Antimicrobial Peptides Role in Human Health and Disease 2016 Harder
Antimicrobial Peptides Role in Human Health and Disease 2016 Harder
Antimicrobial Peptides Role in Human Health and Disease 2016 Harder
Jürgen Harder
Jens-M. Schröder
Editors
Antimicrobial
Peptides
Role in Human Health and Disease
Jürgen Harder • Jens-M. Schröder
Editors
Antimicrobial Peptides
Role in Human Health and Disease
Birkhäuser Advances in Infectious Diseases
Series editors
Stefan H. Kaufmann
Max Planck Institute for Infection Biology, Berlin, Germany
Andrew A. Mercer
Department of Microbiology and Immunology, University of Otago
Dunedin, New Zealand
Olaf Weber
Bonn, Germany
v
vi Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Chapter 1
Antimicrobial Peptides in Cutaneous
Wound Healing
Ole E. Sørensen
Abstract Injury that breached the physical skin barrier increases the likelihood of
infection. The wound healing process is divided into hemostasis, inflammation, pro-
liferation, and tissue remodeling. Antimicrobial peptides play a major role for the
antimicrobial defense at all these stages in wound healing, but the main sources of
antimicrobial peptides vary with the different stages of wound healing coming from
plasma proteins, neutrophils, and keratinocytes. Apart from being part of the antimi-
crobial defense, antimicrobial peptides play other important roles in wound healing
as in angiogenesis, attraction of leukocytes, resolution of inflammation, and prolif-
eration. Future studies will demonstrate whether antimicrobial peptides can be used
therapeutically to improve the wound healing processes and reduce scar formation
in chronic wounds.
1.1 Introduction
The intact skin constitutes a very efficient physical barrier toward surrounding
microbes. Indeed, skin infections are rarely found in intact healthy skin. However,
injury or wounding causes breach in the physical barrier of the skin increasing the
likelihood of infections. Keeping the wound free of overt infection is a prerogative
for successful wound healing (Edwards and Harding 2004). Indeed, chronic non-
healing cutaneous wounds are most often infected with Staphylococcus aureus or
Pseudomonas aeruginosa (Edwards and Harding 2004). Antimicrobial peptides
(AMPs) play a major role for the antimicrobial defense during wound healing.
Indeed, studies of the remarkable ability of the African frog Xenopus laevis to keep
its wounds free of infections under non-sterile conditions led to the first identifica-
tion of antimicrobial peptides (AMPs) from the skin (Zasloff 1987).
O.E. Sørensen
Nuclear Biology Laboratory, Division of Infection Medicine, BMC, B14, Department of
Clinical Sciences, Lund University, Tornavägen 10, Lund SE-221 84, Sweden
e-mail: [email protected]
Wound healing following injury is traditionally divided into four stages: (1) hemostasis,
(2) inflammation, (3) proliferation, and (4) tissue remodeling (Singer and Clark 1999).
AMPs are found at all stages of wound healing; however, the sources of AMPs are differ-
ent at each of these stages of wound healing.
1.2.1 Injury
Injury by itself leads to generation of AMPs. In frog skin, the injury-induced ner-
vous stimulation leads to release of antimicrobial peptides from skin glands
(Simmaco et al. 1998). Though similar mechanisms have not been described in
mammals, injury does lead to activation of proteases that release membrane-bound
growth factor like HB-EGF and amphiregulin that possess antimicrobial activity
(Malmsten et al. 2007). Furthermore, these growth factors play a major role as
inducers of the epidermal AMP expression at later stages in the wound healing pro-
cess (Sørensen et al. 2006, 2008; Roupé et al. 2010). Surely, tissue injury may gen-
erate additional AMPs but a systematic study of antimicrobial peptides released by
tissue injury is still to come.
1.2.2 Hemostasis
After injury, there is extravasation of plasma proteins into the wound with activation
of complement and coagulation cascades. Activation of the complement system
leads to generation of fragments of C3a with antimicrobial activity (Nordahl et al.
2004; Sonesson et al. 2007; Pasupuleti et al. 2007). The coagulation cascade is one
of the principal host defenses in insects and activation of the human coagulation
cascade leads to cleavage of several proteins like kininogen, fibrinogen, and throm-
bin involved in the coagulation cascade, which leads to generation of several anti-
microbial peptides (Frick et al. 2006; Påhlman et al. 2013; Papareddy et al. 2010).
Thrombin knockout mice have increased susceptibility to infection with
Staphylococcus aureus infection in their limited life span (Mullins et al. 2009),
which may indicate an important role of the thrombin-derived AMPs.
1.2.3 Inflammation
Borregaard and Cowland 1997; Borregaard et al. 2007) important for the microbici-
dal activity of these cells (Flannagan et al. 2009). During the inflammatory phase,
the major AMPs present in the wound will be derived from the neutrophils. The
AMPs in neutrophils are found both in neutrophil granules and cytosol (Levy 1996;
Borregaard and Cowland 1997). Neutrophils even produce an AMP like elafin after
migration to the skin (Theilgaard-Mönch et al. 2004); however, the main antimicro-
bial peptides found in neutrophils are the α-defensins (HNPs) in azurophil granules
(Ganz et al. 1985), cathelicidins in specific granules (Sørensen et al. 1997), and
calgranulins (S100A8/S100A9) in the cytosol (Hessian et al. 1993).
The main antimicrobial activity of neutrophil defensins is exerted in the neutro-
phil phagolysosome (Joiner et al. 1989), but neutrophil defensins are also secreted
to the exterior (Ganz 1987; Faurschou et al. 2002). Apart from their direct antibacte-
rial and antiviral activity (Ganz et al. 1985; Daher et al. 1986), defensins also boost
bacterial phagocytosis by macrophages (Soehnlein et al. 2008a). Neutrophil defen-
sins have other functions of importance in wound healing. Human neutrophil defen-
sins have chemotactic activity toward monocytes (Territo et al. 1989), T cells
(Chertov et al. 1996), and immature dendritic cells (Yang et al. 2000a). Furthermore,
these peptides are mitogenic for epithelial cells and fibroblasts (Murphy et al. 1993).
Neutrophil defensins do not seem to have a nonredundant function in wound heal-
ing since mice neutrophils lack defensins (Eisenhauer and Lehrer 1992).
