-ceramide causes the induction of DC [77]. It was therefore hypothesized that cerami-
maturation, secretion of the pro-inflammatory des might regulate neutrophil function in
cytokines IL-12p70 and TNFα, and enhanced response to TNFα. However, later studies have
virus-specific T cell responses in murine models proposed that ceramides activated a negative
of chronic lymphocytic choriomeningitis feedback loop to inhibit superoxide production
virus clone 13 and influenza virus [71]. in human neutrophils. It has been shown that
Collectively, these findings demonstrated ceramides not only did not mediate TNFα-
that DC apoptosis, maturation, and antigen pre- induced superoxide production in human neu-
senting capacity, are finely tuned by the action trophils [78] but also C2-ceramide inhibited the
of endogenous ceramides or treatment with 20:4 (n-6)-mediated superoxide formation in
exogenous ceramide analogs. In addition, human neutrophils [79]. C2-ceramide also inhib-
ceramides are key structural components of ited respiratory burst of N-formylmethionine-
lipid rafts in DC required for binding, uptake, leucyl-phenylalanine (fMLP)-stimulated
and internalization of viruses. These steps are adherent neutrophils [80–82]. Modulation of
key for initiating DC response allowing for viral neutrophil response to polarity in response to a
internalization, virus peptide processing, and chemotactic agent such as fMLP has been
presentation to T cells that subsequently kill shown to be dependent on neutral sphingomye-
virus-infected cells. Since these findings linase activity that converts sphingomyelin to
strongly suggest that ceramides are key effec- ceramide through the modulation of Rac1/2/
tors of DC function, it is important to note that RhoA GTPases [83]. These combined results
future studies elucidating the role of ceramides suggested that induction of ceramides might
on the various DC subsets (e.g. plasmacytoid delay the response of neutrophils to TNFα to
DC, migratory classical DC, tissue resident allow for neutrophil extravasation and migration
classical DC) could be of great therapeutic rele- prior to superoxide production.
vance in numerous immunologic diseases. Ceramides have also been implicated in the
modulation of phagocytosis and migration in
neutrophils. C2-ceramides reduced phagocytosis
15.2.2 Neutrophils of IgG-opsonized erythrocytes by fMLP-
activated neutrophils through the inhibition of
Neutrophil extravasation, migration, production MAP kinase activation and tyrosine phosphoryla-
of cytokines and superoxide, and formation of tion of ERK1 and ERK2 [84]; and by inhibiting
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 175
phospholipase D (PLD) function required for strated to be not only relevant in supporting the
phagocytosis [85, 86]. Similarly, in COS-1 mon- membrane structure but also as a transducer of
key kidney immortalized cells transfected with cellular signals. In vitro studies have revealed
FcγIIA receptor, inhibition of ceramide synthesis that LacCer enhances the upregulation of the
led to enhanced phagocytosis [87]. On the other integrin, CD11b, on neutrophil surface, inducing
hand, chemotaxis, phagocytosis and NETs for- neutrophil adherence to endothelium, and medi-
mation in fMLP-stimulated neutrophils were ating the production of ROS through activation
enhanced by the selective estrogen receptor of NADPH oxidase [96]. NADPH oxidase acti-
antagonist, tamoxifen, through induction of vation in neutrophils results from the association
ceramide accumulation, and subsequently pro- of LacCer in lipid rafts with the Src family
tein kinase C zeta (PKCζ) activation [88]. These kinase Lyn, and subsequent activation of PI3K,
conflicting findings might suggest the opposing mitogen-activated protein kinase (MAPK) and
roles of various ceramides on neutrophil PKC [97]. In addition, LacCer with long-chain
function. fatty acids (C24) are required for the coupling of
A more direct role of ceramides is also preva- Lyn to LacCer lipid rafts resulting in the produc-
lent in neutrophils. Ceramides are pro-apoptotic tion of superoxides and induction of migration
metabolites whose concentration in cells rises in neutrophils [98–100]. Association of Lyn with
prior to the execution of the apoptotic pathway. the LacCer containing C24-fatty acid chain is
Neutrophil apoptosis is mediated by C16- and necessary for the phagocytosis of mycobacteria
C24-ceramides via caspase activation. This is cor- by neutrophils. Interestingly, LacCer-enriched
related with the ability of granulocyte-macro- lipid rafts are modulated by pathogenic bacteria
phage colony-stimulating factor, a neutrophil to evade neutrophil-mediated killing. For
survival factor, to reduce the accumulation of instance, Mycobacterium tuberculosis prevented
ceramides in neutrophils [89]. During early neu- phagolysosome formation in neutrophils through
trophil apoptosis ceramide is generated by acid binding of bacterial mannose-capped lipoarabi-
SMase (aSMase). In aSMase −/− mice, neutro- nomannan to LacCer rafts in neutrophils [101].
phil apoptosis is delayed compared to WT mice In addition, inhibition of inflammation in neu-
[90]. In an anti-microbial setting, Pseudomonas trophils has also been associated with SL biology.
auroginosa release pyocyanin that induce reac- Ceramides inhibit immune cell responses by
tive oxygen species (ROS), which subsequently binding to CD300f inhibitory receptor in sepsis
activates mitochondrial SMase, therefore, [102], LPS-induced skin inflammation [103],
increasing mitochondrial ceramide levels and allergic responses [104], and experimental colitis
inducing cytochrome c release from mitochon- [105]. Interestingly, disruption of ceramide-
dria. This initiates cell death in neutrophils [91]. CD300f binding induces neutrophil infiltration in
In addition, the enzyme sphingomyelin synthase, sepsis and skin inflammation [102]. Ceramide
which mediates the transfer of choline phosphate metabolites have also been implicated in the
to ceramide from phosphatidylcholine, leads to modulation of neutrophil function. For instance,
the production of sphingomyelin and diacylglyc- chemotactic migration of neutrophils in response
erol (DAG) [92, 93] and mediates neutrophil to IL-8 and fMLP is inhibited by S1P, an
killing of fungus Cryptococcus neoformans [94]. immediate metabolite of ceramide [102].
Nevertheless, the aforementioned negative Moreover, IL-8 gene expression and secretion
regulation by ceramides was not reported upon was shown to be induced by S1P in lung H441
stimulation with the glycosphingolipid, lactosyl- epithelial cells [107]. Similarly, C1P dampens
ceramide (LacCer). In neutrophils, LacCer com- inflammation in a model of LPS-induced lung
pose more than two thirds of the glycolipid inflammation. Particularly, C16-C1P and synthetic
molecules in the plasma membrane and is mainly C8-C1P analog inhibit NF-κB activation in neu-
associated with a pro-inflammatory phenotype trophils and LPS-mediated IL-8 production
[95]. This large composition was later demon- [108].
176 S. Albeituni and J. Stiban
Recent studies have also demonstrated induction deficient mice fed with high-fat diet are protected
of PKR/JNK activation [142, 143]. from obesity, glucose intolerance, and insulin
Further investigations aimed to determine resistance suggesting that targeting CerS6 could
whether ceramides induce insulin resistance. be beneficial for the treatment of obesity and type
Analysis of muscle biopsies from insulin intoler- 2 diabetes [20, 156]. It is worth noting that the
ant obese individuals revealed an increased products of CerS5 (C16-ceramide) and CerS2
accumulation of ceramides [144–147]. In addi- (C24-ceramide) antagonized each other’s ability
tion, exogenous C2-ceramide induced apoptosis to form channels in mitochondrial outer mem-
in skeletal muscle myotubes, reducing the cell branes to induce apoptosis [157]. This may be
capacity to uptake glucose, a process that can be another mechanism by which different CerS
reversed by the CerS inhibitor, fumonisin B1 affect insulin resistance and tolerance
[148, 149]. In muscle cells, C2-ceramide also differentially.
mediated insulin resistance by inhibiting Rac Ceramide accumulation has been linked to
activation, thus reducing GLUT4 translocation inflammatory pathways mainly involving signal-
to the plasma membrane in response to insulin ing events downstream of TLR4. More specifi-
[150]. These studies suggested that targeting cally, Holland et al., was the first to provide a link
ceramide accumulation could protect muscles between lipotoxicity and inflammation in the
against insulin resistance. In line with this induction of insulin intolerance [158].
hypothesis, Bruce and coworkers demonstrated Particularly, saturated fatty acids induced de
that overexpression of sphingosine kinase 1 novo ceramide synthesis via TLR4 activation,
(SK1) led to reduced ceramide accumulation which also altered the metabolic program of
and insulin resistance in mice given high-fat diet skeletal muscle, inducing insulin resistance.
[151]. Inhibition of CerS and ceramide synthesis However, Galbo et al. showed that lipid-induced
with the S1P analog, FTY720 [152], also insulin resistance is a result of increased accumu-
reduced insulin tolerance in mice on high-fat lation of DAG and induced DAG-PKCε signal-
diet [153]. Furthermore, in obese rodents, block- ing rather than induced TLR4-ceramide pathway
ade of de novo ceramide synthesis with the SPT) [159]. These conclusions were based on the
inhibitor, myriocin, improved glucose tolerance observation that knockdown of TLR4 or the
[128, 154]. adaptor protein MyD88 prevented hepatic steato-
Since CerS are required for de novo ceramide sis in mice fed with a saturated fat diet through
generation, several studies have focused on the reduction of appetite but not hepatic insulin sig-
role of CerS in inducing insulin tolerance. naling. When mice were given saturated fat by
Overexpression of CerS1, CerS2, CerS4, CerS5, oral gavage, loss of TLR4 or MyD88 did not pro-
and CerS6 in L6 myotubes induced ceramide tect mice from hepatic insulin resistance.
production; however, none of the CerS was able Interestingly, another link between ceramides
to inhibit insulin signaling [139]. Given the and inflammatory pathways in insulin tolerance
opposing role of CerS2 and CerS6 in inflamma- involves stimulation of the Nod-like receptor
tion, current studies have been focusing on the pyrin domain-containing-3 (Nlrp3) inflamma-
role of these CerS in insulin resistance in vivo. some [160]. In obese mice, ceramides activated
CerS2 haploinsufficient mice have altered pat- Nlrp3 inflammasome and IL-1β secretion
terns of ceramide acylation, leading to reduced through caspase-1 activation in adipose tissue
levels of very-long-chain ceramides and increased macrophages in a Nlrp3-dependent manner. This
levels of long-chain C16-ceramide as a compensa- induction subsequently led to T cell activation.
tory mechanism leading to insulin resistance However, this study does not exclude the possi-
when fed with high-fat diet [155]. In agreement bility that inducers other than ceramides present
with these findings, CerS6 expression and levels in the diet induced caspase-1 activation. Further
of C16-ceramide are induced in the adipose tissue studies are required to establish whether cerami-
of obese humans. Moreover, CerS6- and CerS5- des are truly sensed by PRR in immune cells.
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 179
complex class II and reduced expression of IL-10 mune responses against myelin in the central ner-
and CD206,which are markers associated with vous system (CNS) [181]. In experimental
macrophage suppression [176]. autoimmune encephalomyelitis (EAE), de novo
Modulation of TAM function by ceramides synthesis of C16-ceramide by CerS6 is required for
has been also achieved in a mouse model of the production of pro-inflammatory TNFα and
hepatocellular cancer through the administration iNOS in macrophages in response to IFNγ [182].
of nanoliposome-loaded C6-ceramide (LipC6) Upregulation of CerS6 in EAE led to further specu-
[177]. LipC6 exerts its modulatory function by lations on its role in driving MS. Interestingly,
acting as a ROS scavenger, therefore inducing upregulation of CerS6 and TNFα mRNA expres-
macrophage polarization from an M2 to an M1 sion was found to be higher in females compared to
phenotype and resulting in reversal of CD8+ T male littermates in spontaneous relapsing remitting
cell exhaustion and induction of CD8+ cytotoxic EAE [183]. This increase was correlated with the
function against tumor cells. The effect of ability of females to initiate anti-inflammatory
ceramides has also been explored in suppressive responses during the course of the disease, suggest-
MDSC in mice bearing CMS4-metastic tumors. ing that CerS6 could promote anti-inflammatory
Treatment of tumor-bearing mice with acid responses. This observation was later confirmed
ceramidase inhibitor, LCL521, reduced MDSC with experiments showing that EAE progression is
accumulation in tumors without reducing tumor worsened in CerS6 knockout mice [184]. In addi-
growth. In addition, LCL521 treatment led to an tion, EAE was enhanced in chimeric mice in which
increased accumulation of C16-ceramide that only leukocytes were CerS6-deficient, providing
resulted in cathepsin-mediated cell death of further evidence that leukocytes lacking CerS6
tumor and MDSC-like J774 cells [178]. drive the exacerbated phenotype. Moreover,
Suppression of CD8+ T cell function in tumors expression of genes driving leukocyte migration
also results from suppression mediated by Treg and CNS infiltration (e.g. CCL2, CCL5, CXCL2)
cells. To our knowledge, there have been no stud- was increased, especially in CerS6-deficient neu-
ies reporting the effect of ceramides on Treg cell trophils due to increased C-X-C motif chemokine
function in tumor-bearing mice. However, recent receptor type 2 (CXCR2) in response to
studies have described the effect of ceramide granulocyte-colony stimulating factor. On the con-
accumulation on Treg cells in healthy WT and trary, CerS2-deficient mice had delayed develop-
aSMase-deficient mice. Treg cell frequency and ment of EAE. This delay in disease onset was
suppressive function are induced in aSMase-defi- correlated with the reduced expression of
cient mice [179]. In addition, this was associated CXCR2 in CerS2-deficient neutrophils [26]. In
with increased percentage of induced Treg (iTreg) addition, the induction of ceramides was also asso-
cells from aSMase−/− CD4+ T cells treated with ciated with disease development in EAE, since
transforming growth factor beta (TGFβ) and aSMase deficiency protected mice from disease
IL-2. Moreover, aSMase product C6-ceramide [185]. Collectively, these findings suggest that
induced a Th17-associtated cytokine in CD4+ T CerS6 and CerS2 have opposing roles in driving
cell hampering Treg cell induction, suggesting disease progression in EAE.
that aSMase is a negative regulator of Treg cell Excitingly, the S1P receptor agonist, fingoli-
development [180]. The relevance of these find- mod FTY720, was FDA approved in 2010 as a
ings remains to be further explored in models of first-line therapy for the treatment of MS [186,
inflammation for further development of lipid- 187]. FTY720 is phosphorylated in vivo by SK to
targeted therapies. form FTY720-P, which binds with a high affinity
to S1P receptors and competes with its natural
ligand, S1P. Therefore, FTY720 functionally
15.3.4 Multiple Sclerosis antagonizes S1P by strongly binding to S1PR
leading to internalization and inhibition of the
Multiple sclerosis (MS) is a chronic inflammatory receptor [188–190]. By modulating the S1P
disease that results in demyelination due to autoim- receptor, fingolimod prevented autoreactive S1P
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 181
expressing inflammatory T cells from exiting the COX-2 expression, and neutrophil infiltration
lymph nodes, therefore, inhibiting CNS infiltra- [203–205]. In support of these findings, high
tion by autoreactive T cells. Subsequently, this SK1 expression has also been reported in intesti-
resulted in reduced destruction of myelin sheath nal mucosa of patients with ulcerative colitis
surround the axons of nerve cells [188–191]. This [205]. Currently, modulators of S1P receptor,
is a perfect example of the potential outcome of such as ozanimod (RPC1063) and etrasimod
modulating lipid metabolism in immune cells (APD334) are being tested in clinical trials for
driving forward the era of lipid therapeutics. the treatment of IBD [206, 207], further empha-
sizing the importance of investigating the role of
SL metabolism in this disease setting.
15.3.5 Inflammatory Bowel Disease Along with inhibition of S1P receptor func-
tion, modulation of CerS has also been explored
Inflammatory Bowel Disease (IBD) is a group of in IBD, particularly CerS2 and CerS6. In a mouse
diseases, including ulcerative colitis and Crohn’s model of colitis, loss of CerS2 destabilized the
disease, characterized by chronic inflammation epithelial barrier and tight junction in the intesti-
of gastrointestinal tract. Pathology is mediated nal membrane leading to increased intestinal per-
by leukocytes infiltration and massive production meability. CerS2-deficient mice have reduced
of pro-inflammatory cytokines leading to intesti- expression of junctional adhesion molecule A
nal damage [192, 193]. The first link between SL [208] and tight junction protein ZO-1 [209]. This
metabolism and IBD originated from studies outcome is associated with reduced levels of
showing that TNFα, a cytokine that induces the very-long acyl chain ceramides (C24) and
generation of S1P and synthesis of cyclooxygen- increased levels of long-chain sphingoid bases
ase-2 (COX-2) [194, 195], was induced in and C16-ceramides [208]. These findings further
patients with IBD [192, 196]. In addition, TNFα suggest that CerS2 is protective against colitis as
blockade alleviated clinical symptoms in mouse opposed to EAE in which CerS2-deficient mice
models of disease [197, 198]. Since ceramides are protected [26]. Interestingly, further explora-
also induced the activation of MAPK, a key tion of the role of CerS6 in colitis has led to
inducer of inflammatory responses [199], follow opposing conclusions. Scheffel et al. reported
up studies demonstrated that inhibition of that the transfer of CerS6-deficient CD4+ T cells
ceramide accumulation by targeting SMase or is protective in colitis [33]. On the contrary,
S1P receptor diminished clinical manifestations Helke et al. showed that in a model of DSS-
of disease in mouse models of colitis. For exam- induced colitis, loss of CerS6 exacerbates inflam-
ple, in a dextran sulfate sodium (DSS) model of mation [210]. Interestingly, pathology in
colitis, inhibition of SMase with a sphingomy- CerS6-deficient mice is not a result of reduced
elin analogue-7 reduced the formation of intestinal permeability but is associated with
ceramide, levels of cytokines, and intestinal enhanced neutrophil infiltration. However, more
injury [200]. In addition, administration of studies are needed to further elucidate the role of
FTY720 reduced disease symptoms in a mouse CerS in various models of colitis and its imple-
model of colitis, associated with a reduced pro- mentation in the development of CerS targeted
duction of the pro-inflammatory cytokine therapies in humans with IBD.
IL-12p70 and Th1 cytokines, and upregulation of
CD4+ Foxp3+ suppressive Treg cells [201].
Furthermore, administration of a new S1P recep- 15.4 Conclusions and Future
tor agonist, KRP-203, resulted in reduced lym- Directions
phocyte infiltration and production of
pro-inflammatory Th1 cytokines in the colonic While it has been thoroughly reviewed in other
mucosa [202]. Similarly, chemical and genetic cells, the roles of ceramides in immune cells have
inhibition of SK1 that is responsible for the pro- not been proportionally presented. In this chapter
duction of S1P reduced manifestations of colitis, we attempted to summarize the significant effects
182 S. Albeituni and J. Stiban
of ceramides in immune cells and immune dis- nisms of ceramides in immune cell function and
eases. Numerous studies have highlighted the other cell types. Since diversity of ceramides,
importance of ceramides in a variety of pathways similar to other membrane and bioactive lipids,
necessary for the development, function, and arises from the different possible combinations
metabolism of immune cells. Ceramide accumu- of head groups and/or degree of saturation, the
lation has been described as a key step mediating effect of each type of SL and ceramide on every
and regulating various immune cell functions, immune cell opens endless possibilities in target-
including regulation of immune cell responses to ing various ceramides to retune immune cell
viruses, bacteria, and other foreign pathogens, function in cancer, transplantation, diabetes, MS,
migration, phagocytosis, and cytokine produc- IBD and many other diseases.
tion. For instance, ceramides have been impli-
cated in the modulation of responses to LPS, Acknowledgements The authors are indepted to Ms.
cytokine production, antigen presentation, and Yara Khodour for her diligent redrawing of Fig. 15.2 and
her full chapter proof. Dr. Johnny Stiban acknowledges
viral entry in DC; control of migration, cytokine the receipt of two grants from Birzeit University that
and superoxide production, and formation of allowed him to pursue this endeavor (Grant #240193 and
NETs in neutrophils; and response to TLR stimu- #241109).
lation, inducing apoptosis, and regulating
oxidative stress in macrophages. Moreover,
ceramide functions extend to T cells, stabilizing References
the T cell receptor complex and mediating T cell
responses during inflammation. Ceramide metab- 1. Merrill AH Jr (2011) Sphingolipid and glycosphin-
golipid metabolic pathways in the era of sphingolipi-
olites and upstream regulators including CerS in domics. Chem Rev 111:6387–6422
immune cell function have also been thoroughly 2. Park JW, Park WJ, Futerman AH (2014) Ceramide
described in the literature, further emphasizing synthases as potential targets for therapeutic inter-
the important role that ceramides play during vention in human diseases. Biochim Biophys Acta
1841:671–681
inflammation. 3. Don AS, Lim XY, Couttas TA (2014)
Investigating the roles of ceramides and other Re-configuration of sphingolipid metabolism by
SLs as modulators of the biology and metabo- oncogenic transformation. Biomol Ther 4:315–353
lism of immune cells has unraveled new avenues 4. Lopez PH, Schnaar RL (2009) Gangliosides in cell
recognition and membrane protein regulation. Curr
in targeting ceramides in life-threatening inflam- Opin Struct Biol 19:549–557
matory diseases that negatively impact the life of 5. Hannun YA, Obeid LM (2011) Many ceramides.
millions around the globe. For instance, recently, J Biol Chem 286:27855–27862
the FDA approved the CerS inhibitor, FTY720, 6. Abou-Ghali M, Stiban J (2015) Regulation of
ceramide channel formation and disassembly:
as the first-line therapy for MS patients. This insights on the initiation of apoptosis. Saudi J Biol
certainly has raised an interest among immunol- Sci 22:760–772
ogists and lipid biochemists alike in developing 7. Rappocciolo E, Stiban J (2019) Prokaryotic
novel inhibitors to target ceramide metabolism and mitochondrial lipids: a survey of evolution-
ary origins. Adv Exp Med Biol 1159. https://doi.
in various models of inflammation in the labora- org/10.1007/978-3-030-21162-2. (in press)
tory to hopefully translate such findings in the 8. Bikman BT, Summers SA (2011) Ceramides as
clinic. modulators of cellular and whole-body metabolism.
It is also strictly important to consider the J Clin Invest 121:4222–4230
9. Stiban J, Tidhar R, Futerman AH (2010) Ceramide
multiple effects of a plethora of ceramide species synthases: roles in cell physiology and signaling.
on the function of a certain immune cell or differ- Adv Exp Med Biol 688:60–71
ent cells when targeting ceramides in disease. 10. Tidhar R, Futerman AH (2013) The complexity of
Despite the encouraging routes that ceramide sphingolipid biosynthesis in the endoplasmic reticu-
lum. Biochim Biophys Acta 1833:2511–2518
research has taken, further studies and collabora- 11. Boulgaropoulos B, Amenitsch H, Laggner P, Pabst
tions between scientists of multidisciplinary G (2010) Implication of sphingomyelin/ceramide
fields are still needed to elucidate the mecha-
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 183
molar ratio on the biological activity of sphingomy- hepatectomy. Biochem Biophys Res Commun
elinase. Biophys J 99:499–506 493:1176–1183
12. Claus RA, Dorer MJ, Bunck AC, Deigner HP (2009) 24. Chen Z, Han Y, Gu Y, Liu Y, Jiang Z, Zhang M, Cao
Inhibition of sphingomyelin hydrolysis: targeting X (2013) CD11c(high)CD8+ regulatory T cell feed-
the lipid mediator ceramide as a key regulator of cel- back inhibits CD4 T cell immune response via Fas
lular fate. Curr Med Chem 16:1978–2000 ligand-Fas pathway. J Immunol (Baltimore, Md:
13. Kitatani K, Idkowiak-Baldys J, Hannun YA (2008) 1950) 190:6145–6154
The sphingolipid salvage pathway in ceramide 25. Park WJ, Brenner O, Kogot-Levin A, Saada A,
metabolism and signaling. Cell Signal 20:1010–1018 Merrill AH Jr, Pewzner-Jung Y, Futerman AH
14. Mullen TD, Hannun YA, Obeid LM (2012) Ceramide (2015) Development of pheochromocytoma in
synthases at the Centre of sphingolipid metabolism ceramide synthase 2 null mice. Endocr Relat Cancer
and biology. Biochem J 441:789–802 22:623–632
15. Zelnik ID, Rozman B, Rosenfeld-Gur E, Ben- 26. Barthelmes J, de Bazo AM, Pewzner-Jung Y, Schmitz
Dor S, Futerman AH (2019) A stroll down the K, Mayer CA, Foerch C, Eberle M, Tafferner N,
CerS lane. Adv Exp Med Biol 1159. https://doi. Ferreiros N, Henke M, Geisslinger G, Futerman AH,
org/10.1007/978-3-030-21162-2. (in press) Grosch S, Schiffmann S (2015) Lack of ceramide
16. Laviad EL, Albee L, Pankova-Kholmyansky I, synthase 2 suppresses the development of experi-
Epstein S, Park H, Merrill AH Jr, Futerman AH mental autoimmune encephalomyelitis by impairing
(2008) Characterization of ceramide synthase 2: the migratory capacity of neutrophils. Brain Behav
tissue distribution, substrate specificity, and inhi- Immun 46:280–292
bition by sphingosine 1-phosphate. J Biol Chem 27. Chen L, Lu X, Zeng T, Chen Y, Chen Q, Wu W,
283:5677–5684 Yan X, Cai H, Zhang Z, Shao Q, Qin W (2014)
17. Ginkel C, Hartmann D, vom Dorp K, Zlomuzica Enhancement of DEN-induced liver tumourigenesis
A, Farwanah H, Eckhardt M, Sandhoff R, Degen in hepatocyte-specific Lass2-knockout mice coin-
J, Rabionet M, Dere E, Dormann P, Sandhoff K, cident with upregulation of the TGF-beta1-Smad4-
Willecke K (2012) Ablation of neuronal ceramide PAI-1 axis. Oncol Rep 31:885–893
synthase 1 in mice decreases ganglioside levels and 28. Imgrund S, Hartmann D, Farwanah H, Eckhardt
expression of myelin-associated glycoprotein in oli- M, Sandhoff R, Degen J, Gieselmann V, Sandhoff
godendrocytes. J Biol Chem 287:41888–41902 K, Willecke K (2009) Adult ceramide synthase
18. Jennemann R, Rabionet M, Gorgas K, Epstein S, 2 (CERS2)-deficient mice exhibit myelin sheath
Dalpke A, Rothermel U, Bayerle A, van der Hoeven defects, cerebellar degeneration, and hepatocarcino-
F, Imgrund S, Kirsch J, Nickel W, Willecke K, mas. J Biol Chem 284:33549–33560
Riezman H, Grone HJ, Sandhoff R (2012) Loss of 29. Pewzner-Jung Y, Brenner O, Braun S, Laviad EL,
ceramide synthase 3 causes lethal skin barrier dis- Ben-Dor S, Feldmesser E, Horn-Saban S, Amann-
ruption. Hum Mol Genet 21:586–608 Zalcenstein D, Raanan C, Berkutzki T, Erez-Roman
19. Mizutani Y, Kihara A, Igarashi Y (2005) R, Ben-David O, Levy M, Holzman D, Park H,
Mammalian Lass6 and its related family members Nyska A, Merrill AH Jr, Futerman AH (2010a) A
regulate synthesis of specific ceramides. Biochem critical role for ceramide synthase 2 in liver homeo-
J 390:263–271 stasis: II. Insights into molecular changes leading to
20. Gosejacob D, Jager PS, Vom Dorp K, Frejno M, hepatopathy. J Biol Chem 285:10911–10923
Carstensen AC, Kohnke M, Degen J, Dormann P, 30. Pewzner-Jung Y, Park H, Laviad EL, Silva LC,
Hoch M (2016) Ceramide synthase 5 is essential Lahiri S, Stiban J, Erez-Roman R, Brugger B,
to maintain C16:0-ceramide pools and contributes Sachsenheimer T, Wieland F, Prieto M, Merrill
to the development of diet-induced obesity. J Biol AH Jr, Futerman AH (2010b) A critical role for
Chem 291:6989–7003 ceramide synthase 2 in liver homeostasis: I. altera-
21. Tidhar R, Zelnik ID, Volpert G, Ben-Dor S, Kelly S, tions in lipid metabolic pathways. J Biol Chem
Merrill AH Jr, Futerman AH (2018) Eleven residues 285:10902–10910
determine the acyl chain specificity of ceramide syn- 31. Rabionet M, Bayerle A, Jennemann R, Heid H,
thases. J Biol Chem 293:9912–9921 Fuchser J, Marsching C, Porubsky S, Bolenz C,
22. Zhao L, Spassieva SD, Jucius TJ, Shultz LD, Guillou F, Grone HJ, Gorgas K, Sandhoff R (2015)
Shick HE, Macklin WB, Hannun YA, Obeid LM, Male meiotic cytokinesis requires ceramide synthase
Ackerman SL (2011) A deficiency of ceramide 3-dependent sphingolipids with unique membrane
biosynthesis causes cerebellar Purkinje cell neuro- anchors. Hum Mol Genet 24:4792–4808
degeneration and lipofuscin accumulation. PLoS 32. Ebel P, Imgrund S, Vom Dorp K, Hofmann K, Maier
Genet 7:e1002063 H, Drake H, Degen J, Dormann P, Eckhardt M,
23. Jin H, Wang C, Gu D, Zhang Y, Fan S, Xing S, Franz T, Willecke K (2014) Ceramide synthase 4
Wang H, Ruan H, Yang C, Lv Y, Feng H, Yao M, deficiency in mice causes lipid alterations in sebum
Qin W (2017) Liver-specific deletion of LASS2 and results in alopecia. Biochem J 461:147–158
delayed regeneration of mouse liver after partial 33. Scheffel MJ, Helke K, Lu P, Bowers JS, Ogretmen
B, Garrett-Mayer E, Paulos CM, Voelkel-Johnson
184 S. Albeituni and J. Stiban
C (2017) Adoptive transfer of ceramide synthase 6 49. Siskind LJ, Davoody A, Lewin N, Marshall S,
deficient Splenocytes reduces the development of Colombini M (2003) Enlargement and contracture
colitis. Sci Rep 7:15552 of C2-ceramide channels. Biophys J 85:1560–1575
34. Ebel P, Vom Dorp K, Petrasch-Parwez E, Zlomuzica 50. Samanta S, Stiban J, Maugel TK, Colombini M
A, Kinugawa K, Mariani J, Minich D, Ginkel C, (2011) Visualization of ceramide channels by trans-
Welcker J, Degen J, Eckhardt M, Dere E, Dormann mission electron microscopy. Biochim Biophys Acta
P, Willecke K (2013) Inactivation of ceramide syn- 1808:1196–1201
thase 6 in mice results in an altered sphingolipid 51. Siskind LJ, Kolesnick RN, Colombini M (2002)
metabolism and behavioral abnormalities. J Biol Ceramide channels increase the permeability of the
Chem 288:21433–21447 mitochondrial outer membrane to small proteins.