The cathelicidins present in specific granules (or large granules in rudiments) are
stored as inactive proteins in the granules and the biological function is unleashed
following extracellular cleavage with serine proteases from azurophil granules
(Zanetti 2004). In porcine and bovine neutrophils, the cathelicidins are processed
by elastase (Panyutich et al. 1997; Scocchi et al. 1992) while proteinase 3 is respon-
sible for the processing of the human cathelicidin hCAP-18 to the antimicrobial
peptide LL-37 (Sørensen et al. 2001), a peptide with broad-spectrum antimicrobial
activity (Turner et al. 1998). The elastase-mediated processing porcine cathelicidins
has been shown to be important for clearance of bacteria from wounds (Cole et al.
2001). Cathelicidins have other important functions in wound healing, both in the
recruitment of mononuclear cells and in the tissue regeneration. It has long be rec-
ognized that neutrophils play a major role for the subsequent recruitment of mono-
cytes (Ward 1968), and it has now been recognized that LL-37 plays a role for the
recruitment of monocytes (Soehnlein et al. 2008b). Indeed, LL-37 has been shown
to be a chemotactic factor toward monocytes as well as T cells and neutrophils
(Yang et al. 2000b). Porcine and murine cathelicidins have importance for angio-
genesis (Li et al. 2000; Koczulla et al. 2003) and both the porcine PR-39 and the
human LL-37 induce expression of VEGF (Rodriguez-Martinez et al. 2008).
Furthermore, treatment of wounds with the gene delivery of the human cathelicidin
LL-37 promotes wound healing (Jacobsen et al. 2005; Steinstraesser et al. 2014;
Carretero et al. 2008). Transactivation of the epidermal growth factor (EGFR)
occurs in cutaneous wound healing (Tokumaru et al. 2000) and LL-37 has been
shown to cause EGFR transactivation (Tjabringa et al. 2003) and thereby induce
keratinocyte migration (Tokumaru et al. 2005). Furthermore, LL-37 suppresses
both keratinocyte and neutrophil apoptosis (Chamorro et al. 2009; Nagaoka et al.
2006).
4 O.E. Sørensen
As the inflammatory cells recede from the wound, the epidermal keratinocytes
become a major source for AMPs during the proliferative phase of wound healing.
While non-injured epidermis contains constitutively expressed AMPs like hBD-1
and RNase 7, the expression of many AMPs is induced during wound healing and
inflammation. Indeed, many AMPs like hBD-2, hBD-3, RNase 7, and psoriasin
were originally isolated from inflamed epidermis (Gläser et al. 2005; Harder et al.
1997, 2001; Harder and Schröder 2002). Though some epidermal expression of the
human cathelicidin hCAP-18/LL-37 may also be found in the inflammatory stage of
wound healing (Dorschner et al. 2001), the peak expression of epidermal AMPs is
found during the proliferative phase of wound healing (Roupé et al. 2010). The
AMPs with increased epidermal expression during the proliferative phase of wound
healing include hBD-2 (Schmid et al. 2001), hBD-3 (Sørensen et al. 2006), psoria-
sin (S100A7) (Lee and Eckert 2007), S100A8/S100A9 (Thorey et al. 2001),
S100A15 (Roupé et al. 2010), elafin (vanBergen et al. 1996), SLPI (Schmid et al.
2001; Wingens et al. 1998), lactoferrin (Roupé et al. 2010), midkine (Frick et al.
2011), and NGAL (Sørensen et al. 2006). Since the various defensins arise from a
common ancestral gene (Bevins et al. 1996), it is remarkable that neutrophils and
keratinocytes share many of the same AMPs and antimicrobial proteins (see
Table 1.1); however, the AMPs in keratinocytes are induced only during wound
healing and inflammation while the AMPs in neutrophils are synthesized during
normal neutrophil differentiation in the bone barrow (Borregaard et al. 2005).
However, this means that the antimicrobial proteins and peptides are present in the
wound over and extended time, though the cellular source varies.
1 Antimicrobial Peptides in Cutaneous Wound Healing 5
Table 1.1 Antimicrobial peptides and proteins present in both neutrophils and keratinocytes
AMPs in neutrophils and keratinocytes
Neutrophils Keratinocytes
Azurophil granules
α-defensins (HNP-1-4) β-defensins (hBD-1-3)
Lysozyme Lysozyme
Specific and gelatinase granules
Lactoferrin, NGAL, hCAP-18/LL-37 Lactoferrin, NGAL, hCAP-18/LL-37
Transcobalamin, SLPI, lysozyme Transcobalamin, SLPI, lysozyme
Cytosol
Calgranulins (S100A8/S100A9) Calgranulins (S100A8/S100A9)
Produced following extravasation
Elafin Elafin
Keratinocyte AMPs not present in neutrophils
Psoriasin, RNase7, S100A15
Since α- and β-defensins arise from an ancestral gene, it is remarkable how many other antimi-
crobial peptides and proteins found in keratinocytes are also found in neutrophils. See text for
references
Though many studies demonstrate how microbial products induce AMP expression
in keratinocytes mainly through TLR activation (Abtin et al. 2008; Büchau et al.
2007, 2008; Li et al. 2013; Nagy et al. 2005; Gariboldi et al. 2008; Gerstel et al.
2009; Liu et al. 2002), the possible significance of this in acute wound healing in
noninfected wounds is not clear. The bacteria-induced AMP expression may be
more important in chronic wounds with infections (Edwards and Harding 2004).
Furthermore, in this instance, the bacterial proteases could play contradictory roles,
either by induction of AMP expression through protease-activated receptors (Chung
et al. 2004) or by AMP degradation (Schmidtchen et al. 2002). Chronic wounds like
chronic venous ulcers are characterized by chronic inflammation with continuous
recruitment of inflammatory cells, and indeed neutrophil AMPs like neutrophils
defensins are found in high amounts (Lundqvist et al. 2008). The epidermal expres-
sion of hBD-2 and psoriasin is induced in chronic venous ulcers (Butmarc et al.
2004; Dressel et al. 2010), while the expression of LL-37 is decreased (Heilborn
et al. 2003). In diabetic wounds, the high glucose levels may suppress the expres-
sion of both hBD-2 (Lan et al. 2012) and hBD-3 (Lan et al. 2011) in keratinocytes.
To understand the role of AMPs in chronic wound pathology or chronic wound
infections, more detailed studies are needed to delineate the AMP expression in dif-
ferent types of chronic wounds and how this is related to wound infection or under-
lying disease such as diabetes.
AMPs provide part of the antimicrobial defense during wound healing. Topical
application of antimicrobials do not improve normal wound healing (Lipsky and
Hoey 2009), demonstrating adequate antimicrobial defenses. While the AMPs orig-
inating from the coagulation and complement cascades, the neutrophils, and epi-
dermal keratinocytes undoubtedly contribute to the antimicrobial defense in the
8 O.E. Sørensen
wound healing process, it is difficult to decipher the role of individual AMPs due to
overlapping antimicrobial activities. Apart from SLPI (Ashcroft et al. 2000), no
AMP has been demonstrated to possess nonredundant functions during wound
healing.