35. Peters F, Vorhagen S, Brodesser S, Jakobshagen J Biol Chem 277:26796–26803
K, Bruning JC, Niessen CM, Kronke M (2015) 52. Siskind LJ, Kolesnick RN, Colombini M (2006)
Ceramide synthase 4 regulates stem cell homeo- Ceramide forms channels in mitochondrial outer
stasis and hair follicle cycling. J Invest Dermatol membranes at physiologically relevant concentra-
135:1501–1509 tions. Mitochondrion 6:118–125
36. Futerman AH, Hannun YA (2004) The complex life 53. Stiban J, Fistere D, Colombini M (2006)
of simple sphingolipids. EMBO Rep 5:777–782 Dihydroceramide hinders ceramide channel for-
37. Hannun YA, Luberto C (2000) Ceramide in the mation: implications on apoptosis. Apoptosis
eukaryotic stress response. Trends Cell Biol 11:773–780
10:73–80 54. Yamane M, Moriya S, Kokuba H (2017) Visualization
38. Pinto SN, Silva LC, Futerman AH, Prieto M (2011) of ceramide channels in lysosomes following endog-
Effect of ceramide structure on membrane biophysi- enous palmitoyl-ceramide accumulation as an initial
cal properties: the role of acyl chain length and unsat- step in the induction of necrosis. Biochem Biophys
uration. Biochim Biophys Acta 1808:2753–2760 Rep 11:174–181
39. Silva LC, Ben David O, Pewzner-Jung Y, Laviad EL, 55. Steinman RM (2012) Decisions about dendritic
Stiban J, Bandyopadhyay S, Merrill AH Jr, Prieto M, cells: past, present, and future. Annu Rev Immunol
Futerman AH (2012) Ablation of ceramide synthase 30:1–22
2 strongly affects biophysical properties of mem- 56. Steinman RM, Cohn ZA (2007) Pillars article: iden-
branes. J Lipid Res 53:430–436 tification of a novel cell type in peripheral lymphoid
40. Silva LC, Futerman AH, Prieto M (2009) Lipid raft organs of mice. I. Morphology, quantitation, tis-
composition modulates sphingomyelinase activity sue distribution. J Immunol (Baltimore, Md: 1950)
and ceramide-induced membrane physical altera- 178:5–25
tions. Biophys J 96:3210–3222 57. Ashany D, Savir A, Bhardwaj N, Elkon KB (1999)
41. Stiban JS, Silva LC, Futerman AH (2008) Ceramide- Dendritic cells are resistant to apoptosis through the
containing membranes: the interface between bio- Fas (CD95/APO-1) pathway. J Immunol (Baltimore,
physics and biology. Trends Glycosci Glycotechnol Md: 1950) 163:5303–5311
20:297–313 58. Kanto T, Kalinski P, Hunter OC, Lotze MT, Amoscato
42. Stiban J (2019) Introduction: enigmas of sphin- AA (2001) Ceramide mediates tumor-induced den-
golipids. Adv Exp Med Biol 1159. https://doi. dritic cell apoptosis. J Immunol (Baltimore, Md:
org/10.1007/978-3-030-21162-2. (in press) 1950) 167:3773–3784
43. Hannun YA, Obeid LM (2002) The ceramide- 59. Sallusto F, Cella M, Danieli C, Lanzavecchia A
centric universe of lipid-mediated cell regulation: (1995) Dendritic cells use macropinocytosis and the
stress encounters of the lipid kind. J Biol Chem mannose receptor to concentrate macromolecules in
277:25847–25850 the major histocompatibility complex class II com-
44. Nganga R, Oleinik N, Ogretmen B (2018) partment: downregulation by cytokines and bacterial
Mechanisms of ceramide-dependent cancer cell products. J Exp Med 182:389–400
death. Adv Cancer Res 140:1–25 60. Sallusto F, Nicolo C, De Maria R, Corinti S, Testi
45. Kolesnick R, Golde DW (1994) The sphingomyelin R (1996) Ceramide inhibits antigen uptake and
pathway in tumor necrosis factor and interleukin-1 presentation by dendritic cells. J Exp Med 184:
signaling. Cell 77:325–328 2411–2416
46. Maceyka M, Spiegel S (2014) Sphingolipid metabo- 61. Franchi L, Malisan F, Tomassini B, Testi R (2006)
lites in inflammatory disease. Nature 510:58–67 Ceramide catabolism critically controls survival of
47. Colombini M (2019) Ceramide channels. Adv Exp human dendritic cells. J Leukoc Biol 79:166–172
Med Biol 1159. https://doi.org/10.1007/978-3- 62. Joseph CK, Wright SD, Bornmann WG, Randolph
030-21162-2. (in press) JT, Kumar ER, Bittman R, Liu J, Kolesnick RN
48. Siskind LJ, Colombini M (2000) The lipids (1994) Bacterial lipopolysaccharide has structural
C2- and C16-ceramide form large stable chan- similarity to ceramide and stimulates ceramide-
nels. Implications for apoptosis. J Biol Chem activated protein kinase in myeloid cells. J Biol
275:38640–38644 Chem 269:17606–17610
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 185
63. MacKichan ML, DeFranco AL (1999) Role of in induction of apoptosis by tumor necrosis factor-
ceramide in lipopolysaccharide (LPS)-induced sig- alpha in human neutrophils. FEBS Lett 355:267–270
naling. LPS increases ceramide rather than acting as 76. Kim MY, Linardic C, Obeid L, Hannun Y (1991)
a structural homolog. J Biol Chem 274:1767–1775 Identification of sphingomyelin turnover as an effec-
64. Harouse JM, Bhat S, Spitalnik SL, Laughlin M, tor mechanism for the action of tumor necrosis fac-
Stefano K, Silberberg DH, Gonzalez-Scarano F tor alpha and gamma-interferon. Specific role in cell
(1991) Inhibition of entry of HIV-1 in neural cell differentiation. J Biol Chem 266:484–489
lines by antibodies against galactosyl ceramide. 77. Dressler KA, Mathias S, Kolesnick RN (1992)
Science (New York, NY) 253:320–323 Tumor necrosis factor-alpha activates the sphingo-
65. Yahi N, Sabatier JM, Baghdiguian S, Gonzalez- myelin signal transduction pathway in a cell-free
Scarano F, Fantini J (1995) Synthetic multimeric system. Science (New York, NY) 255:1715–1718
peptides derived from the principal neutralization 78. Fiore S, Nicolaou KC, Caulfield T, Kataoka H,
domain (V3 loop) of human immunodeficiency virus Serhan CN (1990) Evaluation of synthetic sphingo-
type 1 (HIV-1) gp120 bind to galactosylceramide sine, lysosphingolipids and glycosphingolipids as
and block HIV-1 infection in a human CD4-negative inhibitors of functional responses of human neutro-
mucosal epithelial cell line. J Virol 69:320–325 phils. Biochem J 266:25–31
66. Alfsen A, Bomsel M (2002) HIV-1 gp41 envelope 79. Robinson BS, Hii CS, Poulos A, Ferrante A
residues 650-685 exposed on native virus act as a (1997) Activation of neutral sphingomyelinase in
lectin to bind epithelial cell galactosyl ceramide. human neutrophils by polyunsaturated fatty acids.
J Biol Chem 277:25649–25659 Immunology 91:274–280
67. Alfsen A, Iniguez P, Bouguyon E, Bomsel M (2001) 80. Ahmed N, Berridge MV (2000) Ceramides that
Secretory IgA specific for a conserved epitope on mediate apoptosis reduce glucose uptake and trans-
gp41 envelope glycoprotein inhibits epithelial tran- porter affinity for glucose in human leukaemic cell
scytosis of HIV-1. J Immunol (Baltimore, Md: 1950) lines but not in neutrophils. Pharmacol Toxicol
166:6257–6265 86:114–121
68. Lingwood CA, Branch DR (2011) The role of 81. Fuortes M, Jin W, Nathan C (1996) Ceramide selec-
glycosphingolipids in HIV/AIDS. Discov Med tively inhibits early events in the response of human
11:303–313 neutrophils to tumor necrosis factor. J Leukoc Biol
69. Magerus-Chatinet A, Yu H, Garcia S, Ducloux E, 59:451–460
Terris B, Bomsel M (2007) Galactosyl ceramide 82. Nakamura T, Abe A, Balazovich KJ, Wu D, Suchard
expressed on dendritic cells can mediate HIV-1 SJ, Boxer LA, Shayman JA (1994) Ceramide regu-
transfer from monocyte derived dendritic cells to lates oxidant release in adherent human neutrophils.
autologous T cells. Virology 362:67–74 J Biol Chem 269:18384–18389
70. Avota E, Gulbins E, Schneider-Schaulies S (2011) 83. Sitrin RG, Sassanella TM, Petty HR (2011) An
DC-SIGN mediated sphingomyelinase-activation obligate role for membrane-associated neutral
and ceramide generation is essential for enhance- sphingomyelinase activity in orienting chemotactic
ment of viral uptake in dendritic cells. PLoS Pathog migration of human neutrophils. Am J Respir Cell
7:e1001290 Mol Biol 44:205–212
71. Pritzl CJ, Seo YJ, Xia C, Vijayan M, Stokes ZD, 84. Suchard SJ, Mansfield PJ, Boxer LA, Shayman JA
Hahm B (2015) A ceramide analogue stimulates (1997b) Mitogen-activated protein kinase activa-
dendritic cells to promote T cell responses upon tion during IgG-dependent phagocytosis in human
virus infections. J Immunol (Baltimore, Md: 1950) neutrophils: inhibition by ceramide. J Immunol
194:4339–4349 (Baltimore, Md: 1950) 158:4961–4967
72. Mantovani A, Cassatella MA, Costantini C, Jaillon 85. Hinkovska-Galcheva V, Kjeldsen L, Mansfield
S (2011) Neutrophils in the activation and regulation PJ, Boxer LA, Shayman JA, Suchard SJ (1998)
of innate and adaptive immunity. Nat Rev Immunol Activation of a plasma membrane-associated neu-
11:519–531 tral sphingomyelinase and concomitant ceramide
73. Wilson E, Wang E, Mullins RE, Uhlinger DJ, Liotta accumulation during IgG-dependent phagocytosis
DC, Lambeth JD, Merrill AH Jr (1988) Modulation in human polymorphonuclear leukocytes. Blood
of the free sphingosine levels in human neutrophils 91:4761–4769
by phorbol esters and other factors. J Biol Chem 86. Suchard SJ, Hinkovska-Galcheva V, Mansfield PJ,
263:9304–9309 Boxer LA, Shayman JA (1997a) Ceramide inhibits
74. Utsumi T, Klostergaard J, Akimaru K, Edashige IgG-dependent phagocytosis in human polymorpho-
K, Sato EF, Utsumi K (1992) Modulation of nuclear leukocytes. Blood 89:2139–2147
TNF-alpha- priming and stimulation-dependent 87. Hinkovska-Galcheva V, Boxer L, Mansfield PJ,
superoxide generation in human neutrophils by Schreiber AD, Shayman JA (2003) Enhanced phago-
protein kinase inhibitors. Arch Biochem Biophys cytosis through inhibition of de novo ceramide syn-
294:271–278 thesis. J Biol Chem 278:974–982
75. Ohta H, Yatomi Y, Sweeney EA, Hakomori S, 88. Corriden R, Hollands A, Olson J, Derieux J, Lopez J,
Igarashi Y (1994) A possible role of sphingosine Chang JT, Gonzalez DJ, Nizet V (2015) Tamoxifen
186 S. Albeituni and J. Stiban
augments the innate immune function of neutrophils in lactosylceramide- mediated superoxide gen-
through modulation of intracellular ceramide. Nat eration and migration in neutrophils. Glycoconj
Commun 6:8369 J 25:357–374
89. Seumois G, Fillet M, Gillet L, Faccinetto C, Desmet 100. Sonnino S, Prinetti A, Nakayama H, Yangida M,
C, Francois C, Dewals B, Oury C, Vanderplasschen Ogawa H, Iwabuchi K (2009) Role of very long fatty
A, Lekeux P, Bureau F (2007) De novo C16- and acid-containing glycosphingolipids in membrane
C24-ceramide generation contributes to spontaneous organization and cell signaling: the model of lacto-
neutrophil apoptosis. J Leukoc Biol 81:1477–1486 sylceramide in neutrophils. Glycoconj J 26:615–621
90. Scheel-Toellner D, Wang K, Assi LK, Webb 101. Nakayama H, Kurihara H, Morita YS, Kinoshita
PR, Craddock RM, Salmon M, Lord JM (2004) T, Mauri L, Prinetti A, Sonnino S, Yokoyama
Clustering of death receptors in lipid rafts initiates N, Ogawa H, Takamori K, Iwabuchi K (2016)
neutrophil spontaneous apoptosis. Biochem Soc Lipoarabinomannan binding to lactosylceramide
Trans 32:679–681 in lipid rafts is essential for the phagocytosis of
91. Manago A, Becker KA, Carpinteiro A, Wilker B, mycobacteria by human neutrophils. Sci Signal
Soddemann M, Seitz AP, Edwards MJ, Grassme H, 9:ra101
Szabo I, Gulbins E (2015) Pseudomonas aeruginosa 102. Izawa K, Maehara A, Isobe M, Yasuda Y, Urai M,
pyocyanin induces neutrophil death via mitochon- Hoshino Y, Ueno K, Matsukawa T, Takahashi M,
drial reactive oxygen species and mitochondrial Kaitani A, Shiba E, Takamori A, Uchida S, Uchida
acid sphingomyelinase. Antioxid Redox Signal K, Maeda K, Nakano N, Yamanishi Y, Oki T,
22:1097–1110 Voehringer D, Roers A, Nakae S, Ishikawa J, Kinjo
92. Tafesse FG, Huitema K, Hermansson M, van der Y, Shimizu T, Ogawa H, Okumura K, Kitamura T,
Poel S, van den Dikkenberg J, Uphoff A, Somerharju Kitaura J (2017) Disrupting ceramide-CD300f inter-
P, Holthuis JC (2007) Both sphingomyelin synthases action prevents septic peritonitis by stimulating neu-
SMS1 and SMS2 are required for sphingomyelin trophil recruitment. Sci Rep 7:4298
homeostasis and growth in human HeLa cells. J Biol 103. Shiba E, Izawa K, Kaitani A, Isobe M, Maehara
Chem 282:17537–17547 A, Uchida K, Maeda K, Nakano N, Ogawa H,
93. Tafesse FG, Ternes P, Holthuis JC (2006) The mul- Okumura K, Kitamura T, Shimizu T, Kitaura
tigenic sphingomyelin synthase family. J Biol Chem J (2017) Ceramide-CD300f binding inhibits
281:29421–29425 lipopolysaccharide-induced skin inflammation.
94. Qureshi A, Subathra M, Grey A, Schey K, Del J Biol Chem 292:2924–2932
Poeta M, Luberto C (2010) Role of sphingomyelin 104. Izawa K, Yamanishi Y, Maehara A, Takahashi M,
synthase in controlling the antimicrobial activity of Isobe M, Ito S, Kaitani A, Matsukawa T, Matsuoka
neutrophils against Cryptococcus neoformans. PLoS T, Nakahara F, Oki T, Kiyonari H, Abe T, Okumura
One 5:e15587 K, Kitamura T, Kitaura J (2012) The receptor LMIR3
95. Symington FW, Hedges DL, Hakomori S (1985) negatively regulates mast cell activation and aller-
Glycolipid antigens of human polymorphonuclear gic responses by binding to extracellular ceramide.
neutrophils and the inducible HL-60 myeloid leu- Immunity 37:827–839
kemia line. J Immunol (Baltimore, Md: 1950) 105. Matsukawa T, Izawa K, Isobe M, Takahashi M,
134:2498–2506 Maehara A, Yamanishi Y, Kaitani A, Okumura K,
96. Arai T, Bhunia AK, Chatterjee S, Bulkley GB (1998) Teshima T, Kitamura T, Kitaura J (2016) Ceramide-
Lactosylceramide stimulates human neutrophils to CD300f binding suppresses experimental colitis by
upregulate Mac-1, adhere to endothelium, and gen- inhibiting ATP-mediated mast cell activation. Gut
erate reactive oxygen metabolites in vitro. Circ Res 65:777–787
82:540–547 106. Kawa S, Kimura S, Hakomori S, Igarashi Y (1997)
97. Iwabuchi K, Nagaoka I (2002) Lactosylceramide- Inhibition of chemotactic motility and trans-
enriched glycosphingolipid signaling domain medi- endothelial migration of human neutrophils by
ates superoxide generation from human neutrophils. sphingosine 1-phosphate. FEBS Lett 420:196–200
Blood 100:1454–1464 107. Chandru H, Boggaram V (2007) The role of sphin-
98. Chiricozzi E, Ciampa MG, Brasile G, Compostella gosine 1-phosphate in the TNF-alpha induction of
F, Prinetti A, Nakayama H, Ekyalongo RC, Iwabuchi IL-8 gene expression in lung epithelial cells. Gene
K, Sonnino S, Mauri L (2015) Direct interac- 391:150–160
tion, instrumental for signaling processes, between 108. Baudiss K, de Paula Vieira R, Cicko S, Ayata
LacCer and Lyn in the lipid rafts of neutrophil-like K, Hossfeld M, Ehrat N, Gomez-Munoz A,
cells. J Lipid Res 56:129–141 Eltzschig HK, Idzko M (2016) C1P attenuates
99. Iwabuchi K, Prinetti A, Sonnino S, Mauri L, lipopolysaccharide-induced acute lung injury by
Kobayashi T, Ishii K, Kaga N, Murayama K, preventing NF-kappaB activation in neutrophils.
Kurihara H, Nakayama H, Yoshizaki F, Takamori J Immunol (Baltimore, Md: 1950) 196:2319–2326
K, Ogawa H, Nagaoka I (2008) Involvement of
very long fatty acid-containing lactosylceramide
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 187
109. Italiani P, Boraschi D (2015) New insights into tissue 121. Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT,
macrophages: from their origin to the development Brickey WJ, Ting JP (2011) Fatty acid-induced
of memory. Immune Netw 15:167–176 NLRP3-ASC inflammasome activation interferes
110. Murray PJ, Wynn TA (2011) Protective and patho- with insulin signaling. Nat Immunol 12:408–415
genic functions of macrophage subsets. Nat Rev 122. Camell CD, Nguyen KY, Jurczak MJ, Christian
Immunol 11:723–737 BE, Shulman GI, Shadel GS, Dixit VD (2015)
111. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy Macrophage-specific de novo synthesis of ceramide
DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv is dispensable for Inflammasome-driven inflamma-
LB, Lawrence T, Locati M, Mantovani A, Martinez tion and insulin resistance in obesity. J Biol Chem
FO, Mege JL, Mosser DM, Natoli G, Saeij JP, 290:29402–29413
Schultze JL, Shirey KA, Sica A, Suttles J, Udalova 123. Serbulea V, Upchurch CM, Ahern KW, Bories
I, van Ginderachter JA, Vogel SN, Wynn TA (2014) G, Voigt P, DeWeese DE, Meher AK, Harris TE,
Macrophage activation and polarization: nomen- Leitinger N (2018) Macrophages sensing oxidized
clature and experimental guidelines. Immunity DAMPs reprogram their metabolism to support
41:14–20 redox homeostasis and inflammation through a
112. Stout RD, Suttles J (2004) Functional plasticity of TLR2-Syk-ceramide dependent mechanism. Mol
macrophages: reversible adaptation to changing Metabol 7:23–34
microenvironments. J Leukoc Biol 76:509–513 124. Hundal RS, Gomez-Munoz A, Kong JY, Salh BS,
113. Bhargava P, Lee CH (2012) Role and function of Marotta A, Duronio V, Steinbrecher UP (2003)
macrophages in the metabolic syndrome. Biochem Oxidized low density lipoprotein inhibits macro-
J 442:253–262 phage apoptosis by blocking ceramide generation,
114. Prieur X, Roszer T, Ricote M (2010) Lipotoxicity thereby maintaining protein kinase B activation and
in macrophages: evidence from diseases associated Bcl-XL levels. J Biol Chem 278:24399–24408
with the metabolic syndrome. Biochim Biophys 125. Aflaki E, Doddapattar P, Radovic B, Povoden S, Kolb
Acta 1801:327–337 D, Vujic N, Wegscheider M, Koefeler H, Hornemann
115. Akira S, Takeda K, Kaisho T (2001) Toll-like recep- T, Graier WF, Malli R, Madeo F, Kratky D (2012)
tors: critical proteins linking innate and acquired C16 ceramide is crucial for triacylglycerol-induced
immunity. Nat Immunol 2:675–680 apoptosis in macrophages. Cell Death Dis 3:e280
116. Beutler B, Hoebe K, Georgel P, Tabeta K, Du X 126. Wang SW, Hojabrpour P, Zhang P, Kolesnick RN,
(2005) Genetic analysis of innate immunity: identifi- Steinbrecher UP, Gomez-Munoz A, Duronio V
cation and function of the TIR adapter proteins. Adv (2015) Regulation of ceramide generation during
Exp Med Biol 560:29–39 macrophage apoptosis by ASMase and de novo syn-
117. Andreyev AY, Fahy E, Guan Z, Kelly S, Li X, thesis. Biochim Biophys Acta 1851:1482–1489
McDonald JG, Milne S, Myers D, Park H, Ryan 127. Zhang Y, Rao E, Zeng J, Hao J, Sun Y, Liu S, Sauter
A, Thompson BM, Wang E, Zhao Y, Brown HA, ER, Bernlohr DA, Cleary MP, Suttles J, Li B (2017)
Merrill AH, Raetz CR, Russell DW, Subramaniam Adipose fatty acid binding protein promotes satu-
S, Dennis EA (2010) Subcellular organelle lipido- rated fatty acid-induced macrophage cell death
mics in TLR-4-activated macrophages. J Lipid Res through enhancing ceramide production. J Immunol
51:2785–2797 (Baltimore, Md: 1950) 198:798–807
118. Dennis EA, Deems RA, Harkewicz R, Quehenberger 128. Holland WL, Brozinick JT, Wang LP, Hawkins ED,
O, Brown HA, Milne SB, Myers DS, Glass CK, Sargent KM, Liu Y, Narra K, Hoehn KL, Knotts TA,
Hardiman G, Reichart D, Merrill AH Jr, Sullards MC, Siesky A, Nelson DH, Karathanasis SK, Fontenot
Wang E, Murphy RC, Raetz CR, Garrett TA, Guan GK, Birnbaum MJ, Summers SA (2007) Inhibition
Z, Ryan AC, Russell DW, McDonald JG, Thompson of ceramide synthesis ameliorates glucocorticoid-,
BM, Shaw WA, Sud M, Zhao Y, Gupta S, Maurya saturated-fat-, and obesity-induced insulin resis-
MR, Fahy E, Subramaniam S (2010) A mouse mac- tance. Cell Metab 5:167–179
rophage lipidome. J Biol Chem 285:39976–39985 129. Nelson DH, Murray DK (1986) Sphingolipids
119. Sims K, Haynes CA, Kelly S, Allegood JC, Wang inhibit insulin and phorbol ester stimulated uptake
E, Momin A, Leipelt M, Reichart D, Glass CK, of 2-deoxyglucose. Biochem Biophys Res Commun
Sullards MC, Merrill AH Jr (2010) Kdo2-lipid A, a 138:463–467
TLR4-specific agonist, induces de novo sphingolipid 130. Kanety H, Hemi R, Papa MZ, Karasik A (1996)
biosynthesis in RAW264.7 macrophages, which is Sphingomyelinase and ceramide suppress insulin-
essential for induction of autophagy. J Biol Chem induced tyrosine phosphorylation of the insulin
285:38568–38579 receptor substrate-1. J Biol Chem 271:9895–9897
120. Schilling JD, Machkovech HM, He L, Sidhu R, 131. Peraldi P, Hotamisligil GS, Buurman WA, White
Fujiwara H, Weber K, Ory DS, Schaffer JE (2013) MF, Spiegelman BM (1996) Tumor necrosis factor
Palmitate and lipopolysaccharide trigger synergistic (TNF)-alpha inhibits insulin signaling through stim-
ceramide production in primary macrophages. J Biol ulation of the p55 TNF receptor and activation of
Chem 288:2923–2932 sphingomyelinase. J Biol Chem 271:13018–13022
188 S. Albeituni and J. Stiban
132. Schmitz-Peiffer C, Craig DL, Biden TJ (1999) 144. Adams JM 2nd, Pratipanawatr T, Berria R, Wang E,
Ceramide generation is sufficient to account for the DeFronzo RA, Sullards MC, Mandarino LJ (2004)
inhibition of the insulin-stimulated PKB pathway in Ceramide content is increased in skeletal muscle
C2C12 skeletal muscle cells pretreated with palmi- from obese insulin-resistant humans. Diabetes
tate. J Biol Chem 274:24202–24210 53:25–31
133. Summers SA, Garza LA, Zhou H, Birnbaum MJ 145. Blachnio-Zabielska AU, Chacinska M, Vendelbo
(1998) Regulation of insulin-stimulated glucose MH, Zabielski P (2016) The crucial role of C18-Cer
transporter GLUT4 translocation and Akt kinase in fat-induced skeletal muscle insulin resistance.
activity by ceramide. Mol Cell Biol 18:5457–5464 Cell Physiol Biochem 40:1207–1220
134. Wang CN, O’Brien L, Brindley DN (1998) Effects 146. Coen PM, Dube JJ, Amati F, Stefanovic-Racic M,
of cell-permeable ceramides and tumor necrosis Ferrell RE, Toledo FG, Goodpaster BH (2010)
factor-alpha on insulin signaling and glucose uptake Insulin resistance is associated with higher intra-
in 3T3-L1 adipocytes. Diabetes 47:24–31 myocellular triglycerides in type I but not type II
135. Zhou H, Summers SA, Birnbaum MJ, Pittman RN myocytes concomitant with higher ceramide con-
(1998) Inhibition of Akt kinase by cell-permeable tent. Diabetes 59:80–88
ceramide and its implications for ceramide-induced 147. Fillmore N, Keung W, Kelly SE, Proctor SD,
apoptosis. J Biol Chem 273:16568–16575 Lopaschuk GD, Ussher JR (2015) Accumulation
136. Zinda MJ, Vlahos CJ, Lai MT (2001) Ceramide of ceramide in slow-twitch muscle contributes to
induces the dephosphorylation and inhibition of the development of insulin resistance in the obese
constitutively activated Akt in PTEN negative JCR:LA-cp rat. Exp Physiol 100:730–741
U87mg cells. Biochem Biophys Res Commun 148. Pickersgill L, Litherland GJ, Greenberg AS, Walker
280:1107–1115 M, Yeaman SJ (2007) Key role for ceramides in
137. Salinas M, Lopez-Valdaliso R, Martin D, Alvarez mediating insulin resistance in human muscle cells.
A, Cuadrado A (2000) Inhibition of PKB/Akt1 J Biol Chem 282:12583–12589
by C2-ceramide involves activation of ceramide- 149. Turpin SM, Lancaster GI, Darby I, Febbraio MA,
activated protein phosphatase in PC12 cells. Mol Watt MJ (2006) Apoptosis in skeletal muscle
Cell Neurosci 15:156–169 myotubes is induced by ceramides and is posi-
138. Schubert KM, Scheid MP, Duronio V (2000) tively related to insulin resistance. Am J Physiol
Ceramide inhibits protein kinase B/Akt by promot- Endocrinol Metab 291:E1341–E1350
ing dephosphorylation of serine 473. J Biol Chem 150. JeBailey L, Wanono O, Niu W, Roessler J, Rudich
275:13330–13335 A, Klip A (2007) Ceramide- and oxidant-induced
139. Frangioudakis G, Diakanastasis B, Liao BQ, Saville insulin resistance involve loss of insulin-dependent
JT, Hoffman NJ, Mitchell TW, Schmitz-Peiffer C Rac-activation and actin remodeling in muscle cells.
(2013) Ceramide accumulation in L6 skeletal mus- Diabetes 56:394–403
cle cells due to increased activity of ceramide syn- 151. Bruce CR, Risis S, Babb JR, Yang C, Kowalski GM,
thase isoforms has opposing effects on insulin action Selathurai A, Lee-Young RS, Weir JM, Yoshioka
to those caused by palmitate treatment. Diabetologia K, Takuwa Y, Meikle PJ, Pitson SM, Febbraio MA
56:2697–2701 (2012) Overexpression of sphingosine kinase 1
140. Stratford S, Hoehn KL, Liu F, Summers SA (2004) prevents ceramide accumulation and ameliorates
Regulation of insulin action by ceramide: dual muscle insulin resistance in high-fat diet-fed mice.
mechanisms linking ceramide accumulation to the Diabetes 61:3148–3155
inhibition of Akt/protein kinase B. J Biol Chem 152. Lahiri S, Park H, Laviad EL, Lu X, Bittman R,
279:36608–36615 Futerman AH (2009) Ceramide synthesis is modu-
141. Teruel T, Hernandez R, Lorenzo M (2001) Ceramide lated by the sphingosine analog FTY720 via a mix-
mediates insulin resistance by tumor necrosis ture of uncompetitive and noncompetitive inhibition
factor-alpha in brown adipocytes by maintaining in an acyl-CoA chain length-dependent manner.
Akt in an inactive dephosphorylated state. Diabetes J Biol Chem 284:16090–16098
50:2563–2571 153. Bruce CR, Risis S, Babb JR, Yang C, Lee-Young
142. Chen CL, Lin CF, Chang WT, Huang WC, Teng CF, RS, Henstridge DC, Febbraio MA (2013) The
Lin YS (2008) Ceramide induces p38 MAPK and sphingosine- 1-phosphate analog FTY720 reduces
JNK activation through a mechanism involving a muscle ceramide content and improves glucose tol-
thioredoxin-interacting protein-mediated pathway. erance in high fat-fed male mice. Endocrinology
Blood 111:4365–4374 154:65–76
143. Hage Hassan R, Pacheco de Sousa AC, Mahfouz 154. Ussher JR, Koves TR, Cadete VJ, Zhang L, Jaswal
R, Hainault I, Blachnio-Zabielska A, Bourron O, JS, Swyrd SJ, Lopaschuk DG, Proctor SD, Keung
Koskas F, Gorski J, Ferre P, Foufelle F, Hajduch E W, Muoio DM, Lopaschuk GD (2010) Inhibition of
(2016) Sustained action of ceramide on THE insu- de novo ceramide synthesis reverses diet-induced
lin signaling pathway in muscle cells: implication of insulin resistance and enhances whole-body oxygen
the double-stranded RNA-activated protein kinase. consumption. Diabetes 59:2453–2464
J Biol Chem 291:3019–3029
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 189
155. Raichur S, Wang ST, Chan PW, Li Y, Ching J, sphingomyelinase- deficient mice. Eur J Immunol
Chaurasia B, Dogra S, Ohman MK, Takeda K, 28:874–880
Sugii S, Pewzner-Jung Y, Futerman AH, Summers 166. Bai A, Kokkotou E, Zheng Y, Robson SC (2015)
SA (2014) CerS2 haploinsufficiency inhibits beta- Role of acid sphingomyelinase bioactivity in human
oxidation and confers susceptibility to diet-induced CD4+ T-cell activation and immune responses. Cell
steatohepatitis and insulin resistance. Cell Metab Death Dis 6:e1828
20:687–695 167. Manoochehri Khoshinani H, Afshar S, Najafi R
156. Turpin SM, Nicholls HT, Willmes DM, Mourier (2016) Hypoxia: A double-edged sword in cancer
A, Brodesser S, Wunderlich CM, Mauer J, Xu E, therapy. Cancer Investig 34:536–545
Hammerschmidt P, Bronneke HS, Trifunovic A, 168. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen
LoSasso G, Wunderlich FT, Kornfeld JW, Bluher AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth
M, Kronke M, Bruning JC (2014) Obesity-induced SC, Phillips GM, Cline GW, Phillips AJ, Medzhitov
CerS6-dependent C16:0 ceramide production pro- R (2014) Functional polarization of tumour-
motes weight gain and glucose intolerance. Cell associated macrophages by tumour-derived lactic
Metab 20:678–686 acid. Nature 513:559–563
157. Stiban J, Perera M (2015) Very long chain cerami- 169. Renner K, Singer K, Koehl GE, Geissler EK, Peter
des interfere with C16-ceramide-induced channel K, Siska PJ, Kreutz M (2017) Metabolic hallmarks
formation: A plausible mechanism for regulating the of tumor and immune cells in the tumor microenvi-
initiation of intrinsic apoptosis. Biochim Biophys ronment. Front Immunol 8:248
Acta 1848:561–567 170. Gude DR, Alvarez SE, Paugh SW, Mitra P, Yu J,
158. Holland WL, Bikman BT, Wang LP, Yuguang
Griffiths R, Barbour SE, Milstien S, Spiegel S
G, Sargent KM, Bulchand S, Knotts TA, Shui G, (2008) Apoptosis induces expression of sphingosine
Clegg DJ, Wenk MR, Pagliassotti MJ, Scherer kinase 1 to release sphingosine-1-phosphate as a
PE, Summers SA (2011) Lipid-induced insulin “come-and-get-me” signal. FASEB J 22:2629–2638
resistance mediated by the proinflammatory recep- 171. Weigert A, Johann AM, von Knethen A, Schmidt
tor TLR4 requires saturated fatty acid-induced H, Geisslinger G, Brune B (2006) Apoptotic cells
ceramide biosynthesis in mice. J Clin Invest promote macrophage survival by releasing the anti-
121:1858–1870 apoptotic mediator sphingosine-1-phosphate. Blood
159. Galbo T, Perry RJ, Jurczak MJ, Camporez JP, Alves 108:1635–1642
TC, Kahn M, Guigni BA, Serr J, Zhang D, Bhanot S, 172. Fadok VA, Bratton DL, Konowal A, Freed PW,
Samuel VT, Shulman GI (2013) Saturated and unsat- Westcott JY, Henson PM (1998) Macrophages that
urated fat induce hepatic insulin resistance indepen- have ingested apoptotic cells in vitro inhibit proin-
dently of TLR-4 signaling and ceramide synthesis flammatory cytokine production through autocrine/
in vivo. Proc Natl Acad Sci U S A 110:12780–12785 paracrine mechanisms involving TGF-beta, PGE2,
160. Vandanmagsar B, Youm YH, Ravussin A, Galgani and PAF. J Clin Invest 101:890–898
JE, Stadler K, Mynatt RL, Ravussin E, Stephens 173. Gordon S (2003) Alternative activation of macro-
JM, Dixit VD (2011) The NLRP3 inflammasome phages. Nat Rev Immunol 3:23–35
instigates obesity-induced inflammation and insulin 174. Hughes JE, Srinivasan S, Lynch KR, Proia RL,
resistance. Nat Med 17:179–188 Ferdek P, Hedrick CC (2008) Sphingosine-1-
161. Ferrara JLM, Chaudhry MS (2018) GVHD: biology phosphate induces an antiinflammatory phenotype
matters. Blood advances 2:3411–3417 in macrophages. Circ Res 102:950–958
162. Rotolo JA, Stancevic B, Lu SX, Zhang J, Suh D, 175. Weigert A, Tzieply N, von Knethen A, Johann
King CG, Kappel LW, Murphy GF, Liu C, Fuks Z, AM, Schmidt H, Geisslinger G, Brune B (2007)
van den Brink MR, Kolesnick R (2009) Cytolytic T Tumor cell apoptosis polarizes macrophages
cells induce ceramide-rich platforms in target cell role of sphingosine- 1-phosphate. Mol Biol Cell
membranes to initiate graft-versus-host disease. 18:3810–3819
Blood 114:3693–3706 176. Weigert A, Schiffmann S, Sekar D, Ley S, Menrad
163. Sofi MH, Heinrichs J, Dany M, Nguyen H, Dai H, Werno C, Grosch S, Geisslinger G, Brune B
M, Bastian D, Schutt S, Wu Y, Daenthanasanmak (2009) Sphingosine kinase 2 deficient tumor xeno-
A, Gencer S, Zivkovic A, Szulc Z, Stark H, Liu C, grafts show impaired growth and fail to polarize
Chang YJ, Ogretmen B, Yu XZ (2017) Ceramide macrophages towards an anti-inflammatory pheno-
synthesis regulates T cell activity and GVHD devel- type. Int J Cancer 125:2114–2121
opment. JCI Insight 2 177. Li G, Liu D, Kimchi ET, Kaifi JT, Qi X, Manjunath
164. Schiffmann S, Hartmann D, Fuchs S, Birod K, Y, Liu X, Deering T, Avella DM, Fox T, Rockey DC,
Ferreiros N, Schreiber Y, Zivkovic A, Geisslinger Schell TD, Kester M, Staveley-O’Carroll KF (2018)
G, Grosch S, Stark H (2012b) Inhibitors of specific Nanoliposome C6-ceramide increases the anti-tumor
ceramide synthases. Biochimie 94:558–565 immune response and slows growth of liver tumors
165. Stoffel B, Bauer P, Nix M, Deres K, Stoffel W in mice. Gastroenterology 154:1024–1036.e1029
(1998) Ceramide-independent CD28 and TCR sig- 178. Liu F, Li X, Lu C, Bai A, Bielawski J, Bielawska
naling but reduced IL-2 secretion in T cells of acid A, Marshall B, Schoenlein PV, Lebedyeva IO, Liu
190 S. Albeituni and J. Stiban
K (2016) Ceramide activates lysosomal cathepsin B 189. Brinkmann V, Davis MD, Heise CE, Albert R,
and cathepsin D to attenuate autophagy and induces Cottens S, Hof R, Bruns C, Prieschl E, Baumruker
ER stress to suppress myeloid-derived suppressor T, Hiestand P, Foster CA, Zollinger M, Lynch KR
cells. Oncotarget 7:83907–83925 (2002) The immune modulator FTY720 targets
179. Hollmann C, Werner S, Avota E, Reuter D, Japtok sphingosine 1-phosphate receptors. J Biol Chem
L, Kleuser B, Gulbins E, Becker KA, Schneider- 277:21453–21457
Schaulies J, Beyersdorf N (2016) Inhibition of 190. Matloubian M, Lo CG, Cinamon G, Lesneski MJ,
acid Sphingomyelinase allows for selective target- Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster
ing of CD4+ conventional versus Foxp3+ regula- JG (2004) Lymphocyte egress from thymus and
tory T cells. J Immunol (Baltimore, Md: 1950) peripheral lymphoid organs is dependent on S1P
197:3130–3141 receptor 1. Nature 427:355–360
180. Zhou Y, Salker MS, Walker B, Munzer P, Borst O, 191. Mandala S, Hajdu R, Bergstrom J, Quackenbush
Gawaz M, Gulbins E, Singh Y, Lang F (2016) Acid E, Xie J, Milligan J, Thornton R, Shei GJ, Card
Sphingomyelinase (ASM) is a negative regulator of D, Keohane C, Rosenbach M, Hale J, Lynch CL,
regulatory T cell (Treg) development. Cell Physiol Rupprecht K, Parsons W, Rosen H (2002) Alteration