Though mice models provide useful insight, it is important to note that differ-
ences exist between humans and mice – also when it comes to both wound healing
and AMPs. Wound contractions are important for wound healing in rodents such as
mice, but not in humans (Davidson 1998). Neutrophils and keratinocytes are major
sources of AMPs during wound healing, but mice have far fewer neutrophils than
humans (Mestas and Hughes 2004) and the mouse epidermis contains far fewer
keratinocytes than human epidermis. Accordingly, though no directly comparative
studies exist, it seems reasonable that the mouse wounds will contain fewer AMPs
than human wounds. There will also be qualitative difference in the AMPs. The
mouse contains many more different β-defensins than humans (Schutte et al. 2002),
while there will be no neutrophil α-defensins in mouse wounds (Eisenhauer and
Lehrer 1992). The neutrophil α-defensins play a role for the antimicrobial function
of the neutrophils (Sørensen et al. 2014), and this will undoubtedly be important for
the antimicrobial defense also in wounds.
While even infected wounds only in some instances benefit from topical treat-
ment with antibiotics (Lipsky and Hoey 2009), gene delivery of AMPs has been
found to have beneficial effects for wound healing (Jacobsen et al. 2005;
Steinstraesser et al. 2014; Carretero et al. 2008). This clearly indicates that AMPs
have beneficial effects in wound healing beyond its antimicrobial properties. AMPs
are generated at all stages of wound healing and it does seem like AMPs participate
in the regulation of some aspects of the wound healing processes, for instance,
LL-37 plays a role for recruitment of monocytes (Soehnlein et al. 2008b).
Resolution of inflammation is now recognized as a regulated process (Serhan and
Savill 2005; Ortega-Gomez et al. 2013), and in wounds, AMPs may play an impor-
tant role here. Dying and necrotic neutrophils are anti-inflammatory due to the
release of neutrophil defensins (Miles et al. 2009) and calgranulin S100A8, which
induce the anti-inflammatory cytokine IL-10. Later, IL-10 may play a role for
downregulation of AMP expression in keratinocytes (Howell et al. 2005). Both
defensins and LL-37 promote proliferation of keratinocytes (Niyonsaba et al. 2007;
Heilborn et al. 2003). Accordingly, AMPs may play a role for inflammation, resolu-
tion of inflammation, and proliferation during wound healing. One of the para-
doxes of psoriasis, a disease with very prominent AMP expression (Harder and
Schröder 2005), is the lack of scarring following the chronic inflammation
(Nickoloff et al. 2006). Future studies will further address the role of AMPs for
regulation of the wound healing process and scar formation. This will hopefully
pave the way for new treatment modalities for chronic wounds and wound
infection.
Acknowledgments This work was supported by grants from the Alfred Österlunds Stiftelse, the
Royal Physiografic Society Lund, Greta och Johan Kocks Stiftelse, Petrus och Augusta Hedlunds
Stiftelse, and the Swedish Research Council.
1 Antimicrobial Peptides in Cutaneous Wound Healing 9
References
Davidson JM (1998) Animal models for wound repair. Arch Dermatol Res 290(Suppl):S1–S11
Dorschner RA, Pestonjamasp VK, Tamakuwala S, Ohtake T, Rudisill J, Nizet V, Agerberth B,
Gudmundsson GH, Gallo RL (2001) Cutaneous injury induces the release of cathelicidin anti-
microbial peptides active against group A Streptococcus. J Invest Dermatol 117:91–97
Dressel S, Harder J, Cordes J, Wittersheim M, Meyer-Hoffert U, Sunderkotter C, Glaser R (2010)
Differential expression of antimicrobial peptides in margins of chronic wounds. Exp Dermatol
19:628–632
Edgeworth J, Gorman M, Bennett R, Freemont P, Hogg N (1991) Identification of p8,14 as a
highly abundant heterodimeric calcium binding protein complex of myeloid cells. J Biol Chem
266:7706–7713
Edwards R, Harding KG (2004) Bacteria and wound healing. Curr Opin Infect Dis 17:91–96
Eisenhauer PB, Lehrer RI (1992) Mouse neutrophils lack defensins. Infect Immun 60:
3446–3447
Faurschou M, Sørensen OE, Johnsen AH, Askaa J, Borregaard N (2002) Defensin-rich granules of
human neutrophils: characterization of secretory properties. Biochim Biophys Acta 1591:
29–35
Feng Z, Dubyak GR, Lederman MM, Weinberg A (2006) Cutting edge: human beta defensin
3 – a novel antagonist of the HIV-1 coreceptor CXCR4. J Immunol 177:782–786
Flannagan RS, Cosio G, Grinstein S (2009) Antimicrobial mechanisms of phagocytes and bacte-
rial evasion strategies. Nat Rev Microbiol 7:355–366
Frick IM, Åkesson P, Herwald H, Mörgelin M, Malmsten M, Nagler DK, Björck L (2006) The
contact system – a novel branch of innate immunity generating antibacterial peptides. EMBO
J 25:5569–5578
Frick IM, Nordin SL, Baumgarten M, Mörgelin M, Sørensen OE, Olin AI, Egesten A (2011)
Constitutive and inflammation-dependent antimicrobial peptides produced by epithelium are
differentially processed and inactivated by the commensal Finegoldia magna and the pathogen
Streptococcus pyogenes. J Immunol 187:4300–4309
Ganz T (1987) Extracellular release of antimicrobial defensins by human polymorphonuclear leu-
kocytes. Infect Immun 55:568–571
Ganz T, Selsted ME, Szklarek D, Harwig SSL, Daher K, Bainton DF, Lehrer RI (1985) Defensins.
Natural peptide antibiotics of human neutrophils. J Clin Invest 6:1427–1435
Gariboldi S, Palazzo M, Zanobbio L, Selleri S, Sommariva M, Sfondrini L, Cavicchini S, Balsari
A, Rumio C (2008) Low molecular weight hyaluronic acid increases the self-defense of skin
epithelium by induction of beta-defensin 2 via TLR2 and TLR4. J Immunol 181:2103–2110
Gerstel U, Czapp M, Bartels J, Schroder JM (2009) Rhamnolipid-induced shedding of flagellin
from Pseudomonas aeruginosa provokes hBD-2 and IL-8 response in human keratinocytes.