Biochem 39:985–995 of lymphocyte trafficking by sphingosine-1-
181. Reich DS, Lucchinetti CF, Calabresi PA (2018) phosphate receptor agonists. Science (New York,
Multiple Sclerosis. N Engl J Med 378:169–180 NY) 296:346–349
182. Schiffmann S, Ferreiros N, Birod K, Eberle M, 192. Baumgart DC, Sandborn WJ (2012) Crohn’s disease.
Schreiber Y, Pfeilschifter W, Ziemann U, Pierre Lancet (London, England) 380:1590–1605
S, Scholich K, Grosch S, Geisslinger G (2012a) 193. Danese S, Fiocchi C (2011) Ulcerative colitis. N
Ceramide synthase 6 plays a critical role in the Engl J Med 365:1713–1725
development of experimental autoimmune enceph- 194. De Palma C, Meacci E, Perrotta C, Bruni P, Clementi
alomyelitis. J Immunol (Baltimore, Md: 1950) E (2006) Endothelial nitric oxide synthase activation
188:5723–5733 by tumor necrosis factor alpha through neutral sphin-
183. Eberle M, Ebel P, Wegner MS, Mannich J, gomyelinase 2, sphingosine kinase 1, and sphingo-
Tafferner N, Ferreiros N, Birod K, Schreiber Y, sine 1 phosphate receptors: a novel pathway relevant
Krishnamoorthy G, Willecke K, Geisslinger G, to the pathophysiology of endothelium. Arterioscler
Grosch S, Schiffmann S (2014) Regulation of Thromb Vasc Biol 26:99–105
ceramide synthase 6 in a spontaneous experimental 195. Pettus BJ, Bielawski J, Porcelli AM, Reames DL,
autoimmune encephalomyelitis model is sex depen- Johnson KR, Morrow J, Chalfant CE, Obeid LM,
dent. Biochem Pharmacol 92:326–335 Hannun YA (2003) The sphingosine kinase 1/
184. Eberle M, Ebel P, Mayer CA, Barthelmes J, Tafferner sphingosine-1-phosphate pathway mediates COX-2
N, Ferreiros N, Ulshofer T, Henke M, Foerch C, de induction and PGE2 production in response to TNF-
Bazo AM, Grosch S, Geisslinger G, Willecke K, alpha. FASEB J 17:1411–1421
Schiffmann S (2015) Exacerbation of experimental 196. Breese EJ, Michie CA, Nicholls SW, Murch
autoimmune encephalomyelitis in ceramide syn- SH, Williams CB, Domizio P, Walker-Smith JA,
thase 6 knockout mice is associated with enhanced MacDonald TT (1994) Tumor necrosis factor alpha-
activation/migration of neutrophils. Immunol Cell producing cells in the intestinal mucosa of children
Biol 93:825–836 with inflammatory bowel disease. Gastroenterology
185. Becker KA, Halmer R, Davies L, Henry BD, Ziobro- 106:1455–1466
Henry R, Decker Y, Liu Y, Gulbins E, Fassbender K, 197. Ngo B, Farrell CP, Barr M, Wolov K, Bailey R,
Walter S (2017) Blockade of experimental multiple Mullin JM, Thornton JJ (2010) Tumor necrosis fac-
sclerosis by inhibition of the acid Sphingomyelinase/ tor blockade for treatment of inflammatory bowel
ceramide system. Neurosignals 25:88–97 disease: efficacy and safety. Curr Mol Pharmacol
186. Sharma S, Mathur AG, Pradhan S, Singh DB, Gupta 3:145–152
S (2011) Fingolimod (FTY720): first approved 198. Popivanova BK, Kitamura K, Wu Y, Kondo T,
oral therapy for multiple sclerosis. J Pharmacol Kagaya T, Kaneko S, Oshima M, Fujii C, Mukaida
Pharmacother 2:49–51 N (2008) Blocking TNF-alpha in mice reduces
187. Singh M, Cugati G, Singh P, Singh AK (2011) colorectal carcinogenesis associated with chronic
Fingolimod: the first oral drug approved by food and colitis. J Clin Invest 118:560–570
drug administration; A breakthrough in treatment of 199. Spiegel S, Foster D, Kolesnick R (1996) Signal
multiple sclerosis. J Pharm Bioallied Sci 3:460–461 transduction through lipid second messengers. Curr
188. Brinkmann V, Cyster JG, Hla T (2004) FTY720: Opin Cell Biol 8:159–167
sphingosine 1-phosphate receptor-1 in the control of 200. Sakata A, Ochiai T, Shimeno H, Hikishima S,
lymphocyte egress and endothelial barrier function. Yokomatsu T, Shibuya S, Toda A, Eyanagi R,
Am J Transplant Off J Am Soc Transplant Am Soc Soeda S (2007) Acid sphingomyelinase inhibition
Transplant Surg 4:1019–1025 suppresses lipopolysaccharide-mediated release
of inflammatory cytokines from macrophages and
15 Roles of Ceramides and Other Sphingolipids in Immune Cell Function and Inflammation 191
protects against disease pathology in dextran sul- for sphingosine kinase 1 in dextran sulfate sodium-
phate sodium-induced colitis in mice. Immunology induced colitis. FASEB J 23:143–152
122:54–64 206. Danese S, Furfaro F, Vetrano S (2018) Targeting
201. Daniel C, Sartory N, Zahn N, Geisslinger G, S1P in inflammatory bowel disease: new avenues for
Radeke HH, Stein JM (2007) FTY720 ameliorates modulating intestinal leukocyte migration. J Crohns
Th1-mediated colitis in mice by directly affect- Colitis 12:S678–s686
ing the functional activity of CD4+CD25+ regu- 207. Peyrin-Biroulet L, Christopher R, Behan D, Lassen
latory T cells. J Immunol (Baltimore, Md: 1950) C (2017) Modulation of sphingosine-1-phosphate
178:2458–2468 in inflammatory bowel disease. Autoimmun Rev
202. Song J, Matsuda C, Kai Y, Nishida T, Nakajima K, 16:495–503
Mizushima T, Kinoshita M, Yasue T, Sawa Y, Ito 208. Kim YR, Volpert G, Shin KO, Kim SY, Shin SH,
T (2008) A novel sphingosine 1-phosphate recep- Lee Y, Sung SH, Lee YM, Ahn JH, Pewzner-
tor agonist, 2-amino-2-propanediol hydrochloride Jung Y, Park WJ, Futerman AH, Park JW (2017)
(KRP-203), regulates chronic colitis in interleu- Ablation of ceramide synthase 2 exacerbates dex-
kin-10 gene-deficient mice. J Pharmacol Exp Ther tran sodium sulphate-induced colitis in mice due to
324:276–283 increased intestinal permeability. J Cell Mol Med
203. Maines LW, Fitzpatrick LR, French KJ, Zhuang 21:3565–3578
Y, Xia Z, Keller SN, Upson JJ, Smith CD (2008) 209. Oertel S, Scholich K, Weigert A, Thomas D,
Suppression of ulcerative colitis in mice by orally Schmetzer J, Trautmann S, Wegner MS, Radeke
available inhibitors of sphingosine kinase. Dig Dis HH, Filmann N, Brune B, Geisslinger G, Tegeder
Sci 53:997–1012 I, Grosch S (2017) Ceramide synthase 2 deficiency
204. Pulkoski-Gross MJ, Uys JD, Orr-Gandy KA, Coant aggravates AOM-DSS-induced colitis in mice: role
N, Bialkowska AB, Szulc ZM, Bai A, Bielawska of colon barrier integrity. Cell Mol Life Sci CMLS
A, Townsend DM, Hannun YA, Obeid LM, Snider 74:3039–3055
AJ (2017) Novel sphingosine kinase-1 inhibitor, 210. Helke K, Angel P, Lu P, Garrett-Mayer E, Ogretmen
LCL351, reduces immune responses in murine DSS- B, Drake R, Voelkel-Johnson C (2018) Ceramide
induced colitis. Prostaglandins Other Lipid Mediat synthase 6 deficiency enhances inflammation in the
130:47–56 DSS model of colitis. Sci Rep 8:1627
205. Snider AJ, Kawamori T, Bradshaw SG, Orr KA,
Gilkeson GS, Hannun YA, Obeid LM (2009) A role
Bioactive Lipids in Cancer,
Inflammation and Related 16
Diseases
Acute and Chronic Mild Traumatic Brain
Injury Differentially Changes Levels
of Bioactive Lipids in the CNS Associated
with Headache
Emma Leishman, Phillip E. Kunkler,
Joyce H. Hurley, Sally Miller,
and Heather B. Bradshaw
16.1 T
raumatic Brain Injury, were also neuroprotective in rodent TBI models
Headache, and Endogenous by reducing blood-brain barrier disruption, as
Cannabinoids measured by sodium fluorescein uptake, which
prevented the entry of pro-inflammatory immune
~40 million people worldwide endure a mild cells, and by attenuating microglial activation, as
traumatic brain injury (mTBI; also known as con- measured by immunohistochemistry [21].
cussion) each year [1], accounting for over 75% Cannabinoid receptor activation is hypothesized
of brain injuries [2]. Headache is a common com- to reduce headache pain [4, 18], suggesting con-
plaint after mTBI [2–6]. In studies of TBI patient vergent biochemistry between headache and TBI.
populations in the United States, around 70% of
participants reported having headaches during
the first-year post-injury [3, 5]. Demonstrating 16.2 Lipoamines as Endogenous
that post-mTBI headache is a global phenome- Activators of Transient
non, a study of TBI patients in China found that, Receptor Potential (TRP)
regardless of TBI severity, 58% of patients had Channels
headaches at 3 months post-injury, 54% at
6 months, and 49% at 1 year [6]. In contrast, only TRP channels are ubiquitous ligand-gated ion
12% of the patients reported pre-injury head- channels that serve a variety of functions through-
aches [6]. Often, post-mTBI headaches are out the body [22–31]. There are 28 known TRP
migraine-like [4, 6–9]. For example, in the study channels in mammalian systems; however, they
by Xu and colleagues, around 70% of the head- are arguably understudied to date as their endog-
aches described by TBI patients were classified enous ligands, distributions, and signaling prop-
as migraine-type [6]. Activation of the trigemino- erties are still in question [26, 29, 31]. In the
vascular system (TVS), the trigeminal innerva- context of disease, and more specifically the
tion of cranial vasculature [10], is hypothesized symptom of pain, TRP vanilloid type 1 (TRPV1)
to underlie migraine pain [11]. Sensitization can has been the focus of intense study and may rep-
occur after repeated TVS activation [11]. After resent the TRP channel being the most studied
the initial mTBI, there is a secondary inflamma- overall [23, 27, 28, 31–34]. More than a dozen
tory biochemical insult that occurs days later [12, bioactive lipids have been shown to activate
13]. These biochemical changes may promote TRPV1 [31, 35–40]. Our group recently identi-
headaches through the activity of inflammatory fied 8 of these [41]. Given the ubiquity of TRPs,
mediators, many of which are bioactive lipids, it is not surprising that they appear to have inter-
that are produced during chronic immune connected responses with other TRPs and
responses [4, 8]. GPCRs. A combination of TRPV1-TRP ankyrin
Bioactive lipids such as endogenous cannabi- 1 (TRPA1) activity has been observed in an ani-
noids (eCBs), the endogenous activators of CB1 mal model for headache wherein ablation of
and CB2 cannabinoid receptors [14–17], help TRPV1 in the trigeminal ganglia (TG) inhibited
regulate inflammation [17] and have emerging TVS activation driven by TRPA1 agonists [42].
roles in the TVS [18]. Brain levels of the eCB We recently showed that TRPA1 activation by
2-arachidonoyl glycerol (2-AG) [15, 16] signifi- acrolein drives increases in TRPV1 endogenous
cantly increased in a mouse model of TBI, start- bioactive lipids in the CNS [43].
ing at 4 h post-injury [19]. As 2-AG was Many endogenous TRPV ligands are
neuroprotective after TBI [12, 19, 20], it was lipoamines, which are conjugations of fatty acids
hypothesized that the increase in 2-AG was a and amines [22, 44–46]. The eCB N-arachidonoyl
compensatory mechanism to mitigate the damage ethanolamine (AEA) [14] is a lipoamine that acti-
of pro-inflammatory mediators released after vates TRPV1, as well as cannabinoid receptors
injury [19]. The effects of 2-AG were mostly [35]. Additional lipoamines activate TRPV1 such
mediated by CB1 [19]. However, CB2 agonists as the N-acyl ethanolamines (NAEs) N-oleoyl
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 195
ethanolamine (OEA) [37, 38], N-linoleoyl etha- measured in the acrolein model [54], showing
nolamine (LEA), and N-docosahexaenoyl etha- these very different models have similar effects
nolamine (DEA) [41], and certain lipoamines on measures of headache-like pain in rodents.
derived from arachidonic acid (AA) such as Given that headache driven by acrolein exposure
N-arachidonoyl GABA (A-GABA) [41] and was linked to alterations in bioactive lipids [43],
N-arachidonoyl taurine (A-Taur) [39]. we wanted to know whether headache caused by
N-arachidonoyl serine (A-Ser) was neuroprotec- a very different model, mTBI, would drive simi-
tive in a mouse model of TBI, and these effects lar alterations in lipid levels and whether the
were attenuated with a TRPV1 antagonist, as effects of repeated mTBIs would differ from the
well as a CB2 antagonist [47, 48]. effects of a single mTBI. To test the hypothesis
that acute and repeated mTBI alters the CNS lipi-
dome, we performed targeted lipidomics screens
16.3 B
ioactive Lipid Levels of the TG, TNC and the CER, from rats exposed
Change in a Model to single or repeated mTBI. Analyzing 74
of Headache lipoamine structural analogs, 2-acyl glycerol
structural analogs, prostaglandins (PGs), and free
Showing cross-talk with TRPV1, TRPA1 is fatty acids through targeted high-pressure liquid
expressed in sensory neurons [49]. The TRPA1 chromatography-coupled tandem mass spec-
agonist acrolein is an irritant and a major compo- trometry (HPLC/MS/MS) we were able to deter-
nent of air pollution [50, 51]. Molecular mecha- mine how this lipidome is modified. Both single
nisms of inflammatory pain and hypersensitization and repeated mTBI modified many bioactive lip-
of the TVS caused by irritants like acrolein par- ids including increased levels of the AEA metab-
tially depend on TRPA1 activity [52, 53]. In a rat olite [58] and GPR18 ligand [59], N-arachidonoyl
model of headache driven by exposure to acro- glycine (NAGly), and alterations of TRPV1 and
lein, TVS sensitization [53] and headache-like CB ligands. Increased levels of PGs were associ-
behaviors [54] were correlated with altered levels ated with repeated mTBI. These findings may
of bioactive lipids in the TVS and the cerebellum shed new light on how bioactive lipids in the
(CER), such as eCBs and endogenous lipoamine CNS play a role in headache associated with TBI.
TPRV1 ligands [43]. TVS tissues analyzed were
the TG, where cell bodies of trigeminal nerve
fibers are located [10], and the trigeminal nucleus 16.4 mTBI Protocols, Tissue
caudalis (TNC), part of the brainstem which Collection, and HPLC/MS/MS
receives sensory signals from the TG and proj- Analysis
ects them onto thalamo-cortical pathways [55].
These experiments provided evidence that acro- All animal procedures were approved by the
lein exposure drives changes in CNS lipids, sup- Institutional Animal Care and Use Committee of
porting a working hypothesis that both TRPA1 Indiana University School of Medicine and were
and TRPV1 activity play a role in headache. in accordance with the ethics guidelines set by
Exposure to a single mTBI caused headache- the International Association for the Study of
like symptoms in rats [56, 57]. Specifically, Pain [60]. In total, 24 adult male (170–250 g)
mTBI caused periorbital allodynia, meaning that Sprague-Dawley rats (Harlan, Indianapolis, IN,
stimuli that were non-nociceptive before the USA) were used: 12 were assigned to the single
injury became painful after the injury [56, 57]. mTBI paradigm and 12 were assigned to the
Providing more evidence of alterations in the repeated mTBI paradigm. Rats were housed in
TVS that could support headache post-TBI, lev- pairs with food and water available ad libitum,
els of pro-nociceptive peptides were upregulated and were kept on a standard light/dark cycle.
in the TNC after TBI [56, 57]. Periorbital allo- Using an Impact One stereotaxic impactor (Leica
dynia was a marker of headache-like symptoms Microsystems, Wetzlar, Germany) with a 3 mm
196 E. Leishman et al.
diameter tip, mTBI was induced by 4.0 m/s (MRM) tailored for each group of structurally
impact to 2.0 mm depth on the right hemisphere similar compounds (Fig. 16.1). HPLC/MS/MS
adjacent to bregma and sagittal sutures. This was data was analyzed using Analyst software
performed once on single mTBI rats and per- (Applied Biosystems) as previously described
formed 4 times, with 48 h between each impact, [41, 43, 46, 65–72]. Chromatograms were gener-
on repeated mTBI rats. 6 of the 12 rats in each ated by determining the retention time of ana-
paradigm (single or repeated) received mTBI and lytes from the C18 analytical column with a
the other 6 received a sham treatment. [M−1] or [M + 1] parent peak and a fragmenta-
To better model concussion, the injury was a tion peak corresponding to the programmed val-
closed-head injury and the rats did not undergo ues. Extraction efficiency was calculated with the
craniotomy [61]. Confirming a mild injury, the d8NAGly or d8AEA-spiked recovery vial as a
impact did not damage the skull. As reviewed by standard as done previously [41, 43, 46, 65–72].
Fehily and Fitzgerald, other rodent models of For each individual lipid in the TG, TNC and
repeated mTBI use similar parameters [62]. For CER, concentrations in moles per gram of wet
example, a study by David Brody’s group tissue weight adjusted for percent recovery from
described modeling repeated mTBI in mice by mTBI animals were compared to sham using a
striking the exposed, intact skull with a rubber tip one-way ANOVA. Statistical tests were carried
at 5.0 m/s to a depth of 3.3 mm [63]. Although out using SPSS Statistics (IBM, Armonk, NY,
this injury caused cognitive deficits, lasting USA) and significance was defined as p < 0.05
microglial activation and disruption to white- and trending significance was defined as p < 0.10.
matter axonal cytoskeletal integrity, there was no Using ANOVAs instead of more flexible t-tests
damage to the skull or any readily observable provides more constraints on this type of larger
damage to the underlying brain tissue [63]. data sets.
Bolton and Saatman tested various impact depths As a first level of analysis presented here, the
in a closed-head mouse model of repeated mTBI percentage of the detected lipids affected by sin-
and found that 2.0 mm impact depth did not dam- gle and repeated mTBI in each tissue type was
age the skull but caused gliosis which was wors- calculated. To calculate the percent of the lipids
ened when mTBI was repeated every 24 h [64]. detected that were unaffected by mTBI, the num-
Thus, our parameters are consistent with models ber of changes (p < 0.10) in a tissue type for a
employed by other groups studying mTBI in specific condition (single or repeated) was first
rodents. Rats were sacrificed via decapitation, subtracted from the number of lipids detected in
either 72 h after the single mTBI or 1 week after that tissue (which represents the number of pos-
the last session in repeated mTBI. Brains were sible changes). This number was then divided by
immediately removed. Following removal of the the number of lipids detected and multiplied by
brain, the left TG was removed from the skull and 100. For example, in the TNC in the repeated
frozen on dry ice. The brains were dissected into mTBI condition, there were 73 lipids detected
targeted regions: TNC and CER, and frozen on and 29 of them changed with repeated mTBI,
dry ice. TG, TNC and CER samples were stored meaning that 44 did not change. 44 divided by 73
at −80 °C before being processed for lipid times 100 is 60.27%, so 60.27% of the detected
extraction. lipidome did not change with repeated mTBI in
Tissue extracts were performed as previously the TNC. This percentage can be viewed in the
described [41, 43, 46, 65–72]. Lipids were ana- left, blue columns of Fig. 16.2. The next portion
lyzed using an Applied Biosystems API 3000 of Fig. 16.2 (middle, green) gives the percentage
triple quadrupole mass spectrometer with elec- of the lipids detected in each tissue that increased
trospray ionization (Foster City, CA, USA) as in mTBI. Sticking with the repeated mTBI TNC,
previously described [43]. Levels of each com- of the 29 changes that were detected, 14 of them
pound were determined by running each sample were increases. 14 is then divided by the number
using a multiple-reactions monitoring method of lipids detected (73 in this case) and multiplied
N-acyl alanine [M - H]- Fragment N-acyl proline [M - H]- Fragment
N-palmitoyl alanine 326.5 88.09 N-palmitoyl proline 352.53 114.12
N-stearoyl alanine 354.55 88.09 N-stearoyl proline 380.59 114.12
N-oleoyl alanine 352.53 88.09 N-oleoyl proline 378.31 114.12
N-linoleoyl alanine 350.52 88.09 N-linoleoyl proline 376.56 114.12
N-arachidonoyl alanine 374.5 88.09 N-arachidonoyl proline 400.58 114.12
N-docosahexaenoyl alanine 398.56 88.09 N-docosahexaenoyl proline 424.6 114.12
N-acyl dopamine [M - H]- Fragment N-acyl serine [M - H]- Fragment
N-oleoyl dopamine 416.3 123.2 N-palmitoyl serine 342.3 74
N-arachidonoyl dopamine 438.4 123.2 N-stearoyl serine 370.3 74
N-acyl ethanolamine [M + H]+ Fragment N-oleoyl serine 368.3 74
N-palmitoyl ethanolamine 300.29 62.1 N-linoleoyl serine 366.27 74
N-stearoyl ethanolamine 328.3 62.1 N-arachidonoyl serine 390.3 74
N-oleoyl ethanolamine 326.3 62.1 N-docosahexaenoyl serine 414.3 74
N-linoleoyl ethanolamine 324.3 62.1 N-acyl taurine [M - H]- Fragment
N-arachidonoyl ethanolamine 348.3 62.1 N-palmitoyl taurine 362.6 124
N-docosahexaenoyl ethanolamine 372.6 62.1 N-stearoyl taurine 390.6 124
N-acyl GABA [M - H]- Fragment N-oleoyl taurine 388.6 124
N-palmitoyl GABA 340.54 102.1 N-arachidonoyl taurine 410.6 124
N-stearoyl GABA 368.58 102.1 N-acyl tryptophan [M - H]- Fragment
N-oleoyl GABA 366.57 102.1 N-palmitoyl tryptophan 441.63 203.1
N-linoleoyl GABA 364.54 102.1 N-stearoyl tryptophan 469.68 203.1
N-arachidonoyl GABA 388.57 102.1 N-oleoyl tryptophan 467.67 203.1
N-docosahexaenoyl GABA 412.59 102.1 N-linoleoyl tryptophan 465.65 203.1
N-acyl glycine [M - H]- Fragment N-arachidonoyl tryptophan 489.67 203.1
N-palmitoyl glycine 312.26 74.2 N-docosahexaenoyl tryptophan 513.69 203.1
N-stearoyl glycine 340.3 74.2 N-acyl tyrosine [M - H]- Fragment
N-oleoyl glycine 338.3 74.2 N-palmitoyl tyrosine 418.59 180.18
N-linoleoyl glycine 336.3 74.2 N-stearoyl tyrosine 446.65 180.18
N-arachidonoyl glycine 360.3 74.2 N-oleoyl tyrosine 444.63 180.18
N-docosahexaenoyl glycine 384.3 74.2 N-linoleoyl tyrosine 442.61 180.18
N-acyl leucine [M - H]- Fragment N-arachidonoyl tyrosine 466 180.18
N-palmitoyl leucine 368.58 130.1 N-docosahexaenoyl tyrosine 490.66 180.18
N-stearoyl leucine 396.63 130.1 N-acyl valine [M - H]- Fragment
N-oleoyl leucine 394.61 130.1 N-palmitoyl valine 354.31 116.31
N-linoleoyl leucine 392.6 130.1 N- stearoyl valine 382.6 116.14
N-docosahexaenoyl leucine 440.64 130.1 N-oleoyl valine 380.59 116.14
N-acyl methionine [M - H]- Fragment N-linoleoyl valine 378.58 116.14
N-palmitoyl methionine 386.62 148.2 N-docosahexaenoyl valine 426.62 116.14
N-stearoyl methionine 414.64 148.2 2-acyl glycerol [M + H]+ Fragment
N-oleoyl methionine 412.65 148.2 2-palmitoyl glycerol 331.5 239.5
N-linoleoyl methionine 410.64 148.2 2-oleoyl glycerol 357.5 265.2
N-arachidonoyl methionine 434.66 148.2 2-linoleoyl glycerol 355.5 245
N-docosahexaenoyl methionine 458.68 148.2 2-arachidonoyl glycerol 379.3 287.5
N-acyl phenylalanine [M - H]- Fragment Free Fatty Acids [M - H]- Fragment
N-palmitoyl phenylalanine 402.59 164.1 Oleic acid 281.5 263
N-stearoyl phenylalanine 430.65 164.1 Linoleic acid 279.5 261
N-oleoyl phenylalanine 428.63 164.1 Arachidonic acid 303.5 285
N-linoleoyl phenylalanine 426.61 164.1 PhosphoNAEs [M - H]- Fragment
N-arachidonoyl phenylalanine 450.64 164.1 PhosphoLEA 403.5 58.5
N-docosahexaenoyl phenylalanine 474.66 164.1 Prostaglandins [M - H]- Fragment
PGE2 351.2 315
PGF2α 353.3 309.2
6-keto-PGF1α 369.3 206.9
Fig. 16.1 Lipids in HPLC/MS/MS screening library parent ion is the only ion allowed to pass through the first
with parent ion and fragment ion masses. Lipids are quadrupole of the API3000. The parent ion is then frag-
grouped by amide family and all members of that lipid mented into the collision chamber and an abundant frag-
family are screened in a multiple reactions monitoring ment can be selected as the fragment ion. Only the
(MRM) method. The parent ion mass is also listed: nega- selected fragment ion can pass from the collision chamber
tive ionization mode, resulting in a [M – H]− parent ion, is into the third quadrupole. Therefore, unknown lipids are
used for all methods except the N-acyl ethanolamine and matched to known standards according to retention time
2-acyl glycerol methods, which uses positive ionization from the analytical column and according to their mass
and generates a parent ion with a mass of [M + H]+. The fingerprint
198 E. Leishman et al.
Fig. 16.2 Summary of effects of single and repeated or repeated mTBI. The middle, green column represents
mild traumatic brain injury (mTBI) on lipid levels in the percentage of lipids detected that increased with single
the rat cerebellum, trigeminal ganglia, and trigeminal and repeated mTBI and the right, orange column repre-
nucleus caudalis. The left, blue column represents the sents the percentage of lipids detected that decreased with
percentage of the lipids detected within each of tissue type single and repeated mTBI relative to sham. See Methods
(row) with concentrations that were unaffected by single for more details on how percentages were calculated
by 100 to yield the percentage increased Immortalized BV2 microglia cells were a gift
(19.18%). The right portion of Fig. 16.2 shows from Dr. N. Stella at University of Washington,
the percentage of the lipids detected that Seattle. Boyden migration chamber assays and
decreased with single or repeated mTBI. The per- analyses were performed identically to those pre-
centage was calculated by taking the number of viously described in our laboratory [59, 73].
lipids with decreased concentrations in each tis-
sue and dividing it by the number of lipids
detected in that tissue. For the TNC, 15 lipids 16.4.1 mTBI Has Broad Effects
decreased with repeated mTBI, giving a percent- on the CER, TG, and TNC
age of 20.55% for the proportion of the lipidome Lipidome
that decreased. When the 3 different percentages
for a tissue are added, they should equal 100. For Of the 74 lipoamines in our screening library
the repeated mTBI TNC, 60.27 plus 19.18 plus (Fig. 16.1), over 50 were detected in the TG,
20.55 equals 100. TNC, and CER. If a lipid was detected in the
Analyzed lipidomics data from the TG, TNC, samples from injured rats it was also found in the
and CER are represented in tabular format illus- corresponding sham samples. In both single and
trating both the direction and magnitude of change. repeated mTBI, 78 lipids were detected in the
For example, the mean level of AEA was CER (92% of the 85 lipids in the screening
9.20 × 10−11 moles per gram in the CER of the rats library). For the TG and TNC, different numbers
subjected to a single mTBI and 7.86 × 10−11 moles of lipids were detected after single and repeated
per gram in the corresponding sham CER. mTBI. In the TG, 80 lipids were detected in the
9.20 × 10−11 divided by 7.86 × 10−11 is 1.17, assign- single condition (94%) and 77 in the repeated
ing it 1 up arrow in the figures because the magni- condition (91%). Fewer lipids were detected in
tude of change was between 1 and 1.5 times higher the TNC (single: 76 lipids – 89%; repeated: 73
than sham. Providing an example of a decrease, lipids – 86%). Tables listing the lipids detected
the average level of AEA in the TNC of rats and their mean levels in each tissue type in sham
exposed to repeated mTBI was 5.22 × 10−12 moles and mTBI tissues are available upon request from
per gram and the average level of AEA in the TNC the authors.
of the sham rats was 7.32 × 10−12 moles per gram; The first level of analysis examined the pro-
5.22 × 10−12 divided by 7.32 × 10−12 equals 0.71, portion of the lipids detected that were
and the reciprocal of 0.71 is 1.41, meaning that the unchanged, upregulated, or downregulated with
decrease is between 1 and 1.5 times sham levels mTBI relative to sham. Effects of single mTBI
and giving it 1 down arrow on our scale. on the lipidome were mostly tissue-dependent
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 199
(Figs. 16.2, 16.3, 16.4, and 16.5). The highest increases in a lipid’s concentration. In contrast,
percentage of changes in lipid levels occurred in changes measured in the TG included decreases
the CER (36 lipids – 46% of the lipids detected), (15 increases, 3 decreases).
followed by the TNC (28 lipids – 37%). The area Repeated mTBI affected a similar number of
with the lowest percentages of changes in lipid the detected lipids (Figs. 16.2, 16.3, 16.4, and
levels was the TG (18 lipids – 22.5%). All of the 16.5). However, the specific tissue most affected
changes driven in the TNC and CER were shifted in repeated mTBI. The highest percent-
Fig. 16.3 Comparison of effects of single or repeated colors represent increases, whereas orange colors repre-
mild traumatic brain injury (mTBI) on levels of N- sent decreases. Darker colors represent significant
acyl ethanolamines, N-acyl glycines, N-acyl taurines, changes of p < 0.05 and lighter colors represent trending
2-acyl glycerols, free fatty acids, and prostaglandins. changes of p < 0.10, as determined by one-way
Levels of targeted bioactive lipids in the cerebellum, tri- ANOVA. The number of arrows indicates the magnitude
geminal ganglia, and trigeminal nucleus caudalis of rats of the difference between mTBI and sham. One arrow
given a single mTBI (mTBI × 1) or 4 repeated mTBIs indicates a magnitude difference of less than 1.5 fold
(mTBI × 4) were compared to respective sham rats. and 2 arrows indicate a 1.5–1.99 fold change. A blank
Cells with shaded arrows indicate a significant or trend- cell indicates that there was no change in the lipid’s level
ing change in that lipid with mTBI. The arrow color due to mTBI. See Methods for more detailed description
indicates the direction of change relative to sham. Green of analysis
200 E. Leishman et al.
Fig. 16.4 Comparison of effects of single or repeated geminal nucleus caudalis (TNC) of rats exposed to a sin-
mild traumatic brain injury (mTBI) on levels of N- gle mild traumatic brain injury (single mTBI – black bars)
docosahexaenoyl ethanolamine (DEA) and N- or sham (open bars) and of rats exposed to a series of 4
arachidonoyl glycine (NAGly). Panel A shows bar mTBIs (repeated mTBI – black bars) or sham (open bars).
graphs for mean levels in moles per gram tissue of DEA in Error bars are ± standard error of the mean. Asterisk (∗)
the cerebellum (CER), trigeminal ganglia (TG), and tri- indicates a difference between mTBI and sham of p < 0.05
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 201
age of changes occurred in the TNC (29 lipids – mental toxin, acrolein [43]. This means that
40% of the lipids detected), followed by the TG increases in NAGly may serve as a marker of
(28 lipids – 36%). Unlike in single mTBI, headache caused by very different stimuli, mTBI
where the most changes were detected in the and TRPA1 activation. There are multiple path-
CER, in repeated mTBI the CER had the fewest ways that regulate NAGly levels. NAGly is a
changes detected (19 lipids – 24%). The ratio of metabolite of AEA and can be formed from AEA
increases and decreases also shifted, with via 2 enzymatic pathways, one mediated by
decreases occurring in every tissue type. In the alcohol dehydrogenase and the other mediated
TG and CER, most of the changes were by fatty acid amide hydrolase (FAAH) [58].
increases (27 increases of 28 changes for TG Indeed, in FAAH knockout (KO) mice, brain
and 17/19 for CER). The TNC stands out, levels of NAGly are significantly reduced,
because it was the only tissue type where most whereas AEA levels are significantly higher
of the changes were decreases, suggesting func- compared to wild-type (WT) [69]. The concur-
tional specialization in this area that can poten- rent reduction in AEA and other NAEs and
tially underlie a more dynamic response to increase in NAGly in the TNC after repeated
injuries (15/29; Figs. 16.2, 16.3, 16.4, and mTBI could potentially be driven by an increase
16.5). Given that the CER was the most affected in AEA metabolism via FAAH. NAGly produc-
area after a single mTBI, a single mTBI may tion can also be mediated by glycine N-acyl
therefore have a stronger effect on bioactive transferase enzymes [75] and by the newly-char-
lipid production in sites closer to the injury, acterized enzyme peptidase M20 domain con-
such as the CER, whereas the effects of repeated taining 1 (PM20D1) [76, 77]. PM20D1 regulates
mTBI on bioactive lipid levels may spread to mitochondrial uncoupling via regulation of
more distant areas. endogenous lipoamines. As mitochondrial dys-
function is a hallmark of neuroinflammation
after TBI [13], the relationship between
16.4.2 Effects of mTBI on Bioactive PM20D1, TBI, and lipoamine regulation should
Lipids with Relatively More be further investigated.
Well-Studied Biosynthetic All other N-acyl glycine lipids that changed
Pathways and Protein Targets with a single mTBI showed an increase
(Fig. 16.3). After a single mTBI, N-oleoyl gly-
16.4.2.1 N -Acyl Glycine Lipid Species cine (O-Gly) increased in all 3 tissue types, along
Are Upregulated After mTBI with the increase in NAGly, which was still pres-
Levels of NAGly increased in all 6 groups in the ent after repeated mTBI. Alterations in O-Gly
mTBI experiments (Figs. 16.3 and 16.4). have previously been associated with TBI in
Increases in NAGly were also found after rodent models, as O-Gly was upregulated in the
repeated exposure to low levels of the environ- mouse insular cortex 24 h after TBI [78]. Levels
Fig. 16.4 (continued) whereas the hash sign (#) indicates nucleus caudalis (TNC) of rats exposed to a single mild
a difference between mTBI and sham of p < 0.10, as traumatic brain injury (single mTBI – black bars) or sham
determined by one-way ANOVA. After single mTBI, (open bars) and of rats exposed to a series of 4 mTBIs
DEA increased in all 3 tissue types, corresponding to (repeated mTBI – black bars) or sham (open bars). Error
green cells with 1 up arrow in Fig. 16.4. After repeated bars are ± standard error of the mean. Asterisk (∗) indi-
mTBI, DEA decreased in the CER and TNC, correspond- cates a difference between mTBI and sham of p < 0.05
ing to orange cells with 1 down arrow in Fig. 16.4, but whereas the hash sign (#) indicates a difference between
increased in the TG, corresponding to a green cell with 1 mTBI and sham of p < 0.10, as determined by one-way
up arrow in Fig. 16.4. Panel B shows bar graphs for mean ANOVA. In all 3 tissue types, levels of NAGly increased
levels in moles per gram tissue of NAGly in the cerebel- after single and repeated mTBI, corresponding to green
lum (CER), trigeminal ganglia (TG), and trigeminal cells with 1 up arrow in Fig. 16.4
202 E. Leishman et al.
Fig. 16.5 Effects of single and repeated mild trau- tophan, N-acyl tyrosine, and N-acyl valine families of
matic brain injury (mTBI) on additional bioactive bioactive lipids, and effects on phosphoLEA. Levels of
lipids in the cerebellum, trigeminal ganglia, and tri- targeted bioactive lipids in the cerebellum, trigeminal
geminal nucleus. Specifically, this heatmap shows ganglia, and trigeminal nucleus caudalis of rats given a
effects of mTBI on members of the N-acyl alanine, single mTBI (mTBI × 1) or 4 repeated mTBIs (mTBI × 4)
N-acyl GABA, N-acyl leucine, N-acyl methionine, N-acyl were compared to respective sham rats. Only lipids that
phenylalanine, N-acyl proline, N-acyl serine, N-acyl tryp- had significant or trending changes with either single or
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 203
of the TRPV1 agonist N-docosahexaenoyl gly- tained up to 1 week after repeated mTBI suggests
cine (D-Gly) [41] also increased in the CER and broad increases in neuroinflammation [79].
N-stearoyl glycine (S-Gly) also increased in the Microglia can release pro-inflammatory lipid sig-
TG. The TNC had the most changes in N-acyl naling molecules such as PGs [4]. It is possible
glycine levels after a single mTBI, with levels of that the increases in PGs observed after repeated
all 6 N-acyl glycines increasing. After repeated mTBI are a consequence of increased microglial
mTBI, the increase in NAGly was the only activation. Experiments are planned to see if
increase in an N-acyl glycine in the TNC. In the microglia release PGs when incubated with
TNC, S-Gly decreased after repeated mTBI, NAGly.
which was the only mTBI-driven decrease mea- Novel data shown here demonstrates that
sured in N-acyl glycines. S-Gly changed in the NAGly is not the only N-acyl glycine that can
opposite direction in the CER, increasing after induce microglial migration. At 10 μM,
repeated mTBI. D-Gly increased in the TG only N-palmitoyl glycine, S-Gly, O-Gly, N-linoleoyl
(Figs. 16.3 and 16.4). glycine, NAGly, and D-Gly induced migration
in BV-2 microglia (Fig. 16.6a, b). This demon-
16.4.2.2 N AGly and Additional N-Acyl strates how BV2 microglia might not be
Glycine Lipids Drive responding to the identity of the acyl chain but
Microglial Migration are instead sensitive to glycine-containing
NAGly drives dose-dependent migration in BV2 lipoamines. Follow-up studies should examine
microglia via activation of GPR18, which can be how combining these compounds affects
inhibited by A-Ser [59]. A-Ser’s effects on migration, as levels of multiple N-acyl glycines
microglia could underlie some of its reported change with mTBI. Unpublished data from the
neuroprotective actions after mTBI [47, 48] Hurley lab confirmed that there were increased
because microglial migration is understood to be numbers of microglia in the corpus callosum of
a marker of neuroinflammation [47]. Other stud- mTBI rats, suggesting some effects on migra-
ies have shown an increase in microglial activa- tion (personal communication). Highlighting
tion after mTBI, with microglia rapidly migrating potential cross-talk between TRPA1 and
towards damaged tissue [79]. However, the utility microglia, endogenous TRPA1 ligands released
of the microglial response is complex [13]. by microglia in the spinal cord are hypothe-
Initially, microglia may be useful at clearing sized to contribute to central sensitization after
away damaged axonal tissue, which can promote a peripheral nerve injury. Both TRPA1 antago-
regeneration of damaged axons [13]. However, nists and blockers of microglia can reduce cen-
sustained microglial activation does not seem to tral pain hypersensitivity in animal models
be as beneficial and appears to promote inflam- [80]. Therefore, it is worth investigating
mation rather than regeneration [13, 79]. The fact whether TRPA1 antagonists or microglial
that the increase in NAGly was widespread, blockers can similarly reduce headache pain
occurring in sites distant from the injury, and sus- secondary to TBI.
Fig. 16.5 (continued) repeated mTBI in at least 1 tissue magnitude of the difference between mTBI and sham.
type are shown here. Cells with shaded arrows indicate a One arrow indicates a magnitude difference of less than
significant or trending change in that lipid with 1.5 fold, 2 arrows indicate a 1.5–1.99 fold change, and 3
mTBI. The arrow color indicates the direction of change arrows indicate a 2–2.99 fold change. A blank cell indi-
relative to sham. Green colors represent increases, cates that there was no change in the lipid’s level due to
whereas orange colors represent decreases. Darker colors mTBI. BAL stands for below analytical limits and means
represent significant changes of p < 0.05 and lighter col- that lipid was not detected in all samples in that tissue
ors represent trending changes of p < 0.10, as determined type. See Methods for more detailed description of
by one-way ANOVA. The number of arrows indicates the analysis
204 E. Leishman et al.
Fig. 16.6 Cellular migration activity of N-acyl gly- level when compared to vehicle. Panel B: At 10 μM,
cines in BV2 microglial cells. Panel A: At 10 μM, N-arachidonoyl glycine (NAGly), N-linoleoyl glycine
N-docosahexaenoyl glycine (DocGly), N-arachidonoyl (LinGly), N-stearoyl glycine (StrGly), and N-palmitoyl
glycine (NAGly), N-linoleoyl glycine (LinGly), and glycine (PalGly) caused cellular migration that was sig-
N-oleoyl glycine (OlGly) caused cellular migration that nificantly greater than the vehicle control. 4 plates were
was significantly greater than the vehicle control. 7 plates analyzed with 20 repeats for each condition per plate. ∗
were analyzed with 20 repeats for each condition per indicates a mean difference significant at the 0.05 level
plate. ∗ indicates a mean difference significant at the 0.05 when compared to vehicle
marker of inflammation, decreased [81]. In rats effects are due to conversion into pro-resolving
given vehicle, plasma levels of DHA were sig- mediators such as D-series resolvins and neuro-
nificantly reduced [81]. DHA had a protective protectin [81].
effect and decreased the number of amyloid pre- The primary biosynthetic pathways for these
cursor protein-positive and caspase-3 positive DHA-derived lipids, 15-lipoxygenase
(apoptotic) neurons in the brainstem [81]. The (15-LOX) can convert DHA into
same group performed a similar follow-up 17S-hydroperoxydocosahexaenoic acid
study, but here they gave rats DHA for 30 days (17S-H(p)-DHA), whereas cyclooxygenase 2
before the injury instead of after the injury [83]. can convert DHA into the enantiomer,
Similar results were uncovered, with DHA hav- 17R-hydroperoxydocosahexaenoic acid
ing a neuroprotective effect against TBI, sug- (17R-H(p)-DHA) [86]. 5-lipoxygenase
gesting that DHA can also be used as a (5-LOX) converts 17S-H(p)-DHA to resolvin
prophylactic to protect the brain against the con- D1, whereas the same enzyme converts
sequences of injury [83]. 17R-H(p)-DHA into aspirin-triggered resolvin
In another study, rats were given a DHA- D1 [86]. Alternatively, 17S-H(p)-DHA and
supplemented diet or a control diet for 30 days 17R-H(p)-DHA can first be converted into
before receiving a mild fluid percussion injury epoxide intermediates which are then hydro-
[82]. Rats on the control diet experienced learn- lyzed to produce neuroprotection D1 [86].
ing difficulties after the injury, as measured by Resolvin D1 can reduce immune cell infiltra-
the Morris water maze, whereas rats that tion and reduces inflammatory pain [87].
received DHA did not show these deficits [82]. Likewise, neuroprotectin D1 reduces inflam-
Furthermore, injury-related decreases in BDNF, matory pain and can also inhibit pain signals
CREB, and synapsin I, were not measured in via TRPV1 at intrathecal doses of just 0.1–
the DHA-supplemented rats and lower levels of 10 ng [88]. Given that TRPV1 ligands are ele-
oxidized proteins were measured in the hippo- vated after a single mTBI, it is possible that
campus of the DHA-rats, suggesting a reduc- neuroprotectin D1 could reduce TVS sensitiza-
tion in oxidative stress [82]. DHA also appears tion and therefore decreases headache pain.
to protect against inflammation after TBI, as Pro-resolving mediators are currently being
rats that were given daily injections of DHA for investigated as neuroprotective therapies for
21 days showed a reduction in markers of acti- brain injuries [89], as they are highly potent anti-
vated microglia, which were elevated after TBI inflammatory bioactive lipids [90]. Treating mice
[85]. Of the activated microglia that were still with diffuse axonal injury with aspirin-triggered
present in the CNS of the DHA-treated rats, resolvin D1 for 3 days pre-injury and 7 days post-
more of these expressed the anti-inflammatory injury partially rescued the cognitive deficits and
marker CD206 and fewer expressed the pro- motor deficits, as measured by the novel object
inflammatory marker CD16/32 [85]. On the recognition task and the rotorod task [90].
other hand, when rats are raised on a diet low in Neuroprotectin D1 formation increased in the
DHA and other omega-3 fatty acids, they show mouse hippocampus after cerebral artery occlu-
worsened recovery from TBI-related motor def- sion, where it inhibited immune cell infiltration
icits and increased neuronal death compared to and pro-inflammatory gene induction [91].
rats on a control diet, as well as a 70% loss of Neuroprotectin D1 also inhibited cytokine pro-
DHA in the brain [84]. This data is an impor- duction in neuronal cell cultures [91].
tant reminder that diet can influence levels of Highlighting the neuroprotective effect of this
bioactive lipids and that a diet rich in omega-3 bioactive lipid, the infarct volume in the hippo-
polyunsaturated fatty acids can have measur- campus was significantly reduced 48 h after the
able health benefits. Although metabolites of cerebral artery occlusion injury in mice given
DHA were not quantified, the authors hypothe- intracerebroventricular infusions of neuroprotec-
sized that some of DHA’s neuroprotective tin D1 [91].
206 E. Leishman et al.
Decreases in DEA after repeated mTBI could [41] and in a rodent model of headache driven by
potentially be driven by conversion to oxygenated acrolein exposure [43], suggesting that these
metabolites. DEA has been identified as a precursor molecules are sensitive to inflammatory insults.
for several bioactive lipids, which are formed via The actions of multiple NAEs at TRP channels
similar pathways as the biosynthesis of resolvins and may override the analgesic effects of AEA via
neuroprotectin from DHA [92]. For example, incu- CB1 [18], and prolonged exposure to elevated lev-
bating mouse brain homogenates with DEA gener- els of NAEs has been associated with a nocicep-
ated 17- hydroxydocosahexaenoyl ethanolamide tive phenotype in response to capsaicin [66].
(17-HDHEA), 4,17-dihydroxydocosahexaenoyl
ethanolamide, as well as smaller amounts of
7,17-dihydroxydocosahexaenoyl ethanolamide, 16.4.4 Additional Bioactive Lipids
10,17-dihydroxydocosahexaenoyl ethanolamide Effected by mTBI
(10,17-diHDHEA), and 15-hydroxy-16(17)-epoxy-
docosapentaenoyl ethanolamide (15-HEDPEA) 16.4.4.1 N-Acyl Taurines
[92]. 10,17-diHDHEA and 15-HEDPEA were ago- All 4 N-acyl taurines, including the TRPV1 and
nists at CB2 with EC50’s in the low nanomolar range TRPV4 agonist [39] A-Taur, increased in the
and blocked platelet-leukocyte aggregation forma- CER and TNC after a single mTBI. However,
tion in human whole blood at concentrations of just none of the targeted N-acyl taurines changed in
10pM [92]. As CB2 ligands were neuroprotective the TG. Fewer changes in N-acyl taurines were
after TBI [21], it is possible that these DEA-derived measured after repeated mTBI and there were no
lipids have neuroprotective effects via that receptor. longer increases in all 4 N-acyl taurines in the
Highlighting the potency of these resolving lipids, CER and TNC. Instead, N-stearoyl taurine
injections of 15-HEDPEA reduced lung injury after decreased in the CER and A-Taur increased in the
reperfusion at doses as low as 1 μg/mouse [92]. TG, which was the only change in N-acyl tau-
Given that these lipids are so potent, they might be rines associated with mTBI in the TG. A-Taur
bioactive at concentrations below the detection lim- also increased in the TNC, as did N-oleoyl tau-
its of our HPLC/MS/MS system. It might instead be rine (Fig. 16.3) [43].
a more viable strategy to measure the activity of
5-LOX after mTBI and acrolein exposure, as this is 16.4.4.2 2-Acyl Glycerols
a rate-limiting enzyme for the formation of resolvins After a single mTBI, 2-acyl glycerol levels were
from DHA and might also be important for the for- only affected in the TNC. Here, there were
mation of oxygenated metabolites of DEA [93]. increases in the eCB 2-AG, as well as 2-palmitoyl
Indeed, blocking 5-LOX with siRNA in head and glycerol (2-PG) and 2-linoleoyl glycerol (2-LG).
neck squamous cell carcinoma cell lines attenuated This makes the TNC the only tissue where levels
the formation of reactive oxygen species and blocked of both of the eCBs, AEA and 2-AG, were
the ability of DEA to inhibit cell proliferation [93]. affected by a single mTBI. Whether levels of
15-LOX is also an important enzyme to monitor, 2-acyl glycerols increased or decreased after
because it mediates the conversion of DEA to repeated mTBI was dependent on tissue type.
17-hydroperoxydocosahexaenoyl ethanolamide, a Changes in 2-acyl glycerols were increases in the
precursor for the CB2 agonists 10,17-diHDHEA and CER and the TG, but decreases in the TNC. In the
15-HEDPEA, as well as 17-HDHEA [92]. CER, 2-PG and 2-LG increased. In contrast, all 4
The fact that multiple NAEs were altered with 2-acyl glycerols in the screening library signifi-
mTBI is important because there are emergent cantly increased in the TG. Only 1 change in
signaling properties when multiple TRPV1- 2-acyl glycerol levels was measured in the TNC –
ligand NAEs are combined in that the potency is a reduction in 2-oleoyl glycerol (Fig. 16.3).
increased when multiple agonists are present As 2-AG has been shown to be neuroprotec-
[43]. Multiple NAEs also changed in brain tis- tive in multiple TBI studies [12, 19, 20], increases
sues of rats in a model of peripheral inflammation in 2-AG in the TG after repeated mTBI and in the
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 207
TNC after single mTBI may be part of a neuro- or deleted. If effects on the lipidome require
protective and regenerative response that medi- DAGL, then more direct measurements of DAGL
ates some of the moderate spontaneous recovery activity can be performed after mTBI or acrolein
that occurs after TBI [74]. mTBI also impacted exposure. Recently, a fluorescent probe was
structurally analogous lipids to the eCB 2-AG, developed to measure DAGL activity [101]. This
especially in the in the TG, where repeated mTBI probe can be used in activity based protein profil-
increased 4 different 2-acyl glycerols. This impli- ing (ABPP) assays in mouse brain proteasomes
cates eCB system enzymes as potential mediators [102]. By performing ABPP after acute and
of mTBI’s effects on lipid levels because these repeated mTBI and after acute and chronic acro-
enzymes regulate lipid levels beyond eCB sub- lein exposure, the impact on DAGL activity can
strates [69, 70, 94]. A decrease in 2-acyl glycerol be quantified. ABPP can also be employed to
hydrolysis via monoacylglycerol lipase (MAGL) detect off-target serine hydrolase activity [103],
could increase 2-acyl glycerol levels, and we which may reveal a more widespread impact of
have shown that MAGL deletion causes wide- mTBI on enzymes important for maintaining lev-
spread increases in 2-acyl glycerols in the mouse els of eCBs and other bioactive lipids.
brain [69]. However, these increases in 2-acyl
glycerols likely came at the expense of levels of 16.4.4.3 T he Potential Link
AA and PGs [69]. Increases in AA and PGs were Between TRPV1 and 2-Acyl
measured in the TG after repeated mTBI, making Glycerol Lipids
MAGL inhibition an unlikely mechanism. What upstream changes can affect DAG produc-
Diacylglycerol lipases (DAGL) synthesize tion? Phospholipase C (PLC) intracellular path-
2-acyl glycerols from their diacylglycerol (DAG) ways stimulate the release of DAGs from
precursors, so an increase in DAGL activity could membrane phospholipids [104]. An earlier study
upregulate 2-acyl glycerols [95]. The DAGLα demonstrated that TBI increased PLC activity in
isoform appears more important for biosynthesis the cat brain [105]. Activation of TRP receptors
of 2-AG in the brain, whereas the DAGLβ iso- can drive PLC pathways [36]. Indeed, we have
form appears more important in the periphery shown that capsaicin activation of TRPV1
[96]. There is evidence that levels of AA and increases 2-acyl glycerol production [43].
AEA can change in the same direction as 2-AG Increases in levels of multiple TRPV1 ligands
upon a change in DAGL activity. Along with like LEA, AEA, and DEA in the TG after repeated
2-AG levels, AEA levels were downregulated in mTBI could potentially increase PLC signaling,
the cerebellum, striatum, and hippocampus of a leading to an increase in DAG substrates for
line of DAGLα KO mice [97]. Another line of 2-acyl glycerol production. AA itself can also
DAGLα KO had an 80% reduction in whole brain activate PLC pathways [106] and levels of AA
2-AG and AA levels and a 40% reduction in AEA were upregulated after repeated mTBI in the TG.
[98]. A third line of DAGLα KO had reduced lev-
els of 2-AG and AA in the forebrain, but did not 16.4.4.4 Free Fatty Acids and PGs
detect a significant reduction in AEA in this area, After a single mTBI, increases in AA and linoleic
hinting at some regional specificity for the acid (LA) were common to all 3 tissues. Oleic
contribution of DAGLα activity to AEA levels acid (OA) also increased in the CER, meaning
[99]. Reductions in whole-brain 2-AG, AA, and that all 3 free fatty acids screened for increased
AEA also occur when DAGLα is pharmacologi- after single mTBI in this tissue type. After
cally inhibited [100]. It isn’t yet known whether repeated mTBI, it was the TG that had increases
increasing DAGL activity causes concurrent in all 3 free fatty acids. AA and LA increased in
increases in AA and AEA, which occurred in the the TG, but OA did not significantly change in
TG after repeated mTBI. this tissue. The only decrease in free fatty acid
Follow-up studies can determine if the effects levels was restricted to the TNC, where LA
of mTBI are abolished when DAGL is inhibited decreased after repeated mTBI (Fig. 16.3).
208 E. Leishman et al.
PGs were also affected by mTBI. A single have some activity at TRP channels. Many of
mTBI increased levels of 2 different PGs, PGF2α these lipids have little data about them beyond
and 6-ketoPGF1α in the CER. In the TG, PGF2α those generated by our laboratory from which to
increased, whereas 6-ketoPGF1α decreased. This compare; therefore, we will have to rely on those
was the only measured decrease in PG levels. In few reports. The lipids are subdivided into
contrast, 2 PGs increased in the TNC, PGE2 and lipoamine “families” as characterized by the
6-ketoPGF1α. After repeated mTBI, PGF2α and amine moiety and each section is labeled with
6-ketoPGF1α increased in all 3 tissue types. In the this family for ease of referencing those that
CER and TG, PGE2 also increased, such that all 3 might be of particular interest to a reader.
PGs in the screening library increased in the CER
and TG (Fig. 16.3). N-Acyl GABAs N-linoleoyl GABA (L-GABA),
Increases in AA in CNS tissues were mea- A-GABA, and N-docosahexaenoyl GABA
sured after mTBI. AA can be directly released (D-GABA) are ligands at TRPV1 [41]. However,
from phospholipids via phospholipase A2, or D-GABA did not change after single mTBI. After
indirectly via PLC [107]. Levels of AA are ele- single mTBI, change in L-GABA was restricted
vated in the CSF of TBI patients and higher levels to the CER, where it increased along with
of AA 1 week post-injury were correlated with A-GABA and 2 additional N-acyl GABAs.
worse outcomes [108]. Increased levels of AA Changes in N-acyl GABAs were decreases in the
are hypothesized to underlie increases in PGs in TG after single TBI and were restricted to N-acyl
the brain after TBI [105] and here there were GABAs that lack TRPV1 activity. N-palmitoyl
increases in levels of all 3 PGs in the CER and GABA changed in the opposite direction in the
TG after repeated mTBI, where levels of AA also TNC, as it increased after a single mTBI, along
increased. with A-GABA. Fewer N-acyl GABAs changed in
Levels of bioactive lipids are differentially the CER after repeated mTBI, as an increase in
affected by mTBI, mostly increasing after single A-GABA was the only detected change.
and repeated mTBI. However, decreases in bio- Decreases in N-acyl GABAs were no longer
active lipid levels were more common after found in the TG after repeated mTBI and instead
repeated mTBI compared to single mTBI, espe- 2 N-acyl GABAs with saturated acyl moieties
cially in the TNC. After a single mTBI, certain increased. On the other hand, 3 N-acyl GABAs
changes were measured in all 3 tissue types and decreased in the TNC, including D-GABA. This
all of these were increases relative to sham. was the only change in D-GABA associated with
Specifically, DEA, O-Gly, NAGly, LA, and AA mTBI. However, A-GABA increased in the
increased. After repeated mTBI, fewer changes TNC. This means that increases in A-GABA
were common to all 3 tissue types, although were present in the CER and TNC after both sin-
levels of PGs increased in all 3 tissues, along gle and repeated mTBI, but other changes in
with NAGly. Thus, DEA levels changed in every N-acyl GABAs were sensitive to the number of
tissue type and NAGly was upregulated in the mTBIs (Fig. 16.5).
CER, TG, and TNC, after a single or repeated
mTBI (Figs. 16.3 and 16.4).
N-Acyl Prolines Although N-docosahexaenoyl
proline is a TRPV1 antagonist [41], this lipid was
16.4.5 Effects of mTBI on Lipoamine not detected in any tissue type. This is likely to do
Families That Include a TRPV with a poor signal to noise characteristic of our
Ligand mass spectrometric analysis than due to a lack of
endogenous production given that all other com-
The following section summarizes some of the binations are measurable. Even with the poor sig-
effects on the lipids that were measured in these nal to noise of this lipoamine family in general,
models but are lipids that have members that we are still able to measure many of the members
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 209
and showed that those N-acyl prolines only single mTBI increased levels of the TRPV2/4
changed after repeated mTBI. N-oleoyl proline agonist N-palmitoyl tyrosine [41] and A-Tyr. An
increased in the CER and TNC, whereas increase in A-Tyr in the TNC was the only change
N-arachidonoyl proline (A-Pro) increased in the in N-acyl tyrosines measured after repeated
TG, which was the only tissue where it was mTBI (Fig. 16.5).
detected in all samples (Fig. 16.5).
as high in the mTBI CER and TG compared to mTBI. Specifically, repeated mTBI increased
sham. In contrast, none of the N-acyl alanines phosphoLEA in the CER and TG (Fig. 16.5) [43].
changed in the TG after repeated mTBI and only Interestingly, these increases did not always
2 changed in the CER. In the repeated condition, accompany an increase in LEA.
the TNC had the most changes, with increases in
4 N-acyl alanines, including A-Ala, and with a Multiple orphan lipids, which lack molecular
decrease in N-stearoyl alanine (Fig. 16.5). targets, change with mTBI suggesting that a
wider variety of signaling systems may be
affected. Those orphan lipids that stand out
N-Acyl Leucines N-acyl leucines only changed include A-Phe, which increased in all 3 tissue
after exposure to 4 mTBIs and did not change after types after a single mTBI. mTBI affected certain
a single mTBI. N-linoleoyl leucine and lipoamines derived from AA, both orphan and
N-docosahexaenoyl leucine increased in the CER, with known molecular targets. For example,
whereas N-palmitoyl leucine and N-stearoyl leu- A-Ala robustly increased in the CER and TG fol-
cine (S-Leu) increased in the TG. In contrast, lowing a single mTBI, A-Met increased in the
S-Leu decreased in the TNC, as did N-oleoyl leu- TNC after repeated mTBI, and A-Pro increased
cine (Fig. 16.5). in the TG after repeated mTBI. In all 3 tissue
types, a single exposure to mTBI increased levels
of A-Phe. Increases in A-Phe were also found in
N-Acyl Methionines Alterations in N-acyl the TG and TNC after repeated mTBI. mTBI
methionines were restricted to the CER and TG might therefore have more of an effect on
after a single mTBI. 2 N-acyl methionines enzymes that preferentially act on substrates con-
increased in each of these tissue types and taining AA.
N-stearoyl methionine increased in both tissue
types. These changes were no longer present
after repeated mTBI. Only the TG and TNC had 16.5 E
ffects on Bioactive Lipids
changes in N-acyl methionines in this condition. Change Over Time But Are
There was a large increase in N-docosahexaenoyl More Similar When
methionine in the TG and a more modest increase Headache-Like Symptoms
in N-arachidonoyl methionine (A-Met) in the Are Present
TNC (Fig. 16.5).