Cell Microbiol 11:842–853
Gläser R, Harder J, Lange H, Bartels J, Christophers E, Schröder JM (2005) Antimicrobial psoria-
sin (S100A7) protects human skin from Escherichia coli infection. Nat Immunol 6:57–64
Harder J, Schröder JM (2002) RNase 7, a novel innate immune defense antimicrobial protein of
healthy human skin. J Biol Chem 277:46779–46784
Harder J, Schröder JM (2005) Psoriatic scales: a promising source for the isolation of human skin-
derived antimicrobial proteins. J Leukoc Biol 77:476–486
Harder J, Bartels J, Christophers E, Schröder JM (1997) A peptide antibiotic from human skin.
Nature 387:861
Harder J, Bartels J, Christophers E, Schröder JM (2001) Isolation and characterization of human
beta -defensin-3, a novel human inducible peptide antibiotic. J Biol Chem 276:5707–5713
Hattori F, Kiatsurayanon C, Okumura K, Ogawa H, Ikeda S, Okamoto K, Niyonsaba F (2014) The
antimicrobial protein S100A7/psoriasin enhances the expression of keratinocyte differentiation
markers and strengthens the skin’s tight junction barrier. Br J Dermatol 171:742–753
Heilborn JD, Nilsson MF, Kratz G, Weber G, Sørensen O, Borregaard N, Stahle-Backdahl M
(2003) The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of
human skin wounds and is lacking in chronic ulcer epithelium. J Invest Dermatol 120:
379–389
1 Antimicrobial Peptides in Cutaneous Wound Healing 11
Hessian PA, Edgeworth J, Hogg N (1993) MRP-8 and MRP-14, two abundant Ca(2+)-binding
proteins of neutrophils and monocytes. J Leukoc Biol 53:197–204
Hiemstra PS, Maassen RJ, Stolk J, Heinzel-Wieland R, Steffens GJ, Dijkman JH (1996)
Antibacterial activity of antileukoprotease. Infect Immun 64:4520–4524
Hiroshima Y, Hsu K, Tedla N, Chung YM, Chow S, Herbert C, Geczy CL (2014) S100A8 induces
IL-10 and protects against acute lung injury. J Immunol 192:2800–2811
Hofmann MA, Drury S, Fu C, Qu W, Taguchi A, Lu Y, Avila C, Kambham N, Bierhaus A, Nawroth
P et al (1999) RAGE mediates a novel proinflammatory axis: a central cell surface receptor for
S100/calgranulin polypeptides. Cell 97:889–901
Howell MD, Novak N, Bieber T, Pastore S, Girolomoni G, Boguniewicz M, Streib J, Wong C,
Gallo RL, Leung DYM (2005) Interleukin-10 downregulates anti-microbial peptide expression
in atopic dermatitis. J Invest Dermatol 125:738–745
Jacobsen F, Mittler D, Hirsch T, Gerhards A, Lehnhardt M, Voss B, Steinau HU, Steinstraesser L
(2005) Transient cutaneous adenoviral gene therapy with human host defense peptide hCAP-
18/LL-37 is effective for the treatment of burn wound infections. Gene Ther 12:1494–1502
Jacobsen LC, Sørensen OE, Cowland JB, Borregaard N, Theilgaard-Mönch K (2008) The secre-
tory leukocyte protease inhibitor (SLPI) and the secondary granule protein lactoferrin are syn-
thesized in myelocytes, colocalize in subcellular fractions of neutrophils, and are coreleased by
activated neutrophils. J Leukoc Biol 83:1155–1164
Jinquan T, Vorum H, Larsen CG, Madsen P, Rasmussen HH, Gesser B, Etzerodt M, Honore B,
Celis JE, Thestrup-Pedersen K (1996) Psoriasin: a novel chemotactic protein. J Invest Dermatol
107:5–10
Joiner KA, Ganz T, Albert J, Rostrosen D (1989) The opsonizing ligand on salmonella typhimurium
influences incorporation of specific, but not azurophil, granule constituents into neutrophil
phagosomes. J Cell Biol 109:2771–2782
Koczulla R, Von Degenfeld G, Kupatt C, Krotz F, Zahler S, Gloe T, Issbrucker K, Unterberger P,
Zaiou M, Lebherz C et al (2003) An angiogenic role for the human peptide antibiotic LL-37/
hCAP-18. J Clin Invest 111:1665–1672
Lan CC, Wu CS, Huang SM, Kuo HY, Wu IH, Wen CH, Chai CY, Fang AH, Chen GS (2011)
High-glucose environment inhibits p38MAPK signaling and reduces human beta-defensin-3
expression in keratinocytes. Mol Med 17:771–779
Lan CC, Wu CS, Huang SM, Kuo HY, Wu IH, Liang CW, Chen GS (2012) High-glucose environ-
ment reduces human beta-defensin-2 expression in human keratinocytes: implications for poor
diabetic wound healing. Br J Dermatol 166:1221–1229
Lee KC, Eckert RL (2007) S100A7 (Psoriasin) – mechanism of antibacterial action in wounds.
J Invest Dermatol 127:945–957
Levy O (1996) Antibiotic proteins of polymorphonuclear leukocytes. Eur J Haematol 56:
263–277
Li J, Post M, Volk R, Gao Y, Li M, Metais C, Sato K, Tsai J, Aird W, Rosenberg RD et al (2000)
PR39, a peptide regulator of angiogenesis. Nat Med 6:49–55
Li J, Raghunath M, Tan D, Lareu RR, Chen Z, Beuerman RW (2006) Defensins HNP1 and HBD2
stimulation of wound-associated responses in human conjunctival fibroblasts. Invest
Ophthalmol Vis Sci 47:3811–3819
Li D, Lei H, Li Z, Li H, Wang Y, Lai Y (2013) A novel lipopeptide from skin commensal activates
TLR2/CD36-p38 MAPK signaling to increase antibacterial defense against bacterial infection.