Effects of mTBI and acrolein exposure were
quite different when comparing acute acrolein
N-Acyl Phenylalanines In all 3 tissue types, a exposure effects to mTBI effects. Specifically,
single exposure to mTBI increased levels of bioactive lipids were more likely to
N-arachidonoyl phenylalanine (A-Phe). Single decrease after the acute acrolein exposure [43].
mTBI also increased N-oleoyl phenylalanine in These lipids tended to increase after mTBI. One
the TNC only. Changes in N-acyl phenylalanines explanation for these differences is that the tissue
were not measured in the CER after 4 mTBIs and was collected immediately after the single 4 h
increases in A-Phe were restricted to the TG and acrolein exposure, whereas tissue was collected
TNC. 2 additional N-acyl phenylalanines at least 72 h later in the case of mTBI. It wasn’t
decreased in the TNC (Fig. 16.5). until after repeated acrolein exposure that
headache-like behaviors appeared [54], whereas
after the single mTBI exposure, there were mea-
PhosphoLEA PhosphoNAEs are intermediates surable headache-like behaviors. Similar altera-
for NAE production [109]. PhosphoLEA is the tions in lipid levels were observed between the
only phosphoNAE that is available in the screen- models at time points when headache behavior
ing library. This lipid only changed after repeated was measurable (4 acrolein exposures or 1 mTBI
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 211
exposure). One key finding is that chronic acro- Systemic pretreatment with the TRPA1 antago-
lein exposure increased levels of multiple TRPV nist AP-18 prevented the sensitization of the TVS
ligands and increased levels of NAGly [43], as due to chronic acrolein [11]. In acrolein-exposed
did mTBI. Thus, bioactive lipids may serve as a rats treated with AP-18, changes in dural blood
marker for the physiological changes in head- flow in response to either TRPA1 or TRPV1 ago-
ache, regardless of the original stimulus that dis- nist challenges did not sensitize the responses,
rupted the TVS. Follow-up studies should implicating TRPA1 in the process of sensitization
examine whether there are similar changes in regardless of the exact receptor action of the
lipid levels immediately after the mTBI, as these agent [11]. The biochemical similarities between
could give insight into the lipid milieu that even- the 2 models (acrolein and mTBI), such as
tually gives rise to inflammation and pain. increases in NAGly [43], as well as the similari-
Providing further evidence that the effects of ties in central and peripheral sensitization [11,
painful insults on bioactive lipids are dynamic 54], suggest that the same therapy may work in
and dependent on time post-insult, in another both models. A follow up study will test the
rodent model of inflammatory pain, we also hypothesis that pretreatment with AP-18 prevents
observed decreases in bioactive lipids in CNS tis- headache driven by mTBI.
sues 1 h after intraplantar carrageenan applica- Novel headache therapeutics that block
tion [41]. For example, A-GABA decreased in microglial activation are currently being devel-
the cerebellum and brainstem and OEA decreased oped [13, 112]. It is possible that attenuating
in the thalamus. At 3 h post-carrageenan, microglial activation could be a therapy for head-
increases were observed in multiple NAEs in all ache due to mTBI, as both share changes in neu-
6 brain areas tested (striatum, hippocampus, cer- roinflammatory mediators that drive microglial
ebellum, thalamus, midbrain, and brainstem) activation [4, 13]. Strengthening the hypothesis
[41]. These effects at 3 h included increases in that blocking microglial migration represents a
NAGly in the striatum, hippocampus, and cere- strategy for treating mTBI, A-Ser, which attenu-
bellum [41]. Therefore, the effects of peripheral ated NAGly-driven microglial migration [59],
insults on bioactive lipids take some time to cen- was also neuroprotective after TBI [47, 48].
tralize, whereas the effects of central insults like A-Ser levels were elevated in the CER and TG
mTBI may take some time to fully permeate the post-repeated mTBI, potentially representing a
periphery. Overall, the effects of peripheral compensatory strategy to reduce the microglial
inflammation, acrolein exposure, and mTBI on activation and migration that occurs after
bioactive lipids look more similar when pain- TBI. Because GPR18 activation drives microglial
related behaviors emerge. migration [59], it should be tested whether
GPR18 antagonists are effective at preventing
microglial migration after mTBI, as this might
16.5.1 Implications for Therapeutics represent a novel strategy to reduce inflamma-
tion. FAAH inhibition also represents a strategy
Current therapeutics for headache are not partic- to reduce NAGly levels [69, 113]. Lipidomics
ularly effective, underscoring the need to better studies have revealed that genetic [69] and phar-
understand the mechanisms that cause headache macological [113] inhibition of FAAH increases
and develop new drugs that target these mecha- levels of AEA and other NAEs but at the expense
nisms [110]. TRPA1 may represent a novel target of other AA-derived lipoamines such as NAGly.
for treating headache [111]. Studies in the TRPA1 Treatment with the FAAH inhibitor URB597
KO mouse [52] suggest that TRPA1 is required reduced microglial and astrocytic activation after
for pain and hypersensitivity associated with TBI in rats and was associated with improvement
chronic inflammation, as TRPA1 functions as a in behavioral measures [20]. The contribution of
gatekeeper of the release of endogenous inflam- NAGly to these findings has not yet been
matory mediators from sensory neurons [24]. investigated.
212 E. Leishman et al.
A, Etinger A, Mechoulam R (1992) Isolation and 27. Julius D (2013) TRP channels and pain. Annu Rev
structure of a brain constituent that binds to the can- Cell Dev Biol 29:355–384
nabinoid receptor. Science 258(5090):1946–1949 28. Moran MM (2018) TRP channels as potential drug
15. Sugiura T, Kondo S, Sukagawa A, Nakane S, targets. Annu Rev Pharmacol Toxicol 58:309–330
Shinoda A, Itoh K, Yamashita A, Waku K (1995) 29. Voets T, Talavera K, Owsianik G, Nilius B (2005)
2-Arachidonoylglycerol: a possible endogenous can- Sensing with TRP channels. Nat Chem Biol
nabinoid receptor ligand in brain. Biochem Biophys 1(2):85–92
Res Commun 215(1):89–97 30. Venkatachalam K, Luo J, Montell C (2014)
16. Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky Evolutionarily conserved, multitasking TRP chan-
M, Kaminski NE, Schatz AR, Gopher A, Almog S, nels: lessons from worms and flies. In: Mammalian
Martin BR, Compton DR et al (1995) Identification transient receptor potential (TRP) cation channels.
of an endogenous 2-monoglyceride, present in Springer, Berlin, pp 937–962
canine gut, that binds to cannabinoid receptors. 31. Nilius B, Szallasi A (2014) Transient receptor poten-
Biochem Pharmacol 50(1):83–90 tial channels as drug targets: from the science of
17. Felder CC, Joyce KE, Briley EM, Mansouri J, basic research to the art of medicine. Pharmacol Rev
Mackie K, Blond O, Lai Y, Ma AL, Mitchell RL 66(3):676–814
(1995) Comparison of the pharmacology and signal 32. Ho KW, Ward NJ, Calkins DJ (2012) TRPV1: a
transduction of the human cannabinoid CB1 and stress response protein in the central nervous system.
CB2 receptors. Mol Pharmacol 48(3):443–450 Am J Neurodegener Dis 1(1):1–14
18. Leimuranta P, Khiroug L, Giniatullin R (2018) 33. Starowicz K, Makuch W, Korostynski M, Malek N,
Emerging role of (Endo) cannabinoids in migraine. Slezak M, Zychowska M, Petrosino S, De Petrocellis
Front Pharmacol 9:420 L, Cristino L, Przewlocka B, Di Marzo V (2013) Full
19. Panikashvili D, Simeonidou C, Ben-Shabat S, Hanuš inhibition of spinal FAAH leads to TRPV1-mediated
L, Breuer A, Mechoulam R, Shohami E (2001) An analgesic effects in neuropathic rats and possible
endogenous cannabinoid (2-AG) is neuroprotective lipoxygenase-mediated remodeling of anandamide
after brain injury. Nature 413(6855):527–531 metabolism. PLoS One 8(4):e60040. https://doi.
20. Katz PS, Sulzer JK, Impastato RA, Teng SX, org/10.1371/journal.pone.0060040
Rogers EK, Molina PE (2015) Endocannabinoid 34. Caterina MJ, Schumacher MA, Tominaga M, Rosen
degradation inhibition improves neurobehavioral TA, Levine JD, Julius D (1997) The capsaicin recep-
function, blood–brain barrier integrity, and neuro- tor: a heat-activated ion channel in the pain pathway.
inflammation following mild traumatic brain injury. Nature 389(6653):816–824
J Neurotrauma 32(5):297–306 35. Zygmunt PM, Petersson J, Andersson DA, Chuang
21. Amenta PS, Jallo JI, Tuma RF, Elliott MB (2012) H, Sorgard M, Di Marzo V, Julius D, Hogestatt ED
A cannabinoid type 2 receptor agonist attenuates (1999) Vanilloid receptors on sensory nerves medi-
blood–brain barrier damage and neurodegenera- ate the vasodilator action of anandamide. Nature
tion in a murine model of traumatic brain injury. 400(6743):452–457. https://doi.org/10.1038/22761
J Neurosci Res 90(12):2293–2305 36. Zygmunt PM, Ermund A, Movahed P, Andersson
22. Bradshaw HB, Raboune S, Hollis JL (2013) DA, Simonsen C, Jonsson BA, Blomgren A, Birnir B,
Opportunistic activation of TRP receptors by Bevan S, Eschalier A, Mallet C, Gomis A, Hogestatt
endogenous lipids: exploiting lipidomics to under- ED (2013) Monoacylglycerols activate TRPV1--a
stand TRP receptor cellular communication. Life link between phospholipase C and TRPV1. PLoS
Sci 92(8–9):404–409. https://doi.org/10.1016/j. One 8(12):e81618. https://doi.org/10.1371/journal.
lfs.2012.11.008 pone.0081618
23. Marrone MC, Morabito A, Giustizieri M, Chiurchiu 37. Ahern GP (2003) Activation of TRPV1 by the
V, Leuti A, Mattioli M, Marinelli S, Riganti L, satiety factor oleoylethanolamide. J Biol Chem
Lombardi M, Murana E, Totaro A, Piomelli D, 278(33):30429–30434. https://doi.org/10.1074/jbc.
Ragozzino D, Oddi S, Maccarrone M, Verderio C, M305051200
Marinelli S (2017) TRPV1 channels are critical 38. Movahed P, Jonsson BA, Birnir B, Wingstrand JA,
brain inflammation detectors and neuropathic pain Jorgensen TD, Ermund A, Sterner O, Zygmunt
biomarkers in mice. Nat Commun 8:15292. https:// PM, Hogestatt ED (2005) Endogenous unsaturated
doi.org/10.1038/ncomms15292 C18 N-acylethanolamines are vanilloid receptor
24. Bautista DM, Pellegrino M, Tsunozaki M (2013) (TRPV1) agonists. J Biol Chem 280(46):38496–
TRPA1: a gatekeeper for inflammation. Annu 38504. https://doi.org/10.1074/jbc.M507429200
Rev Physiol 75:181–200. https://doi.org/10.1146/ 39. Saghatelian A, McKinney MK, Bandell M,
annurev-physiol-030212-183811 Patapoutian A, Cravatt BF (2006) A FAAH-regulated
25. Oxford GS, Hurley JH (2013) The role of TRP chan- class of N-acyl taurines that activates TRP ion chan-
nels in migraine. Open Pain J 6(Suppl 1):37–49 nels. Biochemistry 45(30):9007–9015. https://doi.
26. Clapham DE (2003) TRP channels as cellular sen- org/10.1021/bi0608008
sors. Nature 426(6966):517–524 40. Huang SM, Bisogno T, Trevisani M, Al-Hayani A,
De Petrocellis L, Fezza F, Tognetto M, Petros TJ,
214 E. Leishman et al.
Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti Nature 427(6971):260–265. https://doi.org/10.1038/
P, Walker JM, Di Marzo V (2002) An endogenous nature02282
capsaicin-like substance with high potency at recom- 50. Feng Z, Hu W, Hu Y, Tang MS (2006) Acrolein is
binant and native vanilloid VR1 receptors. Proc Natl a major cigarette-related lung cancer agent: pref-
Acad Sci U S A 99(12):8400–8405. https://doi. erential binding at p53 mutational hotspots and
org/10.1073/pnas.122196999 inhibition of DNA repair. Proc Natl Acad Sci U S
41. Raboune S, Stuart JM, Leishman E, Takacs SM, A 103(42):15404–15409. https://doi.org/10.1073/
Rhodes B, Basnet A, Jameyfield E, McHugh pnas.0607031103
D, Widlanski T, Bradshaw HB (2014) Novel 51. Brone B, Peeters PJ, Marrannes R, Mercken M,
endogenous N-acyl amides activate TRPV1-4 recep- Nuydens R, Meert T, Gijsen HJ (2008) Tear gas-
tors, BV-2 microglia, and are regulated in brain in ses CN, CR, and CS are potent activators of the
an acute model of inflammation. Front Cell Neurosci human TRPA1 receptor. Toxicol Appl Pharmacol
8:195. https://doi.org/10.3389/fncel.2014.00195 231(2):150–156. https://doi.org/10.1016/j.
42. Kunkler PE, Ballard CJ, Pellman JJ, Zhang L, taap.2008.04.005
Oxford GS, Hurley JH (2014) Intraganglionic signal- 52. Bautista DM, Jordt SE, Nikai T, Tsuruda PR, Read
ing as a novel nasal-meningeal pathway for TRPA1- AJ, Poblete J, Yamoah EN, Basbaum AI, Julius D
dependent trigeminovascular activation by inhaled (2006) TRPA1 mediates the inflammatory actions
environmental irritants. PLoS One 9(7):e103086. of environmental irritants and proalgesic agents.
https://doi.org/10.1371/journal.pone.0103086 Cell 124(6):1269–1282. https://doi.org/10.1016/j.
43. Leishman E, Kunkler PE, Manchanda M, Sangani cell.2006.02.023
K, Stuart JM, Oxford GS, Hurley JH, Bradshaw 53. Kunkler PE, Ballard CJ, Oxford GS, Hurley JH
HB (2017) Environmental toxin acrolein alters lev- (2011) TRPA1 receptors mediate environmen-
els of endogenous lipids, including TRP agonists: a tal irritant-induced meningeal vasodilatation.
potential mechanism for headache driven by TRPA1 Pain 152(1):38–44. https://doi.org/10.1016/j.
activation. Neurobiol Pain 1:28–36. https://doi. pain.2010.08.021
org/10.1016/j.ynpai.2017.03.001 54. Kunkler PE, Zhang L, Johnson PL, Oxford GS,
44. Milman G, Maor Y, Abu-Lafi S, Horowitz M, Hurley JH (2018) Induction of chronic migraine
Gallily R, Batkai S, Mo F-M, Offertaler L, Pacher phenotypes in a rat model after environmental irri-
P, Kunos G (2006) N-arachidonoyl L-serine, an tant exposure. Pain 159(3):540–549. https://doi.
endocannabinoid-like brain constituent with vaso- org/10.1097/j.pain.0000000000001124
dilatory properties. Proc Natl Acad Sci U S A 55. Eftekhari S, Edvinsson L (2011) Calcitonin gene-
103(7):2428–2433 related peptide (CGRP) and its receptor components
45. Smoum R, Bar A, Tan B, Milman G, Attar-Namdar in human and rat spinal trigeminal nucleus and spi-
M, Ofek O, Stuart JM, Bajayo A, Tam J, Kram V nal cord at C1-level. BMC Neurosci 12(1):1
(2010) Oleoyl serine, an endogenous N-acyl amide, 56. Daiutolo BV, Tyburski A, Clark SW, Elliott MB
modulates bone remodeling and mass. Proc Natl (2016) Trigeminal pain molecules, allodynia, and
Acad Sci 107(41):17710–17715 photosensitivity are pharmacologically and geneti-
46. Tortoriello G, Rhodes BP, Takacs SM, Stuart JM, cally modulated in a model of traumatic brain injury.
Basnet A, Raboune S, Widlanski TS, Doherty P, J Neurotrauma 33(8):748–760
Harkany T, Bradshaw HB (2013) Targeted lipido- 57. Elliott MB, Oshinsky ML, Amenta PS, Awe OO, Jallo
mics in Drosophila melanogaster identifies novel JI (2012) Nociceptive neuropeptide increases and
2-monoacylglycerols and N-acyl amides. PLoS periorbital allodynia in a model of traumatic brain
One 8(7):e67865. https://doi.org/10.1371/journal. injury. Headache J Head Face Pain 52(6):966–984
pone.0067865 58. Bradshaw HB, Rimmerman N, Hu SS, Benton
47. Mann A, Cohen-Yeshurun A, Trembovler VM, Stuart JM, Masuda K, Cravatt BF, O’Dell DK,
V, Mechoulam R, Shohami E (2016) Are the Walker JM (2009) The endocannabinoid anandamide
endocannabinoid- like compounds N-acyl amino- is a precursor for the signaling lipid N-arachidonoyl
acids neuroprotective after traumatic brain injury? glycine by two distinct pathways. BMC Biochem
J Basic Clin Physiol Pharmacol 27(3):209–216 10:14. https://doi.org/10.1186/1471-2091-10-14
48. Cohen-Yeshurun A, Trembovler V, Alexandrovich 59. McHugh D, Hu SS, Rimmerman N, Juknat
A, Ryberg E, Greasley PJ, Mechoulam R, Shohami A, Vogel Z, Walker JM, Bradshaw HB (2010)
E, Leker RR (2011) N-arachidonoyl-L-serine is neu- N-arachidonoyl glycine, an abundant endogenous
roprotective after traumatic brain injury by reducing lipid, potently drives directed cellular migration
apoptosis. J Cereb Blood Flow Metab 31(8):1768– through GPR18, the putative abnormal cannabi-
1777. https://doi.org/10.1038/jcbfm.2011.53 diol receptor. BMC Neurosci 11:44. https://doi.
49. Jordt SE, Bautista DM, Chuang HH, McKemy DD, org/10.1186/1471-2202-11-44
Zygmunt PM, Hogestatt ED, Meng ID, Julius D 60. Zimmermann M (1983) Ethical guidelines for inves-
(2004) Mustard oils and cannabinoids excite sensory tigations of experimental pain in conscious animals.
nerve fibres through the TRP channel ANKTM1. Pain 16(2):109–110
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 215
61. Velosky AG, Tucker LB, Fu AH, Liu J, McCabe JT impacts of the synthetic cannabinoid CP 55,940 in
(2017) Cognitive performance of male and female adolescent and adult female mouse brains. Front
C57BL/6J mice after repetitive concussive brain Mol Neurosci 11:436
injuries. Behav Brain Res 324:115–124 73. McHugh D, Wager-Miller J, Page J, Bradshaw HB
62. Fehily B, Fitzgerald M (2017) Repeated mild trau- (2012) siRNA knockdown of GPR18 receptors in
matic brain injury: potential mechanisms of damage. BV-2 microglia attenuates N-arachidonoyl glycine-
Cell Transplant 26(7):1131–1155 induced cell migration. J Mol Signal 7(1):10. https://
63. Shitaka Y, Tran HT, Bennett RE, Sanchez L, Levy doi.org/10.1186/1750-2187-7-10
MA, Dikranian K, Brody DL (2011) Repetitive 74. Shohami E, Cohen-Yeshurun A, Magid L, Algali
closed-skull traumatic brain injury in mice causes M, Mechoulam R (2011) Endocannabinoids
persistent multifocal axonal injury and microglial and traumatic brain injury. Br J Pharmacol
reactivity. J Neuropathol Exp Neurol 70(7):551–567 163(7):1402–1410. https://doi.org/10.1111/j.1476-
64. Bolton AN, Saatman KE (2014) Regional neurode- 5381.2011.01343.x
generation and gliosis are amplified by mild trau- 75. Jeffries KA, Dempsey DR, Farrell EK, Anderson
matic brain injury repeated at 24-hour intervals. RL, Garbade GJ, Gurina TS, Gruhonjic I, Gunderson
J Neuropathol Exp Neurol 73(10):933–947 CA, Merkler DJ (2016) Glycine N-acyltransferase
65. Tan B, Bradshaw HB, Rimmerman N, Srinivasan like 3 is responsible for long-chain N-acylglycine
H, Yu YW, Krey JF, Monn MF, Chen JS, Hu SS, formation in N18TG2 cells. J Lipid Res 57(5):781–
Pickens SR, Walker JM (2006) Targeted lipido- 790. https://doi.org/10.1194/jlr.M062042
mics: discovery of new fatty acyl amides. AAPS 76. Long JZ, Roche AM, Berdan CA, Louie SM, Roberts
J 8(3):E461–E465. https://doi.org/10.1208/ AJ, Svensson KJ, Dou FY, Bateman LA, Mina AI,
aapsj080354 Deng Z, Jedrychowski MP, Lin H, Kamenecka
66. Carey LM, Slivicki RA, Leishman E, Cornett TM, Asara JM, Griffin PR, Banks AS, Nomura
B, Mackie K, Bradshaw H, Hohmann AG DK, Spiegelman BM (2018) Ablation of PM20D1
(2016) A pro-nociceptive phenotype unmasked reveals N-acyl amino acid control of metabolism and
in mice lacking fatty-acid amide hydrolase. nociception. Proc Natl Acad Sci U S A 115:E6937–
Mol Pain 12:174480691664919. https://doi. E6945. https://doi.org/10.1073/pnas.1803389115
org/10.1177/1744806916649192 77. Long JZ, Svensson KJ, Bateman LA, Lin H,
67. Stuart JM, Paris JJ, Frye C, Bradshaw HB (2013) Kamenecka T, Lokurkar IA, Lou J, Rao RR, Chang
Brain levels of prostaglandins, endocannabinoids, MR, Jedrychowski MP, Paulo JA, Gygi SP, Griffin
and related lipids are affected by mating strate- PR, Nomura DK, Spiegelman BM (2016) The
gies. Int J Endocrinol 2013:436252. https://doi. secreted enzyme PM20D1 regulates lipidated amino
org/10.1155/2013/436252 acid uncouplers of mitochondria. Cell 166(2):424–
68. Sun X, Deng W, Li Y, Tang S, Leishman E, Bradshaw 435. https://doi.org/10.1016/j.cell.2016.05.071
HB, Dey SK (2016) Sustained endocannabinoid sig- 78. Donvito G, Piscitelli F, Muldoon P, Jackson A,
naling compromises decidual function and promotes Vitale RM, D’Aniello E, Giordano C, Ignatowska-
inflammation-induced preterm birth. J Biol Chem Jankowska BM, Mustafa MA, Guida F (2018)
291(15):8231–8240. https://doi.org/10.1074/jbc. N-Oleoyl-glycine reduces nicotine reward and with-
M115.707836 drawal in mice. Neuropharmacology 148:320–331
69. Leishman E, Cornett B, Spork K, Straiker A, Mackie 79. Blennow K, Hardy J, Zetterberg H (2012) The neu-
K, Bradshaw HB (2016) Broad impact of deleting ropathology and neurobiology of traumatic brain
endogenous cannabinoid hydrolyzing enzymes and injury. Neuron 76(5):886–899
the CB1 cannabinoid receptor on the endogenous 80. Koivisto A (2012) Sustained TRPA1 activation
cannabinoid-related lipidome in eight regions of the in vivo. Acta Physiol 204(2):248–254
mouse brain. Pharmacol Res 110:159–172. https:// 81. Bailes JE, Mills JD (2010) Docosahexaenoic acid
doi.org/10.1016/j.phrs.2016.04.020 reduces traumatic axonal injury in a rodent head
70. Leishman E, Mackie K, Luquet S, Bradshaw HB injury model. J Neurotrauma 27(9):1617–1624.
(2016) Lipidomics profile of a NAPE-PLD KO https://doi.org/10.1089/neu.2009.1239
mouse provides evidence of a broader role of this 82. Wu A, Ying Z, Gomez-Pinilla F (2004) Dietary
enzyme in lipid metabolism in the brain. Biochim omega-3 fatty acids normalize BDNF levels, reduce
Biophys Acta 1861(6):491–500. https://doi. oxidative damage, and counteract learning disability
org/10.1016/j.bbalip.2016.03.003 after traumatic brain injury in rats. J Neurotrauma
71. Leishman E, Murphy M, Mackie K, Bradshaw HB 21(10):1457–1467
(2018) Delta(9)-tetrahydrocannabinol changes the 83. Mills JD, Hadley K, Bailes JE (2011) Dietary supple-
brain lipidome and transcriptome differentially mentation with the omega-3 fatty acid docosahexae-
in the adolescent and the adult. Biochim Biophys noic acid in traumatic brain injury. Neurosurgery
Acta 1863(5):479–492. https://doi.org/10.1016/j. 68(2):474–481
bbalip.2018.02.001 84. Desai A, Kevala K, Kim H-Y (2014) Depletion of
72. Leishman E, Murphy MN, Murphy MI, Mackie K, brain docosahexaenoic acid impairs recovery from
Bradshaw HB (2018) Broad and region-specific traumatic brain injury. PLoS One 9(1):e86472
216 E. Leishman et al.
85. Harvey LD, Yin Y, Attarwala IY, Begum G, Deng J, 96. Reisenberg M, Singh PK, Williams G, Doherty P
Yan HQ, Dixon CE, Sun D (2015) Administration (2012) The diacylglycerol lipases: structure, regu-
of DHA reduces endoplasmic reticulum stress- lation and roles in and beyond endocannabinoid
associated inflammation and alters microglial or signalling. Philos Trans R Soc Lond Ser B Biol
macrophage activation in traumatic brain injury. Sci 367(1607):3264–3275. https://doi.org/10.1098/
ASN Neuro 7(6):1759091415618969 rstb.2011.0387
86. Serhan CN, Recchiuti A (2012) Pro-resolving lipid 97. Tanimura A, Yamazaki M, Hashimotodani Y,
mediators (SPMs) and their actions in regulating Uchigashima M, Kawata S, Abe M, Kita Y,
miRNA in novel resolution circuits in inflammation. Hashimoto K, Shimizu T, Watanabe M, Sakimura
Front Immunol 3:298 K, Kano M (2010) The endocannabinoid
87. Xu Z-Z, Zhang L, Liu T, Park JY, Berta T, Yang 2-arachidonoylglycerol produced by diacylglycerol
R, Serhan CN, Ji R-R (2010) Resolvins RvE1 and lipase alpha mediates retrograde suppression of syn-
RvD1 attenuate inflammatory pain via central and aptic transmission. Neuron 65(3):320–327. https://
peripheral actions. Nat Med 16(5):592–597 doi.org/10.1016/j.neuron.2010.01.021
88. Park C-K, Lü N, Xu Z-Z, Liu T, Serhan CN, Ji R-R 98. Gao Y, Vasilyev DV, Goncalves MB, Howell FV,
(2011) Resolving TRPV1-and TNF-α-mediated Hobbs C, Reisenberg M, Shen R, Zhang MY, Strassle
spinal cord synaptic plasticity and inflamma- BW, Lu P, Mark L, Piesla MJ, Deng K, Kouranova
tory pain with neuroprotectin D1. J Neurosci EV, Ring RH, Whiteside GT, Bates B, Walsh FS,
31(42):15072–15085 Williams G, Pangalos MN, Samad TA, Doherty P
89. Harrison JL, Rowe RK, Lifshitz J (2016) Lipid (2010) Loss of retrograde endocannabinoid signal-
mediators of inflammation in neurological injury: ing and reduced adult neurogenesis in diacylglycerol
shifting the balance toward resolution. Neural Regen lipase knock-out mice. J Neurosci 30(6):2017–2024.
Res 11(1):77–78 https://doi.org/10.1523/JNEUROSCI.5693-09.2010
90. Harrison JL, Rowe RK, Ellis TW, Yee NS, O’Hara 99. Shonesy BC, Bluett RJ, Ramikie TS, Báldi R,
BF, Adelson PD, Lifshitz J (2015) Resolvins AT-D1 Hermanson DJ, Kingsley PJ, Marnett LJ, Winder
and E1 differentially impact functional outcome, DG, Colbran RJ, Patel S (2014) Genetic disruption
post-traumatic sleep, and microglial activation fol- of 2-arachidonoylglycerol synthesis reveals a key
lowing diffuse brain injury in the mouse. Brain role for endocannabinoid signaling in anxiety modu-
Behav Immun 47:131–140 lation. Cell Rep 9(5):1644–1653
91. Marcheselli VL, Hong S, Lukiw WJ, Tian XH, 100. Ogasawara D, Deng H, Viader A, Baggelaar MP,
Gronert K, Musto A, Hardy M, Gimenez JM, Chiang Breman A, den Dulk H, van den Nieuwendijk AM,
N, Serhan CN (2003) Novel docosanoids inhibit Soethoudt M, van der Wel T, Zhou J, Overkleeft HS,
brain ischemia-reperfusion-mediated leukocyte Sanchez-Alavez M, Mori S, Nguyen W, Conti B,
infiltration and pro-inflammatory gene expression. Liu X, Chen Y, Liu QS, Cravatt BF, van der Stelt M
J Biol Chem 278(44):43807–43817 (2016) Rapid and profound rewiring of brain lipid
92. Yang R, Fredman G, Krishnamoorthy S, Agrawal N, signaling networks by acute diacylglycerol lipase
Irimia D, Piomelli D, Serhan CN (2011) Decoding inhibition. Proc Natl Acad Sci U S A 113(1):26–33.
functional metabolomics with docosahexaenoyl https://doi.org/10.1073/pnas.1522364112
ethanolamide (DHEA) identifies novel bioactive sig- 101. van der Wel T, Janssen FJ, Baggelaar MP, Deng
nals. J Biol Chem 286(36):31532–31541. https://doi. H, den Dulk H, Overkleeft HS, van der Stelt M
org/10.1074/jbc.M111.237990 (2015) A natural substrate-based fluorescence assay
93. Park SW, Hah JH, Oh SM, Jeong WJ, Sung MW for inhibitor screening on diacylglycerol lipase α.
(2016) 5-lipoxygenase mediates docosahexae- J Lipid Res 56(4):927–935
noyl ethanolamide and N-arachidonoyl-L-alanine- 102. Baggelaar MP, Janssen FJ, van Esbroeck A, den
induced reactive oxygen species production and Dulk H, Allarà M, Hoogendoorn S, McGuire R,
inhibition of proliferation of head and neck squa- Florea BI, Meeuwenoord N, van den Elst H (2013)
mous cell carcinoma cells. BMC Cancer 16:458. Development of an activity-based probe and in silico
https://doi.org/10.1186/s12885-016-2499-3 design reveal highly selective inhibitors for dia-
94. Bradshaw HB, Leishman E (2017) Lipidomics: cylglycerol lipase-α in brain. Angew Chem Int Ed
a corrective lens for enzyme myopia. In: Methods 52(46):12081–12085
in enzymology, vol 593. Elsevier, Amsterdam, 103. Liu Y, Patricelli MP, Cravatt BF (1999) Activity-
pp 123–141 based protein profiling: the serine hydrolases. Proc
95. Bisogno T, Howell F, Williams G, Minassi A, Cascio Natl Acad Sci 96(26):14694–14699
MG, Ligresti A, Matias I, Schiano-Moriello A, Paul 104. Sugiura T, Kishimoto S, Oka S, Gokoh M (2006)
P, Williams E-J (2003) Cloning of the first sn1-DAG Biochemistry, pharmacology and physiology of
lipases points to the spatial and temporal regulation 2-arachidonoylglycerol, an endogenous cannabi-
of endocannabinoid signaling in the brain. J Cell noid receptor ligand. Prog Lipid Res 45(5):405–446.
Biol 163(3):463–468 https://doi.org/10.1016/j.plipres.2006.03.003
16 Bioactive Lipids in Cancer, Inflammation and Related Diseases 217
105. Wei EP, Lamb RG, Kontos HA (1982) Increased 111. Koivisto A, Chapman H, Jalava N, Korjamo T,
phospholipase C activity after experimental brain Saarnilehto M, Lindstedt K, Pertovaara A (2014)
injury. J Neurosurg 56(5):695–698 TRPA1: a transducer and amplifier of pain and
106. Hwang SC, Jhon D-Y, Bae YS, Kim JH, Rhee SG inflammation. Basic Clin Pharmacol Toxicol
(1996) Activation of phospholipase C-γ by the con- 114(1):50–55. https://doi.org/10.1111/bcpt.12138
certed action of tau proteins and arachidonic acid. 112. Liu Q, Liu C, Jiang L, Li M, Long T, He W, Qin
J Biol Chem 271(31):18342–18349 G, Chen L, Zhou J (2018) α7 nicotinic acetylcho-
107. Axelrod J, Burch RM, Jelsema CL (1988) Receptor- line receptor-mediated anti-inflammatory effect in
mediated activation of phospholipase A2 via a chronic migraine rat model via the attenuation of
GTP-binding proteins: arachidonic acid and its glial cell activation. J Pain Res 11:1129–1140
metabolites as second messengers. Trends Neurosci 113. Han B, Wright R, Kirchhoff AM, Chester JA, Cooper
11(3):117–123 BR, Davisson VJ, Barker E (2013) Quantitative
108. Pilitsis JG, Coplin WM, O’Regan MH, Wellwood LC-MS/MS analysis of arachidonoyl amino acids in
JM, Diaz FG, Fairfax MR, Michael DB, Phillis JW mouse brain with treatment of FAAH inhibitor. Anal
(2003) Free fatty acids in cerebrospinal fluids from Biochem 432(2):74–81. https://doi.org/10.1016/j.
patients with traumatic brain injury. Neurosci Lett ab.2012.09.031
349(2):136–138 114. Russo EB (2016) Clinical endocannabinoid defi-
109. Liu J, Wang L, Harvey-White J, Osei-Hyiaman ciency reconsidered: current research supports the
D, Razdan R, Gong Q, Chan AC, Zhou Z, Huang theory in migraine, fibromyalgia, irritable bowel,
BX, Kim HY, Kunos G (2006) A biosynthetic and other treatment-resistant syndromes. Cannabis
pathway for anandamide. Proc Natl Acad Sci U S Cannabinoid Res 1(1):154–165
A 103(36):13345–13350. https://doi.org/10.1073/ 115. Russo EB, Hohmann AG (2013) Role of cannabi-
pnas.0601832103 noids in pain management. In: Comprehensive treat-
110. Dussor G, Yan J, Xie JY, Ossipov MH, Dodick ment of chronic pain by medical, interventional,
DW, Porreca F (2014) Targeting TRP chan- and integrative approaches. Springer, New York,
nels for novel migraine therapeutics. ACS Chem pp 181–197
Neurosci 5(11):1085–1096. https://doi.org/10.1021/ 116. Reddy D (2017) The utility of cannabidiol in the
cn500083e treatment of refractory epilepsy. Clin Pharmacol
Ther 101(2):182–184
Novel Anti-inflammatory
and Vasodilatory ω-3 17
Endocannabinoid Epoxide
Regioisomers
Lauren N. Carnevale and Aditi Das
Cellular eCB levels are carefully regulated Following their uptake, PUFAs are mobilized
through multiple biosynthetic and degradative via transport proteins to tissues where they esteri-
enzymes. For instance, the eCBs originate from fied and are temporarily stored in the cell mem-
dietary ω-3 and ω-6 polyunsaturated fatty acids brane (Fig. 17.1). For example, AEA is stored as
(PUFAs). PUFAs are class of lipids that contain its membrane precursor N-arachidonoyl phos-
two or more cis carbon-carbon double bonds phatidylethanolamine (NAPE) [18] and is
(points of unsaturation) and are identified as ω-3 released from the membrane by phospholipase
or ω-6 based on the location of the first carbon- D. This catabolic process yields a free eCB [19].
carbon double bond from the terminal methyl After biosynthesis, the eCBs are tightly regulated
group. ω-6 eCBs derived from arachidonic acid by degradative enzymes through hydrolytic inac-
(AA), such as AEA and 2-AG, are the most well tivation [20]. For example, fatty acid amide
studied eCBs. With the recent interest in the hydrolase (FAAH) catalyzes the hydrolytic cleav-
dietary effects of ω-3 PUFAs, there has been an age of AEA to form AA and ethanolamine [21].
increased interest in ω-3 eCBs. The ω-3 eCBs are FAAH is the principal AEA and DHEA-
derived from the ω-3 PUFA docosahexaenoic hydrolyzing enzyme distributed throughout the
acid (DHA), eicosapentanoic acid (EPA, timn- nervous system [22] On the other hand, monoac-
odonic acid), or α-linoleic acid (ALA). ylglycerol lipase (MAGL) is the degradative
Fig. 17.1 Overview of the ω-3 Endocannabinoid to the eCBs DHEA and EPEA, respectively. The metabo-
Epoxide Biosynthetic Pathway. Dietary consumption of lism of DHEA and EPEA by cytochrome P450 epoxygen-
ω-3 polyunsaturated fatty acids results in the storage of ases produces six (DHEA) or five (EPEA) constitutional
DHA and EPA esters within the plasma membrane. Upon isomers at each carbon-carbon double bond. DHEA epox-
cleavage by a phospholipase, DHA and EPA are cleaved ide metabolites are termed EDP-EAs while the EPEA
from their membrane precursor and undergo biosynthesis epoxide metabolites are termed EEQ-EAs
222 L. N. Carnevale and A. Das
enzymes for 2-AG and 2-DHG [20, 23]. Overall, metabolites with novel anti-inflammatory and
these enzymes convert the eCBs back into their potential anti-pain properties as well as vasoac-
free fatty acid form, which can be esterified and tive activities.
stored in the phospholipid membrane bilayer or
used for cellular functions [24].
Based on the widespread tissue distribution of 17.1.2 Endocannabinoid Metabolism
the cannabinoid receptors there is a plethora of by Cytochrome P450 (CYP)
biochemical processes that utilize cannabinoid Epoxygenases
signaling [25–27]. The main role of CB1 is to
mediate neurobehavioral activities, such as appe- In humans, there are 57 CYPs which are
tite regulation and executive functions, while the involved in xenobiotic metabolism, steroid syn-
prominent function of CB2 is to mediate an anti- thesis, and fatty acid metabolism [43]. These
inflammatory immune response. More specifi- enzymes are distributed throughout the human
cally, CB2 is involved in neuroinflammation body, with the highest levels reported in the
through its immunoregulatory activity in murine liver. In 2008, the Hollenberg group was the
brain microglia [5, 28]. Overall, these activities first to demonstrate the metabolism of AEA by
suggest that CB1 and CB2 are neuroprotective CYPs [44]. This represented a novel metabolic
[29–32]. Given the prevalence of pathway for AEA that led to the formation of
neuroinflammatory- based diseases such as the emerging class of ω-6 derived eCB epox-
Alzheimer’s disease (AD), multiple sclerosis ides. Importantly, it was demonstrated that the
(MS), and Parkinson’s disease (PD), selective AEA epoxide, 5,6-EET-EA, had 300-fold selec-
agonists for CB2 activation or modulation of tivity for the CB2 compared to CB1 and 1000-
eCB levels have promising therapeutic potential fold greater affinity for CB2 compared to AEA,
for the control of neuroinflammation [29, 33–37]. indicating unique biological activity of the
Additionally, accumulating evidence suggests epoxides compared to their parent molecule
that eCBs target alternative receptors such as the [44]. To identify CYPs involved in the conver-
nuclear peroxisome proliferator-activated recep- sion of both AEA and 2-AG, the Hollenberg lab
tors (PPAR) and the heat sensing receptor tran- utilized LC-MS/MS to determine that CYP iso-
sient vanilloid type-1 (TRPV1), [38–40] forms, CYP3A4, CYP4F2, and CYP2D6,
prompting studies regarding their role in noci- metabolize AEA in human liver and kidney
ceptive pain. microsomes as well as in brain microsomal and
While the eCBs directly bind to CB1 and mitochondrial preparations [45]. Furthermore,
CB2 receptors, they also serve as substrates for the Das group discovered the role of cardiac
the eicosanoid synthesizing enzymes. CYP, CYP2J2, in the metabolism of 2-AG and
Cyclooxygenases, lipoxygenases, and epoxy- AEA. This finding inspired future studies on
genases oxidize fatty acids to bioactive metabo- CYP2J2-mediated ω-3 and ω-6 eCB metabo-
lites that have novel physiological and lism [12, 13, 46]. CYP2J2 converts PUFAs to
cannabinoid receptor activities that are unique bioactive fatty acid epoxides which display
compared to their parent molecule [5, 41]. anti-inflammatory activities and enhance
Previously, the Serhan group reported the metab- wound repair [47–49]. CYP epoxygenases
olism of DHEA by the lipoxygenase pathway. mediate the metabolism of eCBs to eCB epox-
DHEA is metabolized to 10,12-diHDHA-EA ides [13, 50, 51]. More recent studies have
and 15-HEDPEA, which reduce leukocyte infil- shown that CYP2J2 and other CYPs metabolize
tration (chemotaxis) during inflammation. They ω-3 eCBs such as DHEA and EPEA to more
can activate the CB2 receptor with EC50 values at potent anti-inflammatory epoxide metabolites,
3.9 nM and 1 nM, respectively [42]. In this chap- as illustrated in Figs. 17.1 and 17.2. This meta-
ter, we will discuss the monooxygenation of bolic pathway consists of CYP epoxygenases,
eCBs by epoxygenases to produce eCB epoxide which belong to the cysteinato-hemoprotein
17 Novel Anti-inflammatory and Vasodilatory ω-3 Endocannabinoid Epoxide Regioisomers 223
Fig. 17.2 Endocannabinoid Signaling Regulates tively). These metabolites are more potent at CB2 than
Inflammation and Arterial Tone. Inflammatory stimuli their eCB parent molecule and display CB2 selectivity.
such as LPS up-regulates cytochrome P450 expression 19,20-EDP-EA and 17,18-EEQ-EA diminish pro-
which metabolize eCBs DHEA and EPEA to epoxide inflammatory NO and IL-6 while they elevate anti-
metabolites (19,20-EDP-EA and 17,18-EEQ-EA, respec- inflammatory marker IL-10. In addition, DHEA, EPEA as
well as their epoxide metabolites are vasoactive
superfamily and diflavin enzyme, CYP reduc- MS/MS) detected DHEA in rat brain
tase (CPR), which shuttles electrons from (128.9 ± 27.9 pmol/g), heart (57.7 ± 24.4 pmol/g),
NADPH to the CYP [52]. kidney (49.6 ± 22.2 pmol/g), spleen
(22.9 ± 1.3 pmol/g), liver (195.1 ± 48.7 pmol/g),
pig brain (265.5 ± 100 pmol/g), and human
17.2 Endocannabinoid Epoxides plasma (13.3 ± 0.4 pmol/mL). Interestingly,
dietary supplementation with DHA increased
17.2.1 ω-3 Endocannabinoid DHA the levels of DHEA at the expense of AEA,
Ethanolamide (DHEA) thereby providing a method for elevating the
levels of ω-3 eCBs in vivo [13, 15]. Although an
Encouraged by studies that identified AEA-like exact biosynthetic pathway for DHEA has yet to
fatty acid ethanolamides across several tissues, be determined, it is believed that DHEA is pro-
the Di Marzo group sought to identify their ω-3 duced in a similar manner to AEA. It has been
eCB counterparts. This group demonstrated that suggested that a membrane precursor,
ω-3 eCB, DHEA is located in bovine retina. N-docosahexaenoyl phosphatidylethanolamine
Furthermore, DHEA was found at twofold (NDHPE) is hydrolyzed by phospholipase D2 to
higher levels compared to AEA in the brain produce free DHEA, however, more studies are
(3.77 ± 0.66 ng/g and 1.88 ± 0.46 ng/g, respec- required to confirm this hypothesis [55]. Since
tively) [15, 53]. Additionally, DHEA has been DHEA is structurally similar to AEA and
detected under peripheral inflammation in rat showed a high potential for physiological rele-
contralateral spinal tissue [54]. More recently, a vance, there was significant interest in investi-
targeted lipidomics method using liquid chro- gating its physiological activities in vitro and in
matography tandem mass spectrometry (LC- vivo. Additionally, DHEA displays immune-
224 L. N. Carnevale and A. Das
modulatory activity in RAW264.7 cells. DHEA showed that epoxygenation of DHEA by CYP
dose-dependently reduced monocyte chemoat- epoxygenases led to an altered CB1 and CB2
tractant protein 1 (MCP-1) mRNA, prostaglan- activity profile compared to the DHEA parent
din and thromboxane B2 levels produced by molecule. For example, 19,20-EDP-EA was
COX-2 [56]. observed to be more potent at CB2 compared to
DHEA and had a ten-fold lower activity at CB1
(EC50 at CB1: 108 nM and CB2: 280 nM)
17.2.2 ω-3 Endocannabinoid DHEA (Fig. 17.2) [13]. As 19,20-EDP-EA, was observed
Epoxides to display an altered cannabinoid receptor profile
(Epoxydocosapentaenoic compared to DHEA, it would be interesting to
Acid-Ethanolamide, EDP-EA) characterize the other five DHEA epoxide regioi-
somers produced by the cross-talk of CYP
Recently, the Das group showed that DHEA is a metabolism pathways. From similar studies on
substrate for enzymes in the CYP epoxygenase other regioisomers, it is expected that DHEA
pathway. They designed a targeted lipidomics epoxide regioisomers and enantiomers will dis-
LC-MS/MS method in the multiple reaction play different activities at the CB1 and CB2
monitoring mode (MRM) to simultaneously receptors.
detect and quantify five DHEA epoxide
(EDP-EA) regioisomers in rat brain, heart, kid-
ney, spleen, liver tissues as well as in mouse 17.2.3 Anti-Inflammatory Activity
microglial cells. These isomers share the same of DHA Ethanolamide
molecular weight but differ in the order in which Epoxides (EDP-EAs)
their atoms are connected. Due to the reported
upregulation of CYP epoxygenases in LPS- Multiple studies indicate that eCBs combat
stimulated microglia, the Das lab sought to microglial-promoted neuroinflammation via CB2
investigate these molecules under inflammatory activation [58]. In fact, there is a current drive
conditions [44, 57]. By monitoring the produc- towards the development of CB2-selective ago-
tion of EDP-EAs in LPS-stimulated microglia nists to combat the inflammatory pathology asso-
treated with DHEA, they confirmed that EDP- ciated with progression of neurodegenerative
EAs are produced by CYPs as inhibitors of disease. As the DHEA epoxides (EDP-EAs) are
CYPs reduce their levels. It was further demon- naturally produced by microglia and the microglia
strated that the CYP isozyme, CYP2J2, is express CYPs, the Das group examined the anti-
responsible for this action. Current research is inflammatory effects of these molecules in an in
going on to identify other brain CYPs that are vitro model of neuroinflammation. As mentioned
involved in eCB metabolism. Interestingly, the briefly above, microglial cells are the innate
production of the terminal epoxide, immune cells of the brain, which continuously
19,20-EDP-EA is favored over the other regioi- survey the brain parenchyma for harmful stimuli.
somers in a reconstituted CYP2J2-CYP When harmful stimuli are encountered, microglia
Reductase (CPR) Nanodisc system and by brain exhibit a pro-inflammatory phenotype to promote
microsomes [13]. More research is needed to defense. As microglia clear the harmful stimuli, a
further understand why certain regioisomers are phenotype change occurs that promotes an inflam-
preferred by CYPs. mation-resolution phase. This phenotype switch-
As previous studies consistently showed ing mechanism is classically known as
DHEA as a weak ligand at both CB1 and CB2, macrophage polarization. Out of the EDP-EAs
the Das lab tested to see if DHEA and its epoxide tested, 19,20-EDP-EA showed the most potent
metabolites activated or bound to CB receptors dose-dependent reduction of pro- inflammatory
[56]. Using the PRESTO-Tango assay, they markers. (Fig. 17.2). Correspondingly, it decreased
17 Novel Anti-inflammatory and Vasodilatory ω-3 Endocannabinoid Epoxide Regioisomers 225
pro-inflammatory cytokine IL-6 and increased the 17.2.5 ω-3 Endocannabinoid EPA
production of IL-10 [13]. Additionally, the anti- Ethanolamide (EPEA)
inflammatory effects of 19,20-EDP-EA were con-
firmed using freshly isolated primary piglet The report that several fatty acid ethanolamides
microglial cells stimulated with LPS, which sup- other than AEA have unique activities at the can-
pressed IL-6 production, and thus corroborated nabinoid receptors sparked a study in 1996 by the
the findings in immortalized BV-2 cells. Based on Martin and Razdan groups, which sought to
these findings, it is possible that the DHA-derived examine the structure-activity relationships of
eCB epoxides mediate anti-inflammatory activi- AEA analogs at CB1 [60, 61]. Previous to this
ties in addition to other pathways. Taken together, study, AEA was the only endogenously produced
this indicates that the EDP-EAs and their stable fatty acid ethanolamide to bind and activate the
derivatives may be utilized for treating neuroin- cannabinoid receptors. By understanding the
flammatory conditions. structural requirements necessary for ligands to
bind CB1 it would be possible to develop more
potent ligands for both biological and therapeutic
17.2.4 Vasoactivity of DHA applications. Cannabinoid structure-activity rela-
Ethanolamide Epoxide tionships (SAR) indicated that a phenolic
(EDP-EA) hydroxyl, a lipophilic side chain, and an appro-
priately oriented carbocyclic ring system are
It is well known that CYP epoxygenases convert required for cannabinoid receptor activity. One of
ω-3 PUFAs to PUFA epoxides. For example, the AEA analogs tested in their study was EPA
DHA is converted to DHA epoxides (EDPs). ethanolamide (EPEA). Based on molecular
These molecules are vasoactive in nature, and are dynamic simulations, EPEA was suggested to
more effective at relaxing pre-constricted vessels possess some affinity for CB1 [61].
compared to CYP-derived AA epoxides (EETs) Alongside these studies, the Di Marzo group
[59]. The Das lab demonstrated that CYP determined the in vivo localization of EPEA in
epoxygenase- mediated metabolism of DHEA mouse brains. EPEA was detected at
produced a vasodilatory eCB epoxides, 0.18 ± 0.09 pmol/mg of brain lipid extract, which
19,20-EDP-EA. Compared to 19,20-EDP, as was the lowest level of all of the fatty acid etha-
shown in Fig. 17.1, EDP-EA had a two-fold nolamides detected in their study. EPEA was
higher EC50 at relaxing bovine coronary arteries found at five-fold lower levels than DHEA [53].
(Fig. 17.2) [13]. Although CYP-derived DHEA Compared to EPEA, its parent molecule EPA
epoxides are vasoactive molecules, they do so was found at 443 ± 167 ng/g mice brain.
with reduced potency relative to their parent mol- However, dietary supplementation with EPA
ecule. Furthermore, 19,20-EDP-EA exerts anti- increased the endogenous levels of EPEA,
angiogenic effects in HMVEC cells. Angiogenesis thereby providing a dietary delivery method for
plays several roles in pathological and non- elevating ω -3 eCBs [15]. Furthermore, when
pathological states. As eCBs are regarded as anti- mice were fed a high ω -3 diet consisting of DHA
cancer molecules through their anti-angiogenic and EPA phospholipids, the levels of EPEA
effects, the Das lab tested the ability of increased in adipose tissue, corroborating the
19,20-EDP-EA to inhibit angiogenesis in link between diet and the eCB system. More
HMVEC cells. 1 μM and 3 μM 19,20-EDP-EA recently, a targeted lipidomics method using
decreased tubulogensis by 31 and 75%, respec- LC-MS/MS detected EPEA in rat brain
tively. Previous to this study, the anti-angiogenic (6.2 ± 1.6 pmol/g), heart (9.4 ± 6 pmol/g), kidney
actions of the EDP metabolites were attributed to (37.1 ± 21 pmol/g), spleen (9.2 ± 4.1 pmol/g),
their inhibition of VEGF-stimulated cell migra- liver (80.8 ± 20.1 pmol/g), and pig brain
tion via an unknown epoxyeicosanoic metabolic (2.2 ± 0.4 pmol/g) [13]. Interestingly, EPEA is
pathway [13]. found at a significantly lower level than DHEA
226 L. N. Carnevale and A. Das
in rat brain (DHEA: 128.9 ± 27.9 pmol/g). molecule EPEA. For example, 17,18-EEQ-EA
Together, these findings support that supplemen- had a lower EC50 at CB2 than EPEA (EC50
tation with ω -3 fatty acids can modulate eCB 17,18-EEQ-EA: 1.4 nM and EC50 EPEA:
levels and may increase the bioavailability of 2.1 nM). However, compared to EPEA (EC50
DHA and EPA for physiological function. CB1: 0.1 nM), 17,18-EEQ-EA had a lower
Although the biosynthesis of EPEA has not potency at CB1 (EC50 CB1: 18.5 nM), thus
been proved, the well-studied transacylation- showing a CB2-selectivity for 17,18-EEQ-EA
PDE pathway suggests that n-acyl ethanol- [13] (Fig. 17.2). As mentioned above, discover-
amides are biosynthesized from ing novel CB2-selective molecules may aid in
glycerophospholipids via N-acylphosphatidyl the development of anti-inflammatory therapeu-
ethanolamine (NAPE). Through this pathway, a tics that overcome any potential psychoactive
Ca2+-dependent N-acyltransferase and NAPE- activities that may be mediated through CB1
hydrolyzing PLD render the fatty acid ethanol- activation.
amide free from its membrane precursor [56].
This pathway is similar to the biosynthesis of
AEA and may support the idea of a conserved 17.2.7 Anti-inflammatory Activity
biosynthesis for fatty acid ethanolamides. of EPA Ethanolamide Epoxides
(EEQ-EAs)
17.2.6 ω-3 Endocannabinoid Epoxide Since EPA and EPEA have been shown to pos-
Epoxyeicosatetraenoic Acid- sess anti-inflammatory activities, the anti-
Ethanolamide (EEQ-EA) inflammatory activities of EEQ-EAs were studied
in brain microglia. When BV-2 microglia were
In 2017, the Das lab showed that EPEA, like stimulated with LPS, the levels of EPEA and
DHEA, is a substrate for the CYP epoxygenase total EEQ-EA were elevated. Interestingly, EPEA
pathway. Using a targeted lipidomics LC-MS/MS levels significantly surpassed DHEA levels after
approach they detected and quantified all five 12 h of LPS stimulation (DHEA:
EPEA epoxide (EEQ-EA) regioisomers in rat 6.7 ± 4.6 pmol/106 cells and EPEA:
brain, heart, kidney, spleen, liver tissues as well 2042 ± 939.2 pmol/106 cells). Furthermore, in
as in mouse microglial cells. They found that LPS-stimulated BV-2 cells, 17,18-EEQ-EA
LPS-stimulated and EPEA-treated mouse (10 μM) dose-dependently reduced NO in a CB2-
microglia could convert EPEA to the EEQ-EAs dependent manner, which was attenuated with
via CYP epoxygenases. They further determined CB2 antagonist, AM-630, treatment. In addition,
that one of the CYP isozymes responsible for this 17,18-EEQ-EA dose-dependently decreased IL-6
action is CYP2J2. Using recombinantly expressed and increased IL-10 (Fig. 17.2). Interestingly,
proteins, CYP2J2-CPR Nanodiscs showed a 17,18-EEQ-EA was a more potent inhibitor of
favored production of 17,18-EEQ-EA, as shown NO production than EPA epoxide (17,18-EEQ).
in Fig. 17.1, compared to other regioisomers [13]. This evidence shows that the CYP epoxygenase
To further characterize EPEA and its epoxide pathway may play be responsible for the anti-
metabolites, we were interested in testing inflammatory nature of EPA through conversion
whether the EEQ-EAs were eCBs. Previous of EPEA to its epoxide metabolite. Lastly, similar
studies consistently showed EPEA as a weak to 19,20-EDP-EA, the effects of 17,18-EEQ-EA
agonist for CB1 and CB2 [56]. Through the were confirmed in freshly isolated piglet microg-
PRESTO-Tango assay, the Das lab showed that lia cells, thereby corroborating the anti-
epoxidation of EPEA by CYPs led to an altered inflammatory effects of 17,28-EEQ-EA in
CB1 and CB2 activity compared to their parent primary cells.
17 Novel Anti-inflammatory and Vasodilatory ω-3 Endocannabinoid Epoxide Regioisomers 227
bioactive signals∗. J Biol Chem 286:31532–31541. piglets. Proc Natl Acad Sci 98:6402–6406. https://doi.
https://doi.org/10.1074/jbc.M111.237990 org/10.1073/pnas.101119098
43. Aoki M, Matsumoto NM, Okubo Y, Ogawa R (2019) 54. Dennis M (2010) Inflammatory hyperalgesia induces
Bioelectrochemistry cytochrome P450 genes play essential bioactive lipid production in the spinal cord.
central roles in transcriptional response by keratino- J Neurochem 114:981–993
cytes to a high-voltage alternating current electric fi 55. Bisogno T, Delton-Vandenbroucke I, Milone A,
eld. Bioelectrochemistry 126:163–171. https://doi. Lagarde MDMV (1999) Biosynthesis and inactivation
org/10.1016/j.bioelechem.2018.11.014 of N-arachidonoylethanolamine (anandamide) and
44. Snider NT, Nast JA, Tesmer LA, Hollenberg PF
N-docosahexaenoylethanolamine in bovine retina.
(2009) A cytochrome P450-derived epoxygenated Arch Biochem Biophys 370:300–307
metabolite of anandamide is a potent cannabinoid 56. Meijerink J, Balvers M, Witkamp R (2013) N-acyl
receptor 2-selective agonist. Mol Pharmacol 75:965– amines of docosahexaenoic acid and other n-3 poly-
972. https://doi.org/10.1124/mol.108.053439 unsatured fatty acids – from fishy endocannabinoids
45. Snider NT, Walker VJ, Hollenberg PF (2010)
to potential leads. Br J Pharmacol 169:772–783.
Oxidation of the endogenous cannabinoid arachi- https://doi.org/10.1111/bph.12030
donoyl ethanolamide by the cytochrome P450 mono- 57. Cui X, Kawashima H, Barclay TB et al (2001)
oxygenases: physiological and pharmacological Molecular cloning and regulation of expression of
implications. Pharmacol Rev 62:136–154. https://doi. two novel mouse CYP4F genes: expression in peroxi-
org/10.1124/pr.109.001081 some proliferator-activated receptor alpha-deficient
46. Walker VJ, Griffin AP, Hammar DK, Hollenberg
mice upon lipopolysaccharide and clofibrate chal-
PF (2016) Metabolism of anandamide by human lenges. J Pharmacol Exp Ther 296:542–550
cytochrome P450 2J2 in the reconstituted system 58. Maresz K, Carrier EJ, Ponomarev ED et al (2005)
and human intestinal microsomes. J Pharmacol Modulation of the cannabinoid CB2 receptor
Exp Ther 357:537–544. https://doi.org/10.1124/ in microglial cells in response to inflammatory
jpet.116.232553 stimuli. J Neurochem 95:437–445. https://doi.
47. Kalish BT, Kieran MW, Puder M, Panigrahy D (2013) org/10.1111/j.1471-4159.2005.03380.x
The growing role of eicosanoids in tissue regen- 59. Zhang G, Panigrahy D, Mahakian LM et al (2013)
eration, repair, and wound healing. Prostaglandins Epoxy metabolites of docosahexaenoic acid (DHA)
Other Lipid Mediat 104–105:130–138. https://doi. inhibit angiogenesis, tumor growth, and metasta-
org/10.1016/j.prostaglandins.2013.05.002 sis. Proc Natl Acad Sci 110:6530–6535. https://doi.
48. Node K, Huo Y, Ruan X et al (1999) Anti-inflammatory org/10.1073/pnas.1304321110
properties of cytochrome P450 epoxygenase-derived 60. Hanuš L, Gopher A, Almog S, Mechoulam R (1993)
eicosanoids. Science 285(80):1276–1279. https://doi. Two new unsaturated fatty acid ethanolamides in
org/10.1126/science.285.5431.1276 brain that bind to the cannabinoid receptor. J Med
49. Zhao H, Chen J, Chai J et al (2017) Cytochrome Chem 36:3032–3034. https://doi.org/10.1021/
P450 (CYP) epoxygenases as potential targets in the jm00072a026
management of impaired diabetic wound healing. 61. Thomas BF, Adams IB, Mascarella SW et al (1996)
Lab Investig 97:782–791. https://doi.org/10.1038/ Structure-activity analysis of anandamide ana-
labinvest.2017.21 logs: relationship to a cannabinoid pharmacophore.
50.
McDougle DR, Kambalyal A, Meling DD, J Med Chem 39:471–479. https://doi.org/10.1021/
Das A (2014) Endocannabinoids anandamide jm9505167
and 2-Arachidonoylglycerol are substrates for 62. Morisseau C, Inceoglu B, Schmelzer K et al (2010)
human CYP2J2 Epoxygenase. J Pharmacol Exp Naturally occurring monoepoxides of eicosapentae-
Ther 351:616–627. https://doi.org/10.1124/ noic acid and docosahexaenoic acid are bioactive
jpet.114.216598 antihyperalgesic lipids. J Lipid Res 51:3481–3490.
51. Snider NT, Kornilov AM, Kent UM, Hollenberg PF https://doi.org/10.1194/jlr.M006007
(2007) Anandamide metabolism by human liver and 63. Coussens LM, Werb Z (2002) Inflammation and can-
kidney microsomal cytochrome p450 enzymes to form cer. Pharmaceut Biotechnol 420(6917)860. Nature
hydroxyeicosatetraenoic and epoxyeicosatrienoic acid 420:860–867. https://doi.org/10.1038/nature01322.
ethanolamides. J Pharmacol Exp Ther 321:590–597. Inflammation
https://doi.org/10.1124/jpet.107.119321.hydrolysis 64. Jaudszus A, Gruen M, Watzl B et al (2013) Evaluation
52. Meunier B, de Visser SP, Shaik S (2004) Mechanism of suppressive and pro-resolving effects of EPA and
of oxidation reactions catalyzed by cytochrome P450 DHA in human primary monocytes and T-helper cells.
enzymes. Chem Rev 104:3947–3980. https://doi. J Lipid Res 54:923–935. https://doi.org/10.1194/jlr.
org/10.1021/cr020443g P031260
53. Berger A, Crozier G, Bisogno T et al (2001)
65. Laviano A, Rianda S, Molfino A, Fanelli FR (2013)
Anandamide and diet: inclusion of dietary arachido- Omega-3 fatty acids in cancer. Curr Opin Clin Nutr
nate and docosahexaenoate leads to increased brain Metab Care 16:156–161. https://doi.org/10.1097/
levels of the corresponding N-acylethanolamines in MCO.0b013e32835d2d99
232 L. N. Carnevale and A. Das
66.
Cui PH, Petrovic N, Murray M (2011) The 71. Chen JK, Chen JK, Imig JD et al (2008) Identification
ω-3 epoxide of eicosapentaenoic acid inhibits of novel endogenous cytochrome P450 arachidonate
endothelial cell proliferation by p38 MAP kinase
metabolites with high affinity for cannabinoid recep-
activation and cyclin D1/CDK4 down-regula- tors. J Biol Chem 283:24514–24524. https://doi.
tion. Br J Pharmacol 162:1143–1155. https://doi. org/10.1074/jbc.M709873200
org/10.1111/j.1476-5381.2010.01113.x 72. Bisogno T, Melck D, Bobrov M et al (2000) N-acyl-
67. Guodong Z, Dipak P, Lisa MM et al (2013) Epoxy dopamines: novel synthetic CB1 cannabinoid-receptor
metabolites of docosahexaenoic acid (DHA) inhibit ligands and inhibitors of anandamide inactivation with
angiogenesis, tumor growth, and metastasis. Proc Natl cannabimimetic activity in vitro and in vivo. Biochem
Acad Sci 110:6530–6535. https://doi.org/10.1073/ J 351:817. https://doi.org/10.1042/bj3510817
pnas.1304321110/-/DCSupplemental. www.pnas.org/ 73. Fowler CJ, Tiger G, López-Rodríguez ML et al (2003)
cgi/doi/10.1073/pnas.1304321110 Inhibition of fatty acid amidohydrolase, the enzyme
68. Roy J, Watson JE, Hong IS et al (2018) Antitumorigenic responsible for the metabolism of the endocannabi-
properties of omega-3 endocannabinoid epoxides. noid anandamide, by analogues of arachidonoyl-
J Med Chem 61:5569–5579. https://doi.org/10.1021/ serotonin. J Enzyme Inhib Med Chem 18:225–231.
acs.jmedchem.8b00243 https://doi.org/10.1080/1475636031000080216
69. Hammock BD, Wagner Karen IB (2011) The soluble 74. Wang Y, Balvers MGJ, Hendriks HFJ et al (2017)
epoxide hydrolase as a pharmaceutical target for pain Docosahexaenoyl serotonin emerges as most potent
management. Pain Manag 50:383–386. https://doi. inhibitor of IL-17 and CCL-20 released by blood
org/10.1097/FJC.0b013e3181506445 mononuclear cells from a series of N-acyl serotonins
70. Sasso O, Wagner K, Morisseau C et al (2015)
identified in human intestinal tissue. Biochim Biophys
Peripheral FAAH and soluble epoxide hydro- Acta Mol Cell Biol Lipids 1862:823–831. https://doi.
lase inhibitors are synergistically antinociceptive. org/10.1016/j.bbalip.2017.05.008
Pharmacol Res 97:7–15. https://doi.org/10.1016/j.
phrs.2015.04.001
Overview of Lipid Biomarkers
in Amyotrophic Lateral Sclerosis 18
(ALS)
Andres Trostchansky
obtained about lipid help in the development of processing and compromised mitochondria
drugs for the treatment of ALS. homeostasis, increased oxidative stress, excito-
toxicity, reduced neurotrophic support, and glial
inflammatory responses [7]. Familiar ALS has
18.2 Amyotrophic Lateral been studied using active (SOD1G93A, SOD1G37R)
Sclerosis mice mutants which have a phenotype character-
ized by progressive paralysis and death caused by
Amyotrophic lateral sclerosis is a multifactorial the degeneration of MN, in addition to gliosis
neurodegenerative disease caused by genetic and within the spinal cord, brain stem and cortex [6].
non-inheritable components leading to MN These features suggest a gain of toxic function of
degeneration in the spinal cord, brain stem and the mutant SOD might play a role in the neurode-
primary motor cortex [1]. It is well established generative process. In addition, oxidative stress is
that ALS affects both cortical as well as cranial an important contributor to neuronal death in
and spinal MN with variable evolution. The ALS involving astrocytes [8]. Current research
majority of ALS cases are sporadic, however has shown that ALS is not a single cell type dis-
there exists a 10% of familial cases, some of ease, since it involves microglia and astrocytes
which arise from mutations of the protein surrounding the MN. In fact, our laboratory
Superoxide Dismutase 1(SOD1) [1]. The average reported astrocytes promote neuronal loss by
age of onset of the disease is 55 years, beginning mechanisms involving alterations in mitochon-
later in women than men, and with 90% of the drial functionality, increased production of nitric
patients dying within 5 years after the appearance oxide and nerve growth factor [2, 8–13]. Glial
of the clinical symptoms. For a definitive diagno- cells are the principle innate immune cell of the
sis of ALS the progressive spread of symptoms CNS and pathology associated with these cells is
involving upper and lower motor neurons is referred to as neuroinflammation, a hallmark of
required, according to the revised El Escorial and ALS [14, 15].
the Awaji criteria [4, 5]. The most used clinical Importantly, there exists an increase of neuro-
scores for disease progression is the ALS func- toxic molecules such as pro-inflammatory cyto-
tional rating scale-revised (ALSFRS-R) [5]. This kines, reactive oxygen species (ROS) and
scale is employed to evaluate the pace of disease pro-inflammatory lipid-derived compounds.
progression, predict survival and assess the These molecules may cause further neuronal
effects of disease-modifying drugs in therapeutic damage leading to further glial cell activation
trials. The scale is based on clinical symptoms so resulting in a positive feedback loop of neuroin-
it is imperative to discover biomarkers that allow flammation. Indeed, neuroinflammatory pro-
early diagnosis of the disease. In addition, bio- cesses have been detected in ALS patients as well
markers will aid for therapeutic drug design to as in transgenic models of the disease [16]. Based
improve the quality of life of those affected due on this background, many treatments aim to
to the lack of effective pharmacological treat- inhibit or reduce the pro-inflammatory action of
ments for ALS [5]. The pathogenesis of ALS con- microglia and astrocytes and counteract the pro-
sists of two stages: an early neuroprotective stage gression of the disease. Unfortunately, none of
and a later neurotoxic stage. Multiple mecha- the successful therapies in ALS animal models
nisms have been described for MN death includ- has improved clinical outcomes in patients with
ing glutamate toxicity, mitochondrial dysfunction, ALS [3]. This can be ascribed to different factors.
protein misfolding and apoptosis [6]. However, For example, decreasing or deleting single pro-
ALS appears as a composite syndrome where the inflammatory factors such as TNF-α, IL1-β, and
aberrant cellular pathways may not be ascribed inducible nitric oxide synthase has had little-to-no
solely from a protein misfolding issue, but cellu- effect on overall survival of SOD1G93A mice [8,
lar physiology aspects like deficiencies in RNA 12]. These suggest that a multiplicity of pro-
18 Lipids in ALS 235
inflammatory cytokines can compensate the arachidonic acid (AA) and docosahexaenoic acid
absence of any single factor, and it is unlikely (DHA). Since these PUFA are the precursors of
that continuing efforts to target a single factor in important cell signaling molecules. As examples,
humans will provide significant therapeutic ben- DHA and AAs can be oxidized to give rise to
efit in patients with ALS. Considering the impor- prostaglandins or leukotrienes. The oxidation of
tance of the disease and the fact that inflammatory DHA produces neuroprotectinD1, a signaling
processes are involved, the identification of blood molecule that promotes cell survival under stress.
biomarkers to identify individuals at risk of The latter is part of the denominated specialized
developing the disease or for their recruitment in pro-resolving lipid mediators (SPM) giving to
clinical studies in order to analyze benefits in the these PUFA precursors an important role in neu-
development of drugs for the treatment of ALS roinflammation [20, 21]. The presence of these
would be of biological relevance. Below we will compounds has been implicated in neuronal sig-
discuss the identification and use of lipid-derived naling processes controlling neurogenesis, brain
products as biomarkers since research in the field vesicular activity, central glucose homeostasis,
and the optimization of lipidomic techniques are mood and cognition [22]. Other example is
giving novel and important new data for using Prostaglandin E2 (PGE2), synthetized by
them as disease biomarkers. Prostaglandin Endoperoxide H Synthase-2
(PGHS-2) from AA, which promotes inflamma-
tion after binding to its receptor [23]. Their role
18.3 R
ole of Lipids in Central as biomarkers or footprints of ALS will be dis-
Nervous System and ALS cussed later.