PLoS One 8:e58288
Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, Collins M, Fouser LA
(2006) Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance
expression of antimicrobial peptides. J Exp Med 203:2271–2279
Lim SY, Raftery M, Cai H, Hsu K, Yan WX, Hseih HL, Watts RN, Richardson D, Thomas S, Perry
M et al (2008) S-nitrosylated S100A8: novel anti-inflammatory properties. J Immunol 181:
5627–5636
Lipsky BA, Hoey C (2009) Topical antimicrobial therapy for treating chronic wounds. Clin Infect
Dis 49:1541–1549
12 O.E. Sørensen
Liu AY, Destoumieux D, Wong AV, Park CH, Valore EV, Liu L, Ganz T (2002) Human beta-
defensin-2 production in keratinocytes is regulated by interleukin-1, bacteria, and the state of
differentiation. J Invest Dermatol 118:275–281
Liu L, Roberts AA, Ganz T (2003) By IL-1 signaling, monocyte-derived cells dramatically
enhance the epidermal antimicrobial response to lipopolysaccharide. J Immunol 170:
575–580
Lundqvist K, Sørensen OE, Schmidtchen A (2008) Increased levels of human neutrophil alpha-
defensins in chronic venous leg ulcers. J Dermatol Sci 51:131–134
Maerki C, Meuter S, Liebi M, Muhlemann K, Frederick MJ, Yawalkar N, Moser B, Wolf M (2009)
Potent and broad-spectrum antimicrobial activity of CXCL14 suggests an immediate role in
skin infections. J Immunol 182:507–514
Malmsten M, Davoudi M, Walse B, Rydengard V, Pasupuleti M, Mörgelin M, Schmidtchen A
(2007) Antimicrobial peptides derived from growth factors. Growth Factors 25:60–70
McNeely TB, Dealy M, Dripps DJ, Orenstein JM, Eisenberg SP, Wahl SM (1995) Secretory leu-
kocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency
virus 1 activity in vitro. J Clin Invest 96:456–464
Mestas J, Hughes CC (2004) Of mice and not men: differences between mouse and human immu-
nology. J Immunol 172:2731–2738
Miles K, Clarke DJ, Lu W, Sibinska Z, Beaumont PE, Davidson DJ, Barr TA, Campopiano DJ,
Gray M (2009) Dying and necrotic neutrophils are anti-inflammatory secondary to the release
of alpha-defensins. J Immunol 183:2122–2132
Mork G, Schjerven H, Mangschau L, Soyland E, Brandtzaeg P (2003) Proinflammatory cytokines
upregulate expression of calprotectin (L1 protein, MRP-8/MRP-14) in cultured human kerati-
nocytes. Br J Dermatol 149:484–491
Mullins ES, Kombrinck KW, Talmage KE, Shaw MA, Witte DP, Ullman JM, Degen SJ, Sun W,
Flick MJ, Degen JL (2009) Genetic elimination of prothrombin in adult mice is not compatible
with survival and results in spontaneous hemorrhagic events in both heart and brain. Blood
113:696–704
Murphy CJ, Foster BA, Mannis MJ, Selsted ME, Reid TW (1993) Defensins are mitogenic for
epithelial cells and fibroblasts. J Cell Physiol 155:408–413
Nagaoka I, Tamura H, Hirata M (2006) An antimicrobial cathelicidin peptide, human CAP18/
LL-37, suppresses neutrophil apoptosis via the activation of formyl-peptide receptor-like 1 and
P2X7. J Immunol 176:3044–3052
Nagy I, Pivarcsi A, Koreck A, Szell M, Urban E, Kemeny L (2005) Distinct strains of
Propionibacterium acnes induce selective human beta-defensin-2 and interleukin-8 expression
in human keratinocytes through toll-like receptors. J Invest Dermatol 124:931–938
Nakatsuji T, Chiang HI, Jiang SB, Nagarajan H, Zengler K, Gallo RL (2013) The microbiome
extends to subepidermal compartments of normal skin. Nat Commun 4:1431
Nickoloff BJ, Bonish BK, Marble DJ, Schriedel KA, DiPietro LA, Gordon KB, Lingen MW
(2006) Lessons learned from psoriatic plaques concerning mechanisms of tissue repair, remod-
eling, and inflammation. J Investig Dermatol Symp Proc 11:16–29
Niyonsaba F, Ushio H, Nakano N, Ng W, Sayama K, Hashimoto K, Nagaoka I, Okumura K,
Ogawa H (2007) Antimicrobial peptides human beta-defensins stimulate epidermal keratino-
cyte migration, proliferation and production of proinflammatory cytokines and chemokines.
J Invest Dermatol 127:594–604
Nordahl EA, Rydengard V, Nyberg P, Nitsche DP, Morgelin M, Malmsten M, Bjorck L,
Schmidtchen A (2004) Activation of the complement system generates antibacterial peptides.
Proc Natl Acad Sci U S A 101:16879–16884
Nurjadi D, Herrmann E, Hinderberger I, Zanger P (2013) Impaired beta-defensin expression in
human skin links DEFB1 promoter polymorphisms with persistent Staphylococcus aureus
nasal carriage. J Infect Dis 207:666–674
Ortega-Gomez A, Perretti M, Soehnlein O (2013) Resolution of inflammation: an integrated view.
EMBO Mol Med 5:661–674
Otte JM, Werner I, Brand S, Chromik AM, Schmitz F, Kleine M, Schmidt WE (2008) Human beta
defensin 2 promotes intestinal wound healing in vitro. J Cell Biochem 104:2286–2297
1 Antimicrobial Peptides in Cutaneous Wound Healing 13
Påhlman LI, Mörgelin M, Kasetty G, Olin AI, Schmidtchen A, Herwald H (2013) Antimicrobial
activity of fibrinogen and fibrinogen-derived peptides – a novel link between coagulation and
innate immunity. Thromb Haemost 109:930–939
Panyutich P, Shi J, Boutz PL, Zhao C, Ganz T (1997) Porcine polymorphonuclear leukocytes gen-
erate extracellular microbial activity by elastase-mediated activation of secreted proprotegrins.
Infect Immun 65:978–985
Papareddy P, Rydengard V, Pasupuleti M, Walse B, Morgelin M, Chalupka A, Malmsten M,
Schmidtchen A (2010) Proteolysis of human thrombin generates novel host defense peptides.
PLoS Pathog 6:e1000857
Pasupuleti M, Walse B, Nordahl EA, Morgelin M, Malmsten M, Schmidtchen A (2007)
Preservation of antimicrobial properties of complement peptide C3a, from invertebrates to
humans. J Biol Chem 282:2520–2528
Raftery MJ, Yang Z, Valenzuela SM, Geczy CL (2001) Novel intra- and inter-molecular sulfin-
amide bonds in S100A8 produced by hypochlorite oxidation. J Biol Chem 276:33393–33401
Rodriguez-Martinez S, Cancino-Diaz JC, Vargas-Zuniga LM, Cancino-Diaz ME (2008) LL-37
regulates the overexpression of vascular endothelial growth factor (VEGF) and c-IAP-2 in
human keratinocytes. Int J Dermatol 47:457–462
Röhrl J, Yang D, Oppenheim JJ, Hehlgans T (2010) Human beta-defensin 2 and 3 and their mouse
orthologs induce chemotaxis through interaction with CCR2. J Immunol 184:6688–6694
Roupé KM, Nybo M, Sjöbring U, Alberius P, Schmidtchen A, Sørensen OE (2010) Injury is a
major inducer of epidermal innate immune responses during wound healing. J Invest Dermatol
130:1167–1177
Ryckman C, Vandal K, Rouleau P, Talbot M, Tessier PA (2003) Proinflammatory activities of
S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion.
J Immunol 170:3233–3242
Schmid P, Grenet O, Medina J, Chibout SD, Osborne C, Cox DA (2001) An intrinsic antibiotic
mechanism in wounds and tissue-engineered skin. J Invest Dermatol 116:471–472
Schmidtchen A, Frick IM, Andersson E, Tapper H, Björck L (2002) Proteinases of common patho-
genic bacteria degrade and inactivate the antibacterial peptide LL-37. Mol Microbiol
46:157–168
Schutte BC, Mitros JP, Bartlett JA, Walters JD, Jia HP, Welsh MJ, Casavant TL, McCray PB Jr
(2002) Discovery of five conserved beta -defensin gene clusters using a computational search
strategy. Proc Natl Acad Sci U S A 99:2129–2133
Scocchi M, Skerlavaj B, Romeo D, Gennaro R (1992) Proteolytic cleavage by neutrophil elastase
converts inactive storage proforms to antibacterial bactenecins. Eur J Biochem 209:589–595
Serhan CN, Savill J (2005) Resolution of inflammation: the beginning programs the end. Nat
Immunol 6:1191–1197
Shubbar E, Vegfors J, Carlstrom M, Petersson S, Enerback C (2012) Psoriasin (S100A7) increases
the expression of ROS and VEGF and acts through RAGE to promote endothelial cell prolif-
eration. Breast Cancer Res Treat 134:71–80
Simanski M, Dressel S, Glaser R, Harder J (2010) RNase 7 protects healthy skin from
Staphylococcus aureus colonization. J Invest Dermatol 130:2836–2838
Simanski M, Rademacher F, Schroder L, Schumacher HM, Glaser R, Harder J (2013) IL-17A and
IFN-gamma synergistically induce RNase 7 expression via STAT3 in primary keratinocytes.
PLoS One 8:e59531
Simard JC, Simon MM, Tessier PA, Girard D (2011) Damage-associated molecular pattern
S100A9 increases bactericidal activity of human neutrophils by enhancing phagocytosis.
J Immunol 186:3622–3631
Simmaco M, Mignogna G, Barra D (1998) Antimicrobial peptides from amphibian skin: what do
they tell us? Biopolymers 47:435–450
Singer AJ, Clark RA (1999) Cutaneous wound healing. N Engl J Med 341:738–746
Soehnlein O, Kai-Larsen Y, Frithiof R, Sorensen OE, Kenne E, Scharffetter-Kochanek K, Eriksson
EE, Herwald H, Agerberth B, Lindbom L (2008a) Neutrophil primary granule proteins HBP
and HNP1-3 boost bacterial phagocytosis by human and murine macrophages. J Clin Invest
118:3491–3502
14 O.E. Sørensen
Soehnlein O, Zernecke A, Eriksson EE, Rothfuchs AG, Pham CT, Herwald H, Bidzhekov K,
Rottenberg ME, Weber C, Lindbom L (2008b) Neutrophil secretion products pave the way for
inflammatory monocytes. Blood 112:1461–1471
Sonesson A, Ringstad L, Nordahl EA, Malmsten M, Morgelin M, Schmidtchen A (2007)
Antifungal activity of C3a and C3a-derived peptides against Candida. Biochim Biophys Acta
1768:346–353
Sørensen O, Arnljots K, Cowland JB, Bainton DF, Borregaard N (1997) The human antibacterial
Cathelicidin, hCAP-18, is synthesized in myelocytes and metamyelocytes and localized to spe-
cific granules in neutrophils. Blood 90:2796–2803
Sørensen OE, Follin P, Johnsen AH, Calafat J, Tjabringa GS, Hiemstra PS, Borregaard N (2001)
Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular
cleavage with proteinase 3. Blood 97:3951–3959
Sørensen OE, Cowland JB, Theilgaard-Mönch K, Liu L, Ganz T, Borregaard N (2003) Wound
healing and expression of antimicrobial peptides/polypeptides in keratinocytes, a consequence
of common growth factors. J Immunol 170:5583–5589
Sørensen OE, Thapa DR, Rosenthal A, Liu L, Roberts AA, Ganz T (2005) Differential regulation
of β-defensin expression in human skin by microbial stimuli. J Immunol 174:4870–4879
Sørensen OE, Thapa DR, Roupe KM, Valore EV, Sjobring U, Roberts AA, Schmidtchen A, Ganz
T (2006) Injury-induced innate immune response in human skin mediated by transactivation of
the epidermal growth factor receptor. J Clin Invest 116:1878–1885
Sørensen OE, Schmidtchen A, Roupe KM (2008) EGF receptor: role for innate immunity during
wound healing in human skin. Expert Rev Dermatol 3:587–593
Sørensen OE, Clemmensen SN, Dahl SL, Østergaard O, Heegaard NH, Glenthøj A, Nielsen FC,
Borregaard N (2014) Papillon-Lefevre syndrome patient reveals species-dependent require-
ments for neutrophil defenses. J Clin Invest 124:4539–4548
Steinbakk M, Naess-Andresen CF, Lingaas E, Dale I, Brandtzaeg P, Fagerhol MK (1990)
Antimicrobial actions of calcium binding leukocyte L1 protein, calprotectin. Lancet 336:
763–765
Steinstraesser L, Lam MC, Jacobsen F, Porporato PE, Chereddy KK, Becerikli M, Stricker I,
Hancock RE, Lehnhardt M, Sonveaux P et al (2014) Skin electroporation of a plasmid encod-
ing hCAP-18/LL-37 host defense peptide promotes wound healing. Mol Ther 22:734–742
Territo MC, Ganz T, Selsted ME, Lehrer RI (1989) Monocyte-chemotactic activity of defensins
from human neutrophils. J Clin Invest 84:2017–2020
Theilgaard-Mönch K, Knudsen S, Follin P, Borregaard N (2004) The transcriptional activation
program of human neutrophils in skin lesions supports their important role in wound healing.