In many neurodegenerative diseases, an accu-
Lipids are implicated in a wide variety of biologi- mulation of ceramides has been observed. The
cal processes and can be classified into five cate- latter is considered toxic, since it has been shown
gories: fatty acids, triacylglycerols (TAGs), to promote neuronal death by oxidative stress and
phospholipids, sterol lipids and sphingolipids. apoptosis in both animal models and patients
The brain requires a near constant source of [24–28]. Moreover it has been proposed in
metabolites to maintain function, and contains SOD1G93A mice that sphingomyelin-associated
the second highest concentration of lipids in the ROS production leads to MN death through the
human body [17]. Although the consensus is that p75 factor [12]; in addition, it is reported that an
glucose metabolism almost completely satisfies increase of sphingomyelin in the spinal cord of
the brain energy requirements, it has recently ALS patients mediates MN death via oxidative
been shown that approximately 20% of the total stress [24]. From a structural-functional point of
energy requirement of the brain is met through view, it has been shown that membrane fluidity in
the oxidation of fatty acids, and that this fatty the brain and spinal cord decreases significantly
acid oxidation may take place entirely in astro- over the course of disease in ALS mice [29]. In
cytes [18]. Besides this fact, perhaps the most fact, neuronal membranes rich in phosphatidyl-
essential role for lipids in the brain is as compo- ethanolamine and phosphatidylserine are signifi-
nents of the membrane cellular machinery. This cantly less fluid. Thus, the increase in DHA
is important in the brain since changes in mem- results in more rigid membranes with an impor-
brane fluidity and many signaling processes tant impact of signaling activities in ALS brains
occur in specific intracellular compartments. [30].
Importantly, fatty acids and their derivatives have Along with neuronal degeneration, alterations
been well characterized as drivers of intracellular of lipid metabolism have been reported in
signaling processes [19]. ALS. Different studies have shown that muscle
Two major polyunsaturated fatty acids denervation has a role in the promotion of
(PUFAs) are present at high levels at the brain: abnormal lipid metabolism. In fact, Body Mass
236 A. Trostchansky
studies have shown that levels of 16:1 and 18:1 18.4.2 Complex Lipids
fatty acids, as well as of the enzyme that
introduces the instaurations at the carbon chain, Complex lipids as triglycerides (TG), total cho-
stearoyl-CoA desaturase, are significantly lesterol (TC), phospholipids (PL) were all ana-
increased in blood cells from ALS patients com- lyzed as biomarkers of ALS onset, progression or
pared to healthy controls [4, 44, 45]. It is impor- drug effect on clinical symptoms [43]. The levels
tant to note that the ratio of different of TG and TC in ALS patients were comparable
monounsaturated fatty acids was strongly to those found in control subjects. A recent study
increased in ALS patients. In particular, 16:1 and analyzed the composition of lipids in cerebrospi-
18:1 levels increased significantly in blood cells, nal fluid (CSF) from ALS patients compared to
and higher oleate levels were also observed in controls [43]. In this study, the authors demon-
serum [4]. Fatty acids levels in the blood corre- strated that phosphatidylcholine PC(36:4),
lated with ALSFRS-R, the 16:1/16:0 ratio in ceramide and glucosylceramide levels were
blood cells negatively correlated with ALSFRS-R higher in ALS patients than in control subjects.
decline over a six-month period [4]. Circulating Bioinformatics and statistical analysis showed
free fatty acids were more abundant in ALS six different metabolites that can be followed as
patients, suggesting increased lipid breakdown distinctive between ALS and control groups.
by adipocytes [46]. In fact, a decline of These compound corresponded to the PC deriva-
ALSFRS-R was also significantly associated tives PC (36:2p), PC(36:4p), PC(40:6p), the
with the levels of 16:1 itself. Survival rates were sphingomyelin (SM) derivatives SM(d34:0), and
greater when associated to higher 16:1/16:0 SM(d39:1), and the TG (Triglyceride)
ratios, with a prolonged life expectancy of almost (16:1/18:1/18:2) [43]. The PC and SM com-
11 months in the population of patients with a pounds levels were higher in ALS patients, in
high 16:1/16:0 ratio, as compared to patients with contrast to TG which was lower in ALS patients
low 16:1/16:0 ratio. Higher levels of palmitoleate than in controls. It is worth noting the higher
itself were also significantly associated with level of PC (36:4p and 36:4e) was the strongest
extended life span. In contrast to what was discriminant factor identified by all the statistical
observed for the 16:1/16:0 ratio, no changes in approaches used [43]. However, and importantly,
survival were observed associated to the higher levels of SM(d43:2) were associated to a
18:1/18:0 ratio [4]. Moreover, BMI or circulating lesser decline of the ALSFRS-r score [43].
leptin content did not correlate with ALSFRS-R Similar studies were performed using trans-
decline, nor prognosticated survival based on genic ALS mice. In these experiments, the results
multivariate analysis. In fact, statistical analysis were quite different from those of humans, high-
showed that blood cell 16:1/16:0 ratio was an lighting the difficulties in transferring the data
independent prognostic factor for survival with obtained in animals to humans. In this case the
age, BMI, site of onset and ALSFRS-R as vari- complex lipids, which were found to be discrimi-
ables [4]. The 16:1/16:0 index is an easy-to- nant between control and ALS mice, were PC
handle parameter that predicts survival of ALS (36:2), PC(36:4), PC(40:6). These three lipids
patients. The enzymatically and non-were found in higher levels in the ALS groups
enzymatically oxidation of unsaturated fatty than in controls [43].
acids increase during oxidative stress and may We have previously discussed that accumula-
reflect the presence of an inflammatory process. tion of ceramides (Cer) are toxic for MN [16]. By
Lipid peroxidation susceptibility, assessed by the contrast, increasing the formation and accumula-
peroxidability index (PI) [47], was 16% lower in tion of Cer-derived agents may be protective by
the blood cell fraction of ALS patients as com- reducing ceramide synthesis thus limiting the
pared to controls, independent of the omega-6/ direct toxic effect on MN [16]. The membrane
omega-3 (AA/EPA) index which did not show lipids Glycosphingolipids (GSLs) are a heteroge-
significant modification [4]. neous lipid groups formed through the covalent
238 A. Trostchansky
indicators of ALS. The analysis we did in this 11. Pehar M, Cassina P, Vargas MR, Castellanos R, Viera
L, Beckman JS, Estevez AG, Barbeito L (2004)
review suggests that the benefits of detecting Astrocytic production of nerve growth factor in motor
these biomarkers may help in designing and per- neuron apoptosis: implications for amyotrophic lat-
forming clinical trials whose aim should be not eral sclerosis. J Neurochem 89(2):464–473
only increase life expectancy but also quality of 12. Pehar M, Vargas MR, Robinson KM, Cassina P, Diaz-
Amarilla PJ, Hagen TM, Radi R, Barbeito L, Beckman
life. JS (2007) Mitochondrial superoxide production and
nuclear factor erythroid 2-related factor 2 activation
in p75 neurotrophin receptor-induced motor neuron
References apoptosis. J Neurosci 27(29):7777–7785
13. Vargas MR, Pehar M, Cassina P, Martinez-Palma
L, Thompson JA, Beckman JS, Barbeito L (2005)
1. Al-Chalabi A, Hardiman O (2013) The epidemiology
Fibroblast growth factor-1 induces heme oxygenase-1
of ALS: a conspiracy of genes, environment and time.
via nuclear factor erythroid 2-related factor 2 (Nrf2)
Nat Rev Neurol 9(11):617–628
in spinal cord astrocytes: consequences for motor
2. Barbeito LH, Pehar M, Cassina P, Vargas MR, Peluffo
neuron survival. J Biol Chem 280(27):25571–25579
H, Viera L, Estevez AG, Beckman JS (2004) A role for
14. Boillee S, Cleveland DW (2008) Revisiting oxidative
astrocytes in motor neuron loss in amyotrophic lateral
damage in ALS: microglia, Nox, and mutant SOD1.
sclerosis. Brain Res Brain Res Rev 47(1–3):263–274
J Clin Invest 118(2):474–478
3. Gros-Louis F, Gaspar C, Rouleau GA (2006) Genetics
15. West M, Mhatre M, Ceballos A, Floyd RA, Grammas
of familial and sporadic amyotrophic lateral sclerosis.
P, Gabbita SP, Hamdheydari L, Mai T, Mou S, Pye
Biochim Biophys Acta 1762(11–12):956–972
QN et al (2004) The arachidonic acid 5-lipoxygenase
4. Henriques A, Blasco H, Fleury MC, Corcia P,
inhibitor nordihydroguaiaretic acid inhibits tumor
Echaniz-Laguna A, Robelin L, Rudolf G, Lequeu
necrosis factor alpha activation of microglia and
T, Bergaentzle M, Gachet C et al (2015) Blood cell
extends survival of G93A-SOD1 transgenic mice.
palmitoleate-palmitate ratio is an independent prog-
J Neurochem 91(1):133–143
nostic factor for amyotrophic lateral sclerosis. PLoS
16. Dodge JC, Treleaven CM, Pacheco J, Cooper S,
One 10(7):e0131512
Bao C, Abraham M, Cromwell M, Sardi SP, Chuang
5. Brooks BR, Miller RG, Swash M, Munsat TL, World
WL, Sidman RL et al (2015) Glycosphingolipids
Federation of Neurology Research Group on Motor
are modulators of disease pathogenesis in amyo-
Neuron D (2000) El Escorial revisited: revised cri-
trophic lateral sclerosis. Proc Natl Acad Sci USA
teria for the diagnosis of amyotrophic lateral sclero-
112(26):8100–8105
sis. Amyotrophic Lateral Scler Other Motor Neuron
17. Hamilton JA, Brunaldi K (2007) A model for fatty
Disord 1(5):293–299
acid transport into the brain. J Mol Neurosci MN
6. Philips T, Rothstein JD (2015) Rodent models of
33(1):12–17
amyotrophic lateral sclerosis. Curr Protoc Pharmacol
18. Ebert D, Haller RG, Walton ME (2003) Energy con-
69(5):671–621
tribution of octanoate to intact rat brain metabolism
7. Rossi S, Cozzolino M, Carri MT (2016) Old versus
measured by 13C nuclear magnetic resonance spec-
new mechanisms in the pathogenesis of ALS. Brain
troscopy. J Neurosci 23(13):5928–5935
Pathol 26(2):276–286
19. Gutierrez J, Ballinger SW, rley-Usmar VM, Landar
8. Cassina P, Cassina A, Pehar M, Castellanos R,
A (2006) Free radicals, mitochondria, and oxidized
Gandelman M, de Leon A, Robinson KM, Mason RP,
lipids: the emerging role in signal transduction in vas-
Beckman JS, Barbeito L et al (2008) Mitochondrial
cular cells. Circ Res 99(9):924–932
dysfunction in SOD1G93A-bearing astrocytes pro-
20.
Bazan NG, Molina MF, Gordon WC (2011)
motes motor neuron degeneration: prevention by
Docosahexaenoic acid signalolipidomics in nutrition:
mitochondrial-targeted antioxidants. J Neurosci
significance in aging, neuroinflammation, macular
28(16):4115–4122
degeneration, Alzheimer’s, and other neurodegenera-
9. Miquel E, Cassina A, Martinez-Palma L, Bolatto C,
tive diseases. Annu Rev Nutr 31:321–351
Trias E, Gandelman M, Radi R, Barbeito L, Cassina P
21. Bazan NG, Musto AE, Knott EJ (2011) Endogenous
(2012) Modulation of astrocytic mitochondrial func-
signaling by omega-3 docosahexaenoic acid-derived
tion by dichloroacetate improves survival and motor
mediators sustains homeostatic synaptic and circuitry
performance in inherited amyotrophic lateral sclero-
integrity. Mol Neurobiol 44(2):216–222
sis. PLoS One 7(4):e34776
22. Bazinet RP, Laye S (2014) Polyunsaturated fatty acids
10. Miquel E, Cassina A, Martinez-Palma L, Souza JM,
and their metabolites in brain function and disease.
Bolatto C, Rodriguez-Bottero S, Logan A, Smith RA,
Nat Rev Neurosci 15(12):771–785
Murphy MP, Barbeito L et al (2014) Neuroprotective
23. Miyagishi H, Kosuge Y, Takano A, Endo M, Nango
effects of the mitochondria-targeted antioxidant
H, Yamagata-Murayama S, Hirose D, Kano R, Tanaka
MitoQ in a model of inherited amyotrophic lateral
Y, Ishige K et al (2017) Increased expression of
sclerosis. Free Radic Biol Med 70:204–213
15-Hydroxyprostaglandin dehydrogenase in spinal
240 A. Trostchansky
astrocytes during disease progression in a model of fatty acid eicosapentaenoic acid accelerates disease
amyotrophic lateral sclerosis. Cell Mol Neurobiol progression in a model of amyotrophic lateral sclero-
37(3):445–452 sis. PLoS One 8(4):e61626
24. Cutler RG, Pedersen WA, Camandola S, Rothstein 36. Parakh S, Spencer DM, Halloran MA, Soo KY, Atkin
JD, Mattson MP (2002) Evidence that accumulation JD (2013) Redox regulation in amyotrophic lateral
of ceramides and cholesterol esters mediates oxida- sclerosis. Oxidative Med Cell Longev 2013:408681
tive stress-induced death of motor neurons in amyo- 37. Dupuis L, Oudart H, Rene F, Gonzalez de Aguilar
trophic lateral sclerosis. Ann Neurol 52(4):448–457 JL, Loeffler JP (2004) Evidence for defective energy
25. Brugg B, Michel PP, Agid Y, Ruberg M (1996)
homeostasis in amyotrophic lateral sclerosis: benefit
Ceramide induces apoptosis in cultured mesence- of a high-energy diet in a transgenic mouse model.
phalic neurons. J Neurochem 66(2):733–739 Proc Natl Acad Sci USA 101(30):11159–11164
26. France-Lanord V, Brugg B, Michel PP, Agid Y,
38. Mattson MP, Cutler RG, Camandola S (2007)
Ruberg M (1997) Mitochondrial free radical signal Energy intake and amyotrophic lateral sclerosis.
in ceramide-dependent apoptosis: a putative mecha- NeuroMolecular Med 9(1):17–20
nism for neuronal death in Parkinson's disease. 39. Desport JC, Preux PM, Magy L, Boirie Y, Vallat JM,
J Neurochem 69(4):1612–1621 Beaufrere B, Couratier P (2001) Factors correlated
27. Bras J, Singleton A, Cookson MR, Hardy J (2008) with hypermetabolism in patients with amyotrophic
Emerging pathways in genetic Parkinson’s disease: lateral sclerosis. Am J Clin Nutr 74(3):328–334
potential role of ceramide metabolism in Lewy body 40. Dupuis L, Pradat PF, Ludolph AC, Loeffler JP (2011)
disease. FEBS J 275(23):5767–5773 Energy metabolism in amyotrophic lateral sclerosis.
28. Car H, Zendzian-Piotrowska M, Fiedorowicz A,
Lancet Neurol 10(1):75–82
Prokopiuk S, Sadowska A, Kurek K (2012) The role 41. Ngo ST, Steyn FJ, Huang L, Mantovani S, Pfluger
of ceramides in selected brain pathologies: ischemia/ CM, Woodruff TM, O'Sullivan JD, Henderson RD,
hypoxia, Alzheimer disease. Postepy Hig Med Dosw McCombe PA (2015) Altered expression of metabolic
66:295–303 proteins and adipokines in patients with amyotrophic
29. Miana-Mena FJ, Piedrafita E, Gonzalez-Mingot C, lateral sclerosis. J Neurol Sci 357(1–2):22–27
Larrode P, Munoz MJ, Martinez-Ballarin E, Reiter RJ, 42. Chio A, Calvo A, Ilardi A, Cavallo E, Moglia
Osta R, Garcia JJ (2011) Levels of membrane fluidity C, Mutani R, Palmo A, Galletti R, Marinou K,
in the spinal cord and the brain in an animal model of Papetti L et al (2009) Lower serum lipid levels are
amyotrophic lateral sclerosis. J Bioenerg Biomembr related to respiratory impairment in patients with
43(2):181–186 ALS. Neurology 73(20):1681–1685
30. Ilieva EV, Ayala V, Jove M, Dalfo E, Cacabelos D, 43. Blasco H, Veyrat-Durebex C, Bocca C, Patin F,
Povedano M, Bellmunt MJ, Ferrer I, Pamplona R, Vourc'h P, Kouassi Nzoughet J, Lenaers G, Andres
Portero-Otin M (2007) Oxidative and endoplasmic CR, Simard G, Corcia P et al (2017) Lipidomics
reticulum stress interplay in sporadic amyotrophic lat- reveals cerebrospinal-fluid signatures of ALS. Sci
eral sclerosis. Brain J Neurol 130.(Pt 12:3111–3123 Rep 7(1):17652
31. Reich-Slotky R, Andrews J, Cheng B, Buchsbaum 44. Vinknes KJ, Elshorbagy AK, Drevon CA, Nurk E,
R, Levy D, Kaufmann P, Thompson JL (2013) Body Tell GS, Nygard O, Vollset SE, Refsum H (2013)
mass index (BMI) as predictor of ALSFRS-R score Associations between plasma polyunsaturated fatty
decline in ALS patients. Amyotroph Later Scler acids, plasma stearoyl-CoA desaturase indices and
Frontotemporal Degener 14(3):212–216 body fat. Obesity 21(9):E512–E519
32.
Henriques A, Croixmarie V, Priestman DA, 45. Vinknes KJ, Elshorbagy AK, Nurk E, Drevon CA,
Rosenbohm A, Dirrig-Grosch S, D'Ambra E, Gjesdal CG, Tell GS, Nygard O, Vollset SE, Refsum
Huebecker M, Hussain G, Boursier-Neyret C, H (2013) Plasma stearoyl-CoA desaturase indices:
Echaniz-Laguna A et al (2015) Amyotrophic lateral association with lifestyle, diet, and body composition.
sclerosis and denervation alter sphingolipids and up- Obesity 21(3):E294–E302
regulate glucosylceramide synthase. Hum Mol Genet 46. Falcao-Pires I, Castro-Chaves P, Miranda-Silva D,
24(25):7390–7405 Lourenco AP, Leite-Moreira AF (2012) Physiological,
33. Dupuis L, Corcia P, Fergani A, Gonzalez De Aguilar pathological and potential therapeutic roles of adipo-
JL, Bonnefont-Rousselot D, Bittar R, Seilhean D, kines. Drug Discov Today 17(15–16):880–889
Hauw JJ, Lacomblez L, Loeffler JP et al (2008) 47. Spector AA (1999) Essentiality of fatty acids. Lipids
Dyslipidemia is a protective factor in amyotrophic 34(Suppl):1–3
lateral sclerosis. Neurology 70(13):1004–1009 48. Nordstrom V, Willershauser M, Herzer S, Rozman J,
34. Schmitt F, Hussain G, Dupuis L, Loeffler JP,
von Bohlen Und Halbach O, Meldner S, Rothermel U,
Henriques A (2014) A plural role for lipids in motor Kaden S, Roth FC, Waldeck C et al (2013) Neuronal
neuron diseases: energy, signaling and structure. expression of glucosylceramide synthase in central
Front Cell Neurosci 8(25) nervous system regulates body weight and energy
35. Yip PK, Pizzasegola C, Gladman S, Biggio ML,
homeostasis. PLoS Biol 11(3):e1001506
Marino M, Jayasinghe M, Ullah F, Dyall SC,
Malaspina A, Bendotti C et al (2013) The omega-3
18 Lipids in ALS 241
49. Inokuchi J (2009) Neurotrophic and neuroprotective 52. Pompl PN, Ho L, Bianchi M, McManus T, Qin
actions of an enhancer of ganglioside biosynthesis. Int W, Pasinetti GM (2003) A therapeutic role for
Rev Neurobiol 85:319–336 cyclooxygenase-2 inhibitors in a transgenic mouse
50. Trostchansky A, Mastrogiovanni M, Miquel E,
model of amyotrophic lateral sclerosis. FASEB
Rodriguez-Bottero S, Martinez-Palma L, Cassina P, J 17(6):725–727
Rubbo H (2018) Profile of arachidonic acid-derived 53. Klivenyi P, Kiaei M, Gardian G, Calingasan NY,
inflammatory markers and its modulation by nitro- Beal MF (2004) Additive neuroprotective effects of
oleic acid in an inherited model of amyotrophic lateral creatine and cyclooxygenase 2 inhibitors in a trans-
sclerosis. Front Mol Neurosci 11(131) genic mouse model of amyotrophic lateral sclerosis.
51. Ilzecka J (2003) Prostaglandin E2 is increased in
J Neurochem 88(3):576–582
amyotrophic lateral sclerosis patients. Acta Neurol
Scand 108(2):125–129
Flavonoids Ability to Disrupt
Inflammation Mediated by Lipid 19
and Cholesterol Oxidation
Carlo Barnaba and Ilce G. Medina-Meza
strictly mechanistic aspects: inhibition of ize its bioavailability [46]. In terms of flavonoids,
chemical-induced LDL oxidation, and the effect the primary route of administration is ingestion.
of flavonoids in the monocyte/macrophages acti- Once ingested, flavonoids undergo sulfation,
vation pathways. methylation and glucuronidation in the small
intestine and liver by respective enzymes [26,
44]. These metabolic forms are the ones com-
19.2 Flavonoid Absorption monly found in plasma, rather than the aglycone
and Bioavailability [10, 38, 44, 46, 64]. Several studies performed in
humans have shown that after ingestion of flavo-
The average US daily flavonoid intake is noid-rich food, maximum plasma levels are
189.7 mg/d, subdivided in flavan-3-ols (87%), observed after 1.5–2 h; however, only sub- or
flavanones (7.6%), flavonols (6.8%), and others low-micromolar concentration of the correspond-
[12]. Quercetin is the most important contributor ing aglycone are retrieved. This is the case of
to the estimated intake of flavonoids, mainly green tea catechins (0.6–1.8 μM, depending on
from the consumption of apples and onions [52]. the considered compound) [74], quercetin from a
Data regarding flavonoids bioavailability – i.e. vegetable and fruit-enriched diet (0.14 μM) [17],
the portion of the compound ingested that is as well as from ingestion of apples and onions
digested, absorbed and metabolized – are still (1.5–3.4 μM) [33], among others. From
incomplete. Plasma or urine concentration of a these studies, it is clear that only a small percent-
certain compound is typically used to character- age of flavonoids (<2%) of the initial intake is
19 Flavonoids Ability to Disrupt Inflammation Mediated by Lipid and Cholesterol Oxidation 245
Table 19.1 Individual flavonoids according to their subclass, as found in common berries
Flavonoid subclass Compound Berry species
Flavonols Kaempferol Blueberry, sour cherry, cranberry, raspberry, strawberry
Quercetin Blueberry, sour cherry, cranberry, raspberry, strawberry
Myricetin Blueberry, cranberry, strawberry
Isoharmnetin Sour cherry
Flavones Apigenin –
Luteolin –
Flavanones Hesperitin –
Narigenin Strawberry
Flavan-3-ols (-)-Epicatechin Blueberry, sour cherry, cranberry, raspberry, strawberry
(-)-Epicatechin 3-gallate Strawberry
(-)-epigallocatechin Cranberry, raspberry, strawberry
(-)-epigallocatechin 3-gallate Cranberry, raspberry, strawberry
(+)-catechin Blueberry, sour cherry, cranberry, raspberry, strawberry
(+)-gallocatechin Strawberry
Proanthocyanidins Monomer Blueberry, cranberry, raspberry
Dimer Blueberry, cranberry, raspberry
Trimer Blueberry, cranberry, raspberry
4-6mers Blueberry, cranberry, raspberry
7-10mers Blueberry, cranberry, raspberry
Polymers Blueberry, cranberry,
Anthocyanidins Cyanidin Blueberry, sour cherry, cranberry, raspberry, strawberry
Delphinidin Blueberry, cranberry, raspberry, strawberry
Malvidin Blueberry, cranberry, raspberry, strawberry
Pelargonidin Blueberry, cranberry, raspberry, strawberry
Peonidin Blueberry, sour cherry, cranberry, raspberry, strawberry
Petunidin Raspberry, strawberry
absorbed. Compounds that are not absorbed from actually decrease as the dose increases, suggest-
the intestine migrate to the colon wherein they ing that there is an absorption saturation point
interact with the microflora. The flavonoids that [10]. Because of the overall poor absorption and
re-enter circulation can now be broken into low low bioavailability of flavonoids, a variety of
aglycones by the microbiota and further into low strategies have been explored in order to improve
molecular weight compounds that can be these inefficiencies. Increasing metabolic stabil-
absorbed [64]. ity, intestinal absorption and changing the absorp-
Different polyphenols have different absorp- tion site have been considered to improve
tion rates and metabolic conversions due to their flavonoid bioavailability [52, 64].
variable structures. Regarding flavonols, studies
performed in the Caco-2 model cell line have
shown that the several glycosylated forms are 19.3 Lipid and Cholesterol
poorly absorbed [15, 26]. Aglycones can pass Oxidation in LDL
through the gut wall because microflora hydro- and Atherosclerosis
lyzes the sugar moiety [25]. However, there is
also a compound specificity in the absorption 19.3.1 Chemical-Induced Oxidation
mechanism, i.e. quercetin and daidzein gluco- of LDL
sides seemed to be directly absorbed by active
transport [26]. Not only does polyphenol struc- There is a generous literature regarding ex vivo or
ture affect the absorption, but so can the dose and in vitro oxidation of LDL, and it is out of the
food matrix in which they are contained. It has scope of this chapter a detailed revision of the
been found that the absorption efficiency can field, which can be found elsewhere [42]. Here,
246 C. Barnaba and I. G. Medina-Meza
we will summarize the main strategies used to ity [42]. LDL can be mildly or highly oxidized
assess LDL oxidation and how these experimen- (MM-LDL and the proper ox-LDL, respec-
tal setups are relevant for flavonoids- centered tively), although there is no general consensus
antioxidant studies. LDL are complex macromol- about the active species present in each stage
ecules, made of a lipid core surrounded by a sin- [11, 42, 50].
gle non-exchangeable lipoprotein, known as
apolipoprotein (ApoB). The average lipid core
has been calculated to contain 600 molecules of 19.3.2 Cholesterol Oxidation in LDL
free cholesterol, 1600 molecules of cholesteryl
esters (mainly arachidonate and linoleate esters), Cholesterol is a natural occurring compound that
700 molecules of phospholipids (64% PC, 1.5% has several crucial functions within the body. It
PE, 26% SM, and 11% LPC), 180 molecules of is a key component of all cellular membranes, a
triacylglycerides (TAGs), and approximately 10 precursor to steroid hormones and is involved in
molecules of the lipophilic antioxidant many signal transduction pathways [27].
α-tocopherol [37, 42]. Several exchangeable Cholesterol transport in humans is a complex
lipoproteins are also associated to LDL particle, process. LDL are the lipoproteins that carry
and their function is to modulate interactions esterified cholesterol from the liver to the periph-
with specific cell receptors [7, 16]. In particular, eral tissue [71]. In humans, a balance of choles-
high concentrations of ApoC-III in ApoB lipo- terol is critical for many physiological
proteins have been reported among patients with processing, since accumulation of this molecules
coronary disease and coronary atherosclerotic in cell membrane can affect the function of a
lesions, as well as metabolic syndrome and type variety of enzymes, transporters and receptors.