J Immunol 172:7684–7693
Thorey IS, Roth J, Regenbogen J, Halle JP, Bittner M, Vogl T, Kaesler S, Bugnon P, Reitmaier B,
Durka S et al (2001) The Ca2 + −binding proteins S100A8 and S100A9 are encoded by novel
injury-regulated genes. J Biol Chem 276:35818–35825
Tjabringa GS, Aarbiou J, Ninaber DK, Drijfhout JW, Sørensen OE, Borregaard N, Rabe KF,
Hiemstra PS (2003) The antimicrobial peptide LL-37 activates innate immunity at the airway
epithelial surface by transactivation of the epidermal growth factor receptor. J Immunol
171:6690–6696
Tokumaru S, Higashiyama S, Endo T, Nakagawa T, Miyagawa JI, Yamamori K, Hanakawa Y,
Ohmoto H, Yoshino K, Shirakata Y et al (2000) Ectodomain shedding of epidermal growth
factor receptor ligands is required for keratinocyte migration in cutaneous wound healing.
J Cell Biol 151:209–220
Tokumaru S, Sayama K, Shirakata Y, Komatsuzawa H, Ouhara K, Hanakawa Y, Yahata Y, Dai XJ,
Tohyama M, Nagai H et al (2005) Induction of keratinocyte migration via transactivation of the
epidermal growth factor receptor by the antimicrobial peptide LL-37. J Immunol 175:
4662–4668
Tomee JF, Hiemstra PS, Heinzel-Wieland R, Kauffman HF (1997) Antileukoprotease: an endog-
enous protein in the innate mucosal defense against fungi. J Infect Dis 176:740–747
1 Antimicrobial Peptides in Cutaneous Wound Healing 15
Turner J, Cho Y, Dihn NN, Waring A, Lehrer RI (1998) Activities of LL-37, a Cathelin-associated
antimicrobial peptide of human neutrophils. Antimicrob Agents Chemother 42:2206–2214
Urban CF, Ermert D, Schmid M, Abu-Abed U, Goosmann C, Nacken W, Brinkmann V, Jungblut
PR, Zychlinsky A (2009) Neutrophil extracellular traps contain calprotectin, a cytosolic protein
complex involved in host defense against Candida albicans. PLoS Pathog 5:e1000639
vanBergen BH, Andriessen MPM, Spruijt KIJ, van de Kerkhof PCM, Schalkwijk J (1996)
Expression of SKALP/elafin during wound healing in human skin. Arch Dermatol Res
288:458–462
Vandal K, Rouleau P, Boivin A, Ryckman C, Talbot M, Tessier PA (2003) Blockade of S100A8
and S100A9 suppresses neutrophil migration in response to lipopolysaccharide. J Immunol
171:2602–2609
Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MAD, Nacken W, Foell D, van
der Poll T, Sorg C et al (2007) Mrp8 and Mrp14 are endogenous activators of Toll- like receptor
4, promoting lethal, endotoxin-induced shock. Nat Med 13:1042–1049
Wanke I, Steffen H, Christ C, Krismer B, Gotz F, Peschel A, Schaller M, Schittek B (2011) Skin
commensals amplify the innate immune response to pathogens by activation of distinct signal-
ing pathways. J Invest Dermatol 131:382–390
Ward PA (1968) Chemotaxis of mononuclear cells. J Exp Med 128:1201–1221
Wingens M, van Bergen BH, Hiemstra PS, Meis JF, van Vlijmen WIM, Zeeuwen PL, Mulder J,
Kramps HA, van Ruissen F, Schalkwijk J (1998) Induction of SLPI (ALP/HUSI-I) in epider-
mal keratinocytes. J Invest Dermatol 111:996–1002
Wolf R, Howard OM, Dong HF, Voscopoulos C, Boeshans K, Winston J, Divi R, Gunsior M,
Goldsmith P, Ahvazi B et al (2008) Chemotactic activity of S100A7 (Psoriasin) is mediated by
the receptor for advanced glycation end products and potentiates inflammation with highly
homologous but functionally distinct S100A15. J Immunol 181:1499–1506
Yang D, Chertov O, Bykovskaia N, Chen Q, Buffo MJ, Shogan J, Anderson M, Schroder JM,
Wang JM, Howard OMZ et al (1999) beta-defensins: linking innate and adaptive immunity
through dendritic and T cell CCR6. Science 286:525–528
Yang D, Chen Q, Chertov O, Oppenheim JJ (2000a) Human neutrophil defensins selectively che-
moattract naive T and immature dendritic cells. J Leukoc Biol 68:9–14
Yang D, Chen Q, Schmidt AP, Anderson GM, Wang JM, Wooters J, Oppenheim JJ, Chertov O
(2000b) LL-37, the Neutrophil Granule- and epithelial cell-derived cathelicidin, utilizes formyl
peptide receptor -like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutro-
phil, monocytes, and T cells. J Exp Med 192:1069–1074
Zanetti M (2004) Cathelicidins, multifunctional peptides of the innate immunity. J Leukoc Biol
75:39–48
Zanger P, Holzer J, Schleucher R, Scherbaum H, Schittek B, Gabrysch S (2010) Severity of
Staphylococcus aureus infection of the skin is associated with inducibility of human beta-
defensin 3 but not human beta-defensin 2. Infect Immun 78:3112–3117
Zanger P, Nurjadi D, Vath B, Kremsner PG (2011) Persistent nasal carriage of Staphylococcus
aureus is associated with deficient induction of human beta-defensin 3 after sterile wounding
of healthy skin in vivo. Infect Immun 79:2658–2662
Zasloff M (1987) Magainins, a class of antimicrobial peptides from Xenopus skin: isolation, char-
acterization of two active forms, and partial cDNA sequence of a precursor. Proc Natl Acad Sci
U S A 84:5449–5453
Zhu J, Nathan C, Jin W, Sim D, Ashcroft GS, Wahl SM, Lacomis L, Erdjument-Bromage H,
Tempst P, Wright CD et al (2002) Conversion of proepithelin to epithelins: roles of SLPI and
elastase in host defense and wound repair. Cell 111:867–878
Chapter 2
Antimicrobial Peptides as Endogenous
Antibacterials and Antivirals at the Ocular
Surface
2.1 Introduction
The ocular surface and its adnexal structures comprise the cornea, conjunctiva
with the bulbar, the fornical, and palpebral parts, the main lacrimal gland, the
glands of the eyelids (including the meibomian glands, the glands of Moll, and
accessory lacrimal glands), as well as the nasolacrimal system (or efferent tear
ducts with the upper and lower puncta, the paired lacrimal canaliculi, the lacri-
mal sac, and the nasolacrimal duct). The nasolacrimal system collects the tear
fluid from the ocular surface and conveys it into the nasal cavity. All the other
structures contribute to formation of the preocular tear film. The tear film
together with the glands and cells that produce it (lacrimal glands, accessory
lacrimal glands, ocular surface epithelia, meibomian glands, glands of Moll, and
the sensory and motor nerves connecting these) has been assigned the general
term lacrimal functional unit (LFU) (Stern et al. 1998). Disease in or damage to
any component of the LFU can destabilize the tear film and lead to dry eye dis-
ease (DED), a condition expressed by signs and/or symptoms of ocular irrita-
tion, redness, and foreign body sensation. DED afflicts millions of people around
the world and makes the ocular surface more vulnerable to infection (Gayton
2009).