2 diabetes [16, 61]. Cholesterol is susceptible of being oxidized by
Ex vivo and in vitro oxidation of LDL is an naturally occurring oxidative agents, like reac-
experimental strategy largely adopted to either tive oxygen and nitrogen species (ROS and RNS,
characterize products of lipid oxidation, or respectively), that can alter its chemical back-
assess the ability of antioxidants to dampen bone by oxygenating carbons at several posi-
oxidation itself. Oxidation can be triggered via tions. Commonly, auto-oxidation of cholesterol
several mechanisms, including but not limited (i.e. by chemical and physical means) occurs at
to free radicals generation, Fenton reaction, and position C-7 proximal to the C5-C6 double bond,
peroxynitrite oxidation agents [19]. α,α′- or to the C-25 tertiary carbon [45, 47]. However,
azodiisobutyramidine dihydrochloride (AAPH) first oxidative derivatives can be further oxidized
has been widely used as ROS inducer, since it and go subsequent chemical rearrangements. To
degrades to highly reactive radical species that date, more than 80 compounds, broadly known
oxidized molecular oxygen to peroxy radicals as cholesterol oxidation products (COPs) have
[19]. Fenton reaction is triggered by micromo- been identified. COPs are more polar than the
lar solution of Cu2+ (or less commonly Fe2+) parent cholesterol, thus present different chemi-
salts, which are able to abstract a proton from cal and biophysical properties in cell environ-
preformed lipid peroxides to generate peroxyl ment. Their different chemistry is the major
radicals [28]. Finally, peroxynitrite is obtained source of health- associated issues, since they
from a variety of precursor, including linsido- can interfere with cholesterol homeostasis at
mine (SIN-1) [62, 65]. Oxidation of LDL can several level. It is not the scope of this chapter to
potentially affect each of its constituents, detail the putative biological effects due to the
including the apoB, and cross-reactions exposure of COPs, as from cell, in vivo and
between different fractions (i.e. protein/lipid (marginally) clinical studies. The reader can
chemical interaction) can exponentially multi- refer to our recent review [45] or other compel-
ply the chemistry of the oxidative products, to a ling works in the area. Briefly, in vitro works
variety of lipids with reported biological activ- using several cell lines have reported that admin-
19 Flavonoids Ability to Disrupt Inflammation Mediated by Lipid and Cholesterol Oxidation 247
istration of high concentration of COPs – either the biomarkers of oxidative stress: PL-esterified
individually or as a mixture of compounds – can isoprostanes are known to have pro-inflamma-
trigger several physiological responses able to tory action, and stimulate monocyte adhesion to
induce pro-inflammatory, pro- fibrogenic, and the endothelial cell [1, 2]. Oxysterols, in partic-
pro-apoptotic effects. A consistent body of liter- ular 7-ketocholesterol, 7α-OH and 7β-OH cho-
ature has also assessed the cytotoxic, mutagenic lesterol, and cholesterol epoxides are abundant
and carcinogenic properties of COPs. As afore- in LDL, and are believed to exert cytotoxic
mentioned, cholesterol homeostasis is strictly effects on monocyte-macrophages ([5, 13]).
linked to COPs accumulation in fluids and cell Although toxic, ROS-induced COPs (particu-
compartments [45]. larly B-ring derivatives) seem unable to activate
liver-X receptors (LXR-α and β) and
peroxisome-proliferator-activated (PPARs)
19.3.3 OxLDL Activate Monocytes/ receptors in macrophages, and so trigger the
Macrophages Pathway anti-atherogenic response [6, 37, 69]. On the
other side, enzymatic side-chain COPs like
The mechanism by which LDL activates mono- 22-OH, 25-OH and 27-OH cholesterols can trig-
cytes in the intima towards their conversion to ger both LXRα and PPARs [69], which is
macrophages is still controversial, although a explained by specific structural requirements
rough phenomenological description has been needed for these ligand-receptor interactions to
provided [59]. There is scientific consensus that occur [36]. Other inflammatory mechanisms
oxLDLs, rather than the parent LDL, are the pri- have been proposed for B-ring oxysterols, as
mary stimulus leading endothelial activation 7-keto and 7-OH cholesterol isomers. A increas-
and subsequent inflammatory response. Under ing literature body has demonstrated that B-ring
particular conditions, LDL can cross the endo- oxysterols can stimulate inflammatory signaling
thelial tissue of the blood vessels and being in monocytes by interacting with Toll-like
incorporated into the intima. According to the receptors [18], particularly TLR-4 [34]. 7β-OH
current response-to-injury hypothesis, oxidation cholesterol stimulate the increase of IL-8 secre-
of LDLs favors the expression of cell adhesion tion via the MEK/ERK1/2 pathway [69].
molecules by endothelial cells, such as the cell Oxidation of the fatty acid moiety of
adhesion molecule-1 (ICAM-1) and the vascu- cholesteryl-esters also forms compounds – like
lar adhesion molecule-1 (VCAM-1) [59]. These 9-oxononanoyl cholesterol – able to activate
cell adhesion molecules favor LDL migration adhesion of monocytes [5, 11]. These oxidative
towards the intima. ROS and RNS-induced CE (OxCE) are also able to trigger inflammation
action on LDLs can also occur at the intima through several mechanisms, including TLR-
level [23, 40]. oxLDL-induced activation of mediated response, as recently discussed in detail
macrophages is the second stage of atheroscle- by Miller and Shyy [50].
rosis development. Both chemical and enzy- The discussed inflammatory responses trigger
matic oxidative processes act on the whole LDL the differentiation of monocytes, as well as their
chemical structure, including phospholipids, recruitment from the vascular vessels to the arte-
cholesterol (free and esterified), apolipoprotein rial wall, via trans-endothelial migration [43, 59].
and TAGs [11]. Lipids species derived from oxi- The monocyte recruitment molecular process is
dation are responsible to “activate” monocytes, still under debate, and involve several signaling
via specific interaction with membrane recep- mechanisms, as discussed elsewhere [43]. In the
tors. Extensive oxidation is necessary for LDL intima, monocytes then differentiate into macro-
to bind macrophages receptors SR-A or CD36, phages, that uptake oxLDL via scavenging recep-
including up to 50% cholesterol oxidation, apoB tors, resulting in the transformation of
fragmentation, and diffused oxidation of unsat- macrophages to foam cells. Accumulation of
urated fatty acids [6, 37]. Isoprostane are one of foam cells and platelets is the boundary stone for
248 C. Barnaba and I. G. Medina-Meza
the formation of the atherosclerotic lesions showed that the contribution of the different
known as fatty streaks. functional groups to the inhibition of LDL oxi-
dation is well correlated to the energy needed to
form the flavonoid radical by abstraction of a
19.4 Flavonoids Against LDL hydrogen atom. In other words, it is possible to
Oxidation classify flavonols according to their inhibition
potential towards radical and copper induced
19.4.1 Mechanisms of Flavonoids oxidation, being more effective those com-
Action Against Lipid pounds having two adjacent hydroxyls at the
Oxidation B-ring, like quercetin and catechin. Solubility
and partitioning behavior of flavonoids could
Several studies in recent years have examined the also play a role in defining their scavenging
relation between flavonoids and oxidation or activity, considering that their effectiveness as
inflammation. The powerful antioxidant activity LDL oxidative protectors relies in their ability to
exhibited by flavonoids is due to their action as physically interact with the lipid core [48].
free radical scavengers, singlet oxygen quench- However, Paganga et al. [54] demonstrated that
ers, and metal ion chelators [26]. Several in vitro superiority of quercetin vs. catechin in inhibiting
systems have been used to evaluate the antioxi- LDL peroxidation cannot be accounted to parti-
dant potential of flavonoids. In this chapter, we tioning considerations, but to the exceptional
will focus in LDL oxidation and macrophages ability of the former to chelate copper. Another
activation. interesting SAR study was performed by Yi and
LDL oxidation, as aforementioned, is the first coworkers [75], where twenty-three 4-oxo-flavo-
critical step in triggering the inflammatory cas- noids (i.e. C-4 is oxidized to ketone function)
cade that finally brings to atheroma formation were assessed against oxLDL-induced endothe-
and consequent CVD manifestation. Early works lial dysfunction. Similarly to the previous stud-
by Frankel’s group [20, 48] have extensively ies [48, 49, 68], flavonols – morin, myricetin,
explored the ability of fruits flavonoids extracts fisetin and quercetin – showed higher activity in
to dampen oxidation in LDL. Flavonoids possess micromolar range. The promising “hits” shared
lower redox potential than lipids, and are thus higher oxy-functionalization at the flavan ring:
oxidized by ROS and RNS, resulting in more 3′,4′-o-dihydroxyl on B-ring, a 3-hydroxyl on
stable and less-reactive ortho-semiquinone radi- C-ring, a 2,3-double bond and a 5,7-m-dihy-
cal compared to lipid radicals and peroxy radi- droxyl on the A-ring were all required for the
cals [3, 4, 29]. When evaluated individually, observed inhibitory effect.
several pure flavonoids (catechin, quercetin,
cyanidin, caffeic acid, and ellagic acid) showed
up to 95% inhibition of Cu2+-induced hexanal 19.4.2 Dietary Flavonoids and LDL
production in micromolar ranges (2.5–7.5 μM). Oxidation
However, specific binary and ternary mixtures of
flavonoids showed antagonistic effects, particu- A substantial amount of literature has used
larly if caffeic and ellagic acids coexisted in grape and wine as flavonoids-enriched matrix to
equal amounts [48]. Dissimilarities among flavo- leverage their LDL-protectant function, mainly
noids AO activity can be explained taking into supported by early hypothesis regarding the so-
account differences in their chemical structures; called “French paradox” formulated by epide-
particularly, structural variation in the C-ring miologists in the early ‘80. The “French
and the oxidative state of C3 seemed to confer paradox” is the observation of low coronary
different reactivity [29, 48]. A structure-activity heart disease (CHD) death rates despite high
relationship (SAR) study by Vaya et al. [68] intake of dietary cholesterol and saturated fat in
19 Flavonoids Ability to Disrupt Inflammation Mediated by Lipid and Cholesterol Oxidation 249
southern French diet [58]. Red wine contains up intrinsic antioxidants are still present. Similar
to 250 mg/mL phenols, depending on type and results have been obtained with flavonoids
variety [44]. Experiments performed using Petit extract from different species of berries [30,
Syrah wine showed that 5 μM concentration of 51], as well as other fruits [44].
individual catechin, myricetin, epicatechin, An obvious following step in CVD-related fla-
rutin and quercetin were able to inhibit LDL vonoids research are in vivo and clinical studies.
oxidation by Cu2+ in the range 60–75% [63], Various studies have exposed either animal mod-
higher than α-tocopherol. More interesting, pro- els or humans subjects to flavonoids-enriched
cyanidins (i.e. oligomeric forms of flavonols) diets, recording controversial results that par-
were higher inhibitors of hexanal formation (up tially debunked the “anti-oxidative claim” [44].
to 80% at 5 μM), which can be explain by their Part of this controversy is due to the lack of reli-
better ability to interact with the LDL constitu- able biomarkers for measuring plasma antioxi-
ents. A more extensive work performed on 14 dant activity, lack of long-term studies and
grapes varieties demonstrated similar effects on underestimation of gut and liver bio-
the inhibition of hexanal generation, with the transformation [44, 73]. A few examples of in
additional information that the inhibitory poten- vivo works follow, and a good critical review can
tial was positively correlated with the content of be found elsewhere [44].
phenols, and in less extent anthocyanins and fla- O’Reilly and co-workers coupled in vitro
vonols [49]. An interesting paper by Frémont assessment of a flavonoid mixture against Cu2+
and co-workers more deeply investigated the oxidation of LDL with dietary intervention
effects of wine extract and individual flavonoids study. All the tested flavonoids – including quer-
in in vitro LDL oxidation by copper and AAPH cetin, luteolin, catechin, and kaempferol, among
[21]. Their results discriminated between che- others – showed sub-micromolar ability to halve
lating agents (resveratrol), which are active TBARS value and decrease LDL peroxides.
against Cu2+ oxidation, and radical scavengers Surprisingly, when 32 subjects were exposed to
(catechin) that inhibit AAPH lipid peroxide for- a high-flavonoids diet, no significant effects
mation. Furthermore, in their studies highly were found in ex vivo LDL oxidation [53]. From
unsaturated cholesteryl-esters, rather than phos- the in vitro effects on LDL, Pignatelli and others
pholipids, were mostly affected by both oxida- [56] administered red and white wine to 20
tion mechanisms, and this was demonstrated by healthy volunteers, finding a significant reduc-
altered electrophoresis mobility of apoB after tion in plasma conjugate dienes, as well as uri-
oxidation. These effects were negligible in nary PGF-2α-III, a marker of oxidative stress.
white wines, which contain lower amounts of Flavonols from concentrated cranberry juice
flavonoids, although imposing a longer grape- decreased plasma oxLDL and cell adhesion mol-
skin contact during winemaking can led to “red- ecules ICAM-1 and VCAM-1 in 30 men, over
like” white wines with analogue antioxidant three successive periods of 4 weeks [60]. In their
capacity [22]. Beside grape, other flavonol-rich review, Lotito and collaborators [44] list a few
dietary extracts have been tested as antioxidants works that failed to demonstrate in vivo efficacy
against LDL chemically-induced oxidation. of flavonoids in inhibit plasma oxidation. The
Viana et al. [70] used an extract of Vaccinium conclusion is that, although epidemiology stud-
myrtillus containing anthocyan, catechin, chal- ies are fairly convincing about their antioxidant
cone and other flavonoids to dampen Cu2+- effects, is unlikely that flavonoids per se acts
induced LDL oxidation. They showed a against LDL oxidation, given their poor bio-
significant increase of the lag phase of lipid per- availability. Likely, observed effects are due to
oxidation associated with the presence of vita- upregulation of endogenous antioxidants
min E, indicating that the effects of flavonoids enzymes, like those of the uric acid biosynthetic
are limited to the early stages of oxidation, when pathway.
250 C. Barnaba and I. G. Medina-Meza
flavonoids-conjugates, with unknown or reduced 8. Chan MM-Y, Fong D, Ho C-T, Huang H-I (1997)
Inhibition of inducible nitric oxide synthase gene
biological activity. In order to positively merge expression and enzyme activity by epigallocatechin
the in vitro observations with the diffused epide- gallate, a natural product from green tea. Biochem
miological studies, research should be focused in Pharmacol 54(12):1281–1286
validate those speculated mechanisms in more 9. Chang YP, Ka SM, Hsu WH, Chen A, Chao LK, Lin
CC, Hsieh CC, Chen MC, Chiu HW, Ho CL (2015)
physiological settings. Among various aspects Resveratrol inhibits NLRP3 inflammasome activation
worth of research interests, the authors believe by preserving mitochondrial integrity and augmenting
that priority should be given to a deep under- autophagy. J Cell Physiol 230(7):1567–1579
standing of the “fate” of flavonoids in our organ- 10. Charron CS, Kurilich AC, Clevidence BA, Simon PW,
Harrison DJ, Britz SJ, Baer DJ, Novotny JA (2009)
ism, a critical effort that should consider Bioavailability of anthocyanins from purple carrot
adsorption, bioavailability, and the actions of juice: effects of acylation and plant matrix. J Agric
intrinsic metabolism, inasmuch as gut microbi- Food Chem 57(4):1226–1230
ota. Also, the “flavonoids effect” should be better 11. Choi SH, Sviridov D, Miller YI (2017) Oxidized cho-
lesteryl esters and inflammation. Biochim Biophys
contextualized in the overall diet, thus facilitating Acta Mol Cell Biol Lipids 1862(4):393–397
the comprehension of combined effects between 12. Chun OK, Chung SJ, Song WO (2007) Estimated
other macro and micro-nutrients, that can poten- dietary flavonoid intake and major food sources of US
tially act as synergistic/antagonist. Thus, a holis- adults. J Nutr 137(5):1244–1252
13. Clare K, Hardwick SJ, Carpenter KL, Weeratunge
tic rather than a criterion-based approach can led N, Mitchinson MJ (1995) Toxicity of oxysterols
to more realistic outcomes and expectations on to human monocyte-macrophages. Atherosclerosis
the consumer. 118(1):67–75
14. Comalada M, Ballester I, Bailon E, Sierra S, Xaus J,
Galvez J, de Medina FS, Zarzuelo A (2006) Inhibition
Acknowledgements This work has been partially funded
of pro-inflammatory markers in primary bone marrow-
by Michigan State University start-up to I.G.M.M. Mr.
derived mouse macrophages by naturally occurring
Matthew Schweiss is acknowledged for his help in prepar-
flavonoids: analysis of the structure–activity relation-
ing Table 19.1.
ship. Biochem Pharmacol 72(8):1010–1021
15. Dai J-y, Yang J-l, Li C (2008) Transport and metab-
olism of flavonoids from Chinese herbal remedy
References Xiaochaihu-tang across human intestinal Caco-2 cell
monolayers. Acta Pharmacol Sin 29(9):1086–1093
16. Davidsson P, Hulthe J, Fagerberg B, Olsson B-M,
1. Berliner JA, Gharavi NM (2008) Endothelial cell
Hallberg C, Dahllöf B, Camejo G (2005) A proteomic
regulation by phospholipid oxidation products. Free
study of the apolipoproteins in LDL subclasses in
Radic Biol Med 45(2):119–123
patients with the metabolic syndrome and type 2 dia-
2. Berliner JA, Leitinger N, Tsimikas S (2009) The role
betes. J Lipid Res 46(9):1999–2006
of oxidized phospholipids in atherosclerosis. J Lipid
17. Erlund I (2004) Review of the flavonoids quercetin,
Res 50(Suppl):S207–S212
hesperetin, and naringenin. Dietary sources, bioac-
3. Bors W, Michel C, Stettmaier K (2001) Structure-
tivities, bioavailability, and epidemiology. Nutr Res
activity relationships governing antioxidant capaci-
24(10):851–874
ties of plant polyphenols. In: Methods in enzymology.
18.
Erridge C, Webb DJ, Spickett CM (2007)
Elsevier, Amsterdam, pp 166–180
25-hydroxycholesterol, 7β-hydroxycholesterol and
4. Bors W, Saran M (1987) Radical scavenging by
7-ketocholesterol upregulate interleukin-8 expres-
flavonoid antioxidants. Free Radic Res Commun
sion independently of toll-like receptor 1, 2, 4 or 6
2(4–6):289–294
signalling in human macrophages. Free Radic Res
5. Brown AJ, Dean RT, Jessup W (1996) Free and esteri-
41(3):260–266
fied oxysterol: formation during copper-oxidation of
19. Esterbauer H, Jiirgens G (1993) Mechanistic and
low density lipoprotein and uptake by macrophages.
genetic aspects of susceptibility of LDL to oxidation.
J Lipid Res 37(2):320–335
Curr Opin Lipidol 4(2):114–124
6. Brown AJ, Jessup W (1999) Oxysterols and athero-
20. Frankel E, German J, Kinsella J, Parks E, Kanner
sclerosis. Atherosclerosis 142(1):1–28
J (1993) Inhibition of oxidation of human low-density
7. Campos H, Perlov D, Khoo C, Sacks FM (2001)
lipoprotein by phenolic substances in red wine. Lancet
Distinct patterns of lipoproteins with apoB defined
341(8843):454–457
by presence of apoE or apoC-III in hypercholes-
21.
Frémont L, Belguendouz L, Delpal S (1999)
terolemia and hypertriglyceridemia. J Lipid Res
Antioxidant activity of resveratrol and alcohol-free
42(8):1239–1249
252 C. Barnaba and I. G. Medina-Meza
wine polyphenols related to LDL oxidation and poly- vonoids: genistein, kaempferol, quercetin, and daid-
unsaturated fatty acids. Life Sci 64(26):2511–2521 zein inhibit STAT-1 and NF-κB activations, whereas
22. Fuhrman B, Volkova N, Suraski A, Aviram M (2001) flavone, isorhamnetin, naringenin, and pelargonidin
White wine with red wine-like properties: increased inhibit only NF-κB activation along with their inhibi-
extraction of grape skin polyphenols improves the tory effect on iNOS expression and NO production in
antioxidant capacity of the derived white wine. activated macrophages. Mediat Inflamm 2007:45673
J Agric Food Chem 49(7):3164–3168 36. Janowski BA, Grogan MJ, Jones SA, Wisely GB,
23. Förstermann U, Xia N, Li H (2017) Roles of vascular Kliewer SA, Corey EJ, Mangelsdorf DJ (1999)
oxidative stress and nitric oxide in the pathogenesis of Structural requirements of ligands for the oxysterol
atherosclerosis. Circ Res 120(4):713–735 liver X receptors LXRα and LXRβ. Proc Natl Acad
24.
García-Lafuente A, Guillamón E, Villares A, Sci 96(1):266–271
Rostagno MA, Martínez JA (2009) Flavonoids as 37. Jessup W, Kritharides L, Stocker R (2004) Lipid oxi-
anti-inflammatory agents: implications in cancer and dation in atherogenesis: an overview. Biochem Soc
cardiovascular disease. Inflamm Res 58(9):537–552 Trans 32(1):134–138
25. Georgiev V, Ananga A, Tsolova V (2014) Recent 38. Kawai Y, Nishikawa T, Shiba Y, Saito S, Murota K,
advances and uses of grape flavonoids as nutraceuti- Shibata N, Kobayashi M, Kanayama M, Uchida K,
cals. Nutrients 6(1):391–415 Terao J (2008) Macrophage as a target of querce-
26. González R, Ballester I, López-Posadas R, Suárez tin glucuronides in human atherosclerotic arteries
M, Zarzuelo A, Martinez-Augustin O, Medina FSD implication in the anti-atherosclerotic mechanism of
(2011) Effects of flavonoids and other polyphenols on dietary flavonoids. J Biol Chem 283(14):9424–9434
inflammation. Crit Rev Food Sci Nutr 51(4):331–362 39. Lairon D, Amiot MJ (1999) Flavonoids in food and
27. Guizzetti M, Costa LG (2005) Disruption of choles- natural antioxidants in wine. Curr Opin Lipidol
terol homeostasis in the developing brain as a potential 10(1):23–28
mechanism contributing to the developmental neuro- 40. Leeuwenburgh C, Hardy MM, Hazen SL, Wagner
toxicity of ethanol: an hypothesis. Med Hypotheses P, Oh-ishi S, Steinbrecher UP, Heinecke JW (1997)
64(3):563–567 Reactive nitrogen intermediates promote low density
28. Gutteridge JM (1986) Iron promoters of the Fenton lipoprotein oxidation in human atherosclerotic intima.
reaction and lipid peroxidation can be released J Biol Chem 272(3):1433–1436
from haemoglobin by peroxides. FEBS Lett 41. Leiro J, Alvarez E, Arranz JA, Laguna R, Uriarte E,
201(2):291–295 Orallo F (2004) Effects of cis-resveratrol on inflam-
29. Heim KE, Tagliaferro AR, Bobilya DJ (2002)
matory murine macrophages: antioxidant activity and
Flavonoid antioxidants: chemistry, metabolism and down-regulation of inflammatory genes. J Leukoc
structure-activity relationships. J Nutr Biochem Biol 75(6):1156–1165
13(10):572–584 42. Levitan I, Volkov S, Subbaiah PV (2010) Oxidized
30. Heinonen IM, Meyer AS, Frankel EN (1998)
LDL: diversity, patterns of recognition, and patho-
Antioxidant activity of berry phenolics on human physiology. Antioxid Redox Signal 13(1):39–75
low-density lipoprotein and liposome oxidation. 43. Ley K, Miller YI, Hedrick CC (2011) Monocyte
J Agric Food Chem 46(10):4107–4112 and macrophage dynamics during atherogenesis.
31. Hertog MG, Feskens EJ, Kromhout D, Hollman P, Arterioscler Thromb Vasc Biol 31(7):1506–1516
Katan M (1993) Dietary antioxidant flavonoids and 44. Lotito SB, Frei B (2006) Consumption of flavonoid-
risk of coronary heart disease: the Zutphen Elderly rich foods and increased plasma antioxidant capacity
Study. Lancet 342(8878):1007–1011 in humans: cause, consequence, or epiphenomenon?
32. Hertog MG, Kromhout D, Aravanis C, Blackburn
Free Radic Biol Med 41(12):1727–1746
H, Buzina R, Fidanza F, Giampaoli S, Jansen A, 45. Maldonado-Pereira L, Schweiss M, Barnaba C,
Menotti A, Nedeljkovic S (1995) Flavonoid intake Medina-Meza IG (2018) The role of cholesterol oxi-
and long-term risk of coronary heart disease and dation products in food toxicity. Food Chem Toxicol
cancer in the seven countries study. Arch Intern Med 118:908–939
155(4):381–386 46. McGhie TK, Walton MC (2007) The bioavailabil-
33. Hollman PC, Van Trijp JM, Buysman MN, vd Gaag ity and absorption of anthocyanins: towards a better
MS, Mengelers MJ, De Vries JH, Katan MB (1997) understanding. Mol Nutr Food Res 51(6):702–713
Relative bioavailability of the antioxidant flavonoid 47. Medina-Meza IG, Barnaba C (2013) Kinetics of cho-
quercetin from various foods in man. FEBS Lett lesterol oxidation in model systems and foods: current
418(1–2):152–156 status. Food Eng Rev 5(3):171–184
34. Huang J-D, Amaral J, Lee JW, Rodriguez IR (2014) 48.
Meyer AS, Heinonen M, Frankel EN (1998)
7-Ketocholesterol-induced inflammation signals Antioxidant interactions of catechin, cyanidin, caffeic
mostly through the TLR4 receptor both in vitro and acid, quercetin, and ellagic acid on human LDL oxi-
in vivo. PLoS One 9(7):e100985 dation. Food Chem 61(1–2):71–75
35. Hämäläinen M, Nieminen R, Vuorela P, Heinonen M, 49. Meyer AS, Yi O-S, Pearson DA, Waterhouse AL,
Moilanen E (2007) Anti-inflammatory effects of fla- Frankel EN (1997) Inhibition of human low-density
19 Flavonoids Ability to Disrupt Inflammation Mediated by Lipid and Cholesterol Oxidation 253
lipoprotein oxidation in relation to composition Cu 2+, AAPH or SIN-1 and at decreasing superoxide
of phenolic antioxidants in grapes (Vitis vinifera). THP-1 cell production. A comparison to other extracts
J Agric Food Chem 45(5):1638–1643 or compounds. Free Radic Res 37(5):573–584
50. Miller YI, Shyy JY-J (2017) Context-dependent role 63. Teissedre PL, Frankel EN, Waterhouse AL, Peleg H,
of oxidized lipids and lipoproteins in inflammation. German JB (1996) Inhibition ofIn vitrohuman LDL
Trends Endocrinol Metab 28(2):143–152 oxidation by phenolic antioxidants from grapes and
51. Miranda-Rottmann S, Aspillaga AA, Pérez DD,
wines. J Sci Food Agric 70(1):55–61
Vasquez L, Martinez AL, Leighton F (2002) Juice 64. Thilakarathna SH, Rupasinghe HP (2013) Flavonoid
and phenolic fractions of the berry Aristotelia chilen- bioavailability and attempts for bioavailability
sis inhibit LDL oxidation in vitro and protect human enhancement. Nutrients 5(9):3367–3387
endothelial cells against oxidative stress. J Agric Food 65. Thomas SR, Davies MJ, Stocker R (1998) Oxidation
Chem 50(26):7542–7547 and antioxidation of human low-density lipoprotein
52. Nijveldt RJ, Van Nood E, Van Hoorn DE, Boelens PG, and plasma exposed to 3- morpholinosydnonimine
Van Norren K, Van Leeuwen PA (2001) Flavonoids: a and reagent peroxynitrite. Chem Res Toxicol
review of probable mechanisms of action and poten- 11(5):484–494
tial applications. Am J Clin Nutr 74(4):418–425 66. Tsai SH, Lin-Shiau SY, Lin JK (1999) Suppression of
53. O'Reilly JD, Sanders TA, Wiseman H (2000)
nitric oxide synthase and the down-regulation of the
Flavonoids protect against oxidative damage to LDL activation of NFκB in macrophages by resveratrol. Br
in vitro: use in selection of a flavonoid rich diet and J Pharmacol 126(3):673–680
relevance to LDL oxidation resistance ex vivo? Free 67. Tsao R (2010) Chemistry and biochemistry of dietary
Radic Res 33(4):419–426 polyphenols. Nutrients 2(12):1231–1246
54. Paganga G, Al-Hashim H, Khodr H, Scott BC,
68. Vaya J, Mahmood S, Goldblum A, Aviram M, Volkova
Aruoma OI, Hider RC, Halliwell B, Rice-Evans CA N, Shaalan A, Musa R, Tamir S (2003) Inhibition
(1996) Mechanisms of antioxidant activities of quer- of LDL oxidation by flavonoids in relation to their
cetin and catechin. Redox Rep 2(6):359–364 structure and calculated enthalpy. Phytochemistry
55. Pan W, Yu H, Huang S, Zhu P (2016) Resveratrol 62(1):89–99
protects against TNF-α-induced injury in human 69. Vejux A, Lizard G (2009) Cytotoxic effects of oxy-
umbilical endothelial cells through promoting sterols associated with human diseases: induction of
sirtuin-1-induced repression of NF-KB and p38 cell death (apoptosis and/or oncosis), oxidative and
MAPK. PLoS One 11(1):e0147034 inflammatory activities, and phospholipidosis. Mol
56. Pignatelli P, Ghiselli A, Buchetti B, Carnevale R,
Asp Med 30(3):153–170
Natella F, Germano G, Fimognari F, Di Santo S, Lenti 70. Viana M, Barbas C, Bonet B, Bonet MV, Castro
L, Violi F (2006) Polyphenols synergistically inhibit M, Fraile MV, Herrera E (1996) In vitro effects
oxidative stress in subjects given red and white wine. of a flavonoid- rich extract on LDL oxidation.
Atherosclerosis 188(1):77–83 Atherosclerosis 123(1–2):83–91
57. Price NL, Gomes AP, Ling AJ, Duarte FV, Martin- 71. Von Eckardstein A, Nofer J-R, Assmann G (2001)
Montalvo A, North BJ, Agarwal B, Ye L, Ramadori High density lipoproteins and arteriosclerosis: role of
G, Teodoro JS (2012) SIRT1 is required for AMPK cholesterol efflux and reverse cholesterol transport.
activation and the beneficial effects of resveratrol on Arterioscler Thromb Vasc Biol 21(1):13–27
mitochondrial function. Cell Metab 15(5):675–690 72. Wadsworth TL, Koop DR (1999) Effects of the wine
58. Renaud S d, de Lorgeril M (1992) Wine, alcohol, polyphenolics quercetin and resveratrol on pro-
platelets, and the French paradox for coronary heart inflammatory cytokine expression in RAW 264.7
disease. Lancet 339(8808):1523–1526 macrophages. Biochem Pharmacol 57(8):941–949
59. Ross R (1999) Atherosclerosis—an inflammatory dis- 73. Williamson G, Manach C (2005) Bioavailability and
ease. N Engl J Med 340(2):115–126 bioefficacy of polyphenols in humans. II. Review
60. Ruel G, Pomerleau S, Couture P, Lemieux S,
of 93 intervention studies. Am J Clin Nutr 81(1):
Lamarche B, Couillard C (2008) Low-calorie cran- 243S–255S
berry juice supplementation reduces plasma oxidized 74. Wiseman S, Mulder T, Rietveld A (2001) Tea fla-
LDL and cell adhesion molecule concentrations in vonoids: bioavailability in vivo and effects on cell
men. Br J Nutr 99(2):352–359 signaling pathways in vitro. Antioxid Redox Signal
61. Sacks FM (2015) The crucial roles of apolipoproteins 3(6):1009–1021
E and C-III in apoB lipoprotein metabolism in nor- 75. Yi L, Jin X, Chen C-Y, Fu Y-J, Zhang T, Chang
molipidemia and hypertriglyceridemia. Curr Opin H, Zhou Y, Zhu J-D, Zhang Q-Y, Mi M-T (2011)
Lipidol 26(1):56–63 Chemical structures of 4-oxo-flavonoids in relation to
62. Shafiee M, Carbonneau M-A, Urban N, Descomps B, inhibition of oxidized low-density lipoprotein (LDL)-
Leger CL (2003) Grape and grape seed extract capaci- induced vascular endothelial dysfunction. Int J Mol
ties at protecting LDL against oxidation generated by Sci 12(9):5471–5489
Index
F M
Flavonoids, 2, 243–251 Macrophages, 4, 16, 28, 38, 47, 67, 79, 129, 137, 150,
220, 244
Maresins (MaR), 1, 5, 17–19, 29, 31, 38,
G 51, 53–55, 66, 67
Glaucoma, 2, 5, 159 Mass spectrometry (MS), 19, 71, 82, 83, 103–105, 108,
Goblet cells (GC), 2, 14–23 120, 126, 134–139, 142, 143, 160, 195–198,
206, 208, 223
Memory, 27, 28
H Metabolomics, 101–109, 120, 236
Healing, 5–7, 9, 19, 28, 31, 48 Microglia, 29, 30, 137, 155, 156, 159, 194, 196,
Heart failure, 2, 37, 41, 46, 105 198, 203–205, 211, 212, 220, 222, 224,
Hemorrhage, 117, 125, 127–129, 157 226, 233, 234
Hemorrhagic transformation, 125, 127 Microsomal prostaglandin E synthase-1 (mPGES-1),
12-Hydroxyeicosatetraenoic acid (12-HETE), 18, 55, 56, 89–97, 137
126, 129, 238 Mild traumatic brain injury (mTBI), 194–196,
15-Hydroxyeicosatetraenoic acid (15-HETE), 56, 83, 198–212
126, 238 mPGES-1 inhibition, 93, 95, 96
Mucins, 15, 16, 20
Myocardial infarction (MI), 38–40, 46, 48, 106
I
Immune cells, 2, 14, 29, 49, 51, 52, 91, 93,
120, 135, 136, 139, 150, 151, 154, 156, N
157, 160, 170–182, 194, 205, 220, N-arachidonoyl glycine (NAGly), 195, 200–201,
224, 234, 250 203, 204, 208, 211, 212
Infection, 1, 3, 5–9, 28, 47, 49, 54, 71, 72, 150, 151, Neuroblastoma, 2, 89–97
173, 176, 177 Neurodegeneration, 29, 30, 135, 155
Inflammation, 1, 3, 17, 28, 37, 47, 66, 81, Neuroimmunology, 134–143
104, 134, 149, 172, 203, 222, Neuroinflammation, 2, 27–32, 154, 155, 157, 201, 203,
235, 243 212, 220, 222, 234–236, 238
Ischemia, 40, 52, 126, 127, 158 Neuroprotection, 29, 129, 142, 205, 212
Ischemic stroke, 2, 125–129
O
K Ocular surface, 2, 3, 7–9, 13–23, 160
Keratitis, 1, 3–9 Omega-3 fatty acids, 54, 204, 205
Oxidative stress, 39, 40, 54, 157, 158, 177, 182,
205, 234–237, 243, 247, 249, 250
L Oxysterols, 247
Lactosylceramide (LacCer), 151, 154, 160,
175, 176, 238
Leukocytes, 3, 5, 7, 9, 19, 37, 40, 41, 47–49, P
51, 52, 54, 57, 58, 66–69, 71, 73, Phospholipases A2 (PLA2), 2, 21–23, 136, 139, 142
150, 154, 157, 159, 180, 181, Platelet-activating factor (PAF), 139
206, 222 Polyunsaturated fatty acids (PUFA), 4, 29, 38, 41, 51, 55,
Leukotriene B4 (LTB4), 6, 38, 53, 54, 73, 119, 138 105–107, 115, 116, 118, 126, 135,
Lipid biomarkers, 233–239 205, 221, 222, 225, 235
Lipid mediators, 1, 3–9, 28, 29, 38, 39, 50, 52, 54, 57, Pro-resolving mediators, 2, 7–9, 49, 51, 54–56, 58,
67, 69, 73, 86, 89, 105, 108, 139, 65–74, 205
142, 143, 228, 235 Prostaglandin E2 (PGE2), 2, 4, 20, 38, 79, 80, 89–91,
Lipid oxidation, 2, 246, 248, 250 93–95, 97, 107, 108, 119, 154, 235, 238
Lipidomics, 55, 103–105, 107, 137, 177, 195, 198, Prostaglandin glycerol ester, 77–86
211, 223–226, 235, 236 Prostaglandins (PGs), 2, 4, 19, 38, 56, 66, 77–86,
Lipoamines, 194, 195, 198, 201, 203, 208–211 107, 126, 150, 195, 224, 235
Lipoxins (LX), 1, 4, 5, 17–19, 29, 30, 38, 40, Psychiatric disorders, 101–109
51, 53, 54, 56, 57, 66, 107, 138
Lipoxygenase (LOX), 1, 2, 4, 6, 18, 38, 49,
52, 53, 55, 57, 119, 125–129, 138, 150, R
205, 222, 238 Resolution, 4, 28, 38, 47, 67, 83, 105, 224
Low-density lipoprotein (LDL), 236, 244–248 Resolution of inflammation, 1, 4, 6, 28, 31, 39,
Lysophosphatidic acid (LPA), 139–142 47–57, 67, 107
Index 257