The tear film coats the cornea and conjunctiva and is a complex structure com-
posed of an inner mucous layer that is part of the glycocalyx and is attached to the
epithelial cells (composed mainly of membrane-bound mucins), an aqueous com-
ponent consisting, beside water (99 %) of ions, soluble mucins and a wide range
of different proteins and peptides (such as trefoil factor family peptides that func-
tion as linker molecules for mucins to form gels), as well as an outer, anterior-most
lipid component that prevents evaporation. Ocular mucins and trefoil factor family
(TFF) peptides take a variety of different functions which provide protection at the
ocular surface. Mucins are a family of high molecular weight, heavily glycosyl-
ated hydrophilic proteins. Membrane-anchored mucins, MUC1, MUC4, und
MUC16, are part of the epithelial glycocalyx and provide a continuous anatomic
barrier across the ocular surface which prevents pathogen penetration. The mem-
brane-spanning molecules also have signaling capabilities that influence epithelial
activity. The negative charge of the mucins (glycocalyx) occurs in an accumulation
of positive-charged tear components, like lysozyme, SLPI, as well as AMPs (see
below), which enhance the local concentration of antimicrobial agents at the ocu-
lar surface. The high molecular secreted mucins are distinguished in gel-forming
mucins (mainly MUC5AC from conjunctival goblet cells and MUC5B from the
lacrimal gland) and small soluble mucins (MUC7 from the lacrimal gland). The
secreted mucins as well as TFFs are responsible for the rheological properties of
the tear film, enabling movement and spreading of the tear film. TFF peptides have
many other physiological functions in addition to their rheological properties,
such as promotion of epithelial cell migration, antiapoptotic properties, induction
of cell scattering, epithelial restitution, and neuropeptide functions (Paulsen and
Berry 2006).
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 19
Like other epithelia, also the ocular surface is constantly in contact with various
microorganism and pathogen-associated molecular patterns (PAMPs). The ocular
surface epithelia and associated structures of the lacrimal apparatus are effective in
counteracting invasion and colonization of microorganisms. They produce the tear
film that contains up to 1500 different proteins (Zhou et al. 2012). More than 90 %
of the total amount of tear proteins are four antimicrobial compounds: lysozyme,
lactoferrin, tear lipocalin and secreted immunoglobulin A (sIgA). The concentration
of these four proteins in the tear fluid is in the mg/ml range. These tear compounds
are well known and show broad antimicrobial activity. Lysozyme cleaves the pepti-
doglycan backbone of bacterial cell walls and is also able to digest fungal cell walls.
Lactoferrin binds free iron at the ocular surface and inhibits bacterial and fungal
growth. It has also been shown to have iron-independent anti-HIV activity. Tear
lipocalin binds microbial siderophores (iron chelating compounds) and thus has a
bacteriostatic effect. Lysozyme, lactoferrin, and tear lipocalin are built mainly by
the acinar cells of the main lacrimal gland as well as by accessory lacrimal glands
in the eyelids (glandulae conjunctivales [glands of Krause and Wolfring]). In con-
trast, sIgA is produced as a dimer by subepithelial plasma cells, transported via
transcytosis, bound to a protein termed “secretory component,” through the epithe-
lium and then is secreted into the tear film (Mcdermott 2013; Tiffany 2008).
In addition to these principal antimicrobial factors, tear fluid contains a great
bulk of other tear compounds, several of them with antimicrobial activity. The con-
centration of these compounds is in the range of μg/ml to pg/ml. Beside their anti-
microbial activity, several of these compounds have no antimicrobial activity as
main biological function and several are with yet unknown function (Mcdermott
2013). Among those constituents with antimicrobial activity is secretory phospholi-
pase A2 (sPLA2). It binds to anionic bacterial membranes of Gram-positive bacte-
ria due to its cationic domains and kills bacteria by its enzymatic activity. sPLA2
shows activity against Gram-positive bacteria in the physiological tear environment
(Qu and Lehrer 1998). Antimicrobial activity and anti-inflammatory properties
were shown for members of the whey acidic protein (WAP) family like secretory
leukocyte protease inhibitor (SLPI) as well as elafin, which are characterized by a
cysteine-rich region with four intramolecular disulfide bonds. SLPI was identified
by its antiprotease activity and shows also activity against Gram-positive and Gram-
negative bacteria, fungi, and HIV (Sallenave 2010). High amounts could be detected
in closed-eye tears. SLPI may inhibit neutrophil elastase derived from immigrating
neutrophilic granulocytes and protects the ocular surface from enzymatic degrada-
tion effects during the sleep (Sathe et al. 1998). Furthermore, bactericidal/
permeability-increasing protein (BPI) was initially identified in neutrophils, but it is
also expressed by ocular surface epithelial cells and could be detected in tears. BPI
binds lipopolysaccharides (LPS) liberated from Gram-negative bacteria (Peuravuori
et al. 2006). Clinical trials with recombinant modified BPI molecule (rBPI21)
20 F. Garreis et al.
During recent years, the well-known lung surfactant proteins (SPs) have also been
described in other mucosae such as in the ocular system (Schicht et al. 2010). Here,
they have been associated with surface tension regulating activities of the tear film
as well as with immunological functions. Tear SP-D has been shown to protect the
ocular surface from infection with Gram-negative bacteria, in particular
Pseudomonas aeruginosa (Mcdermott 2013). With regard to virus infection, SP-A
and SP-D are able to bind and agglutinate viruses (such as influenza A virus, adeno-
virus, hepatitis B virus [HBV], and herpes simplex virus [HSV]) and they have been
shown to initiate phagocytosis by macrophages (Harrod et al. 1999; Kishore et al.
2006; Levine et al. 2001). In chronic herpes virus infection, a protein misfolding
shall lead to functional loss of SP-C with losing of the surface regulatory properties
(Lawson et al. 2008). Also, the recently discovered surfactant protein H (SFTA3)
has been demonstrated to occur at the ocular surface (Schicht 2012). Also, this sur-
factant protein has immunological functions by mediating activation of macro-
phages (Diler et al. 2014). However, so far, there exist no data demonstrating any
function of surfactant proteins with regard to viral eye infection.
AMPs are defined as small endogenous proteins displaying direct antimicrobial activ-
ity. In general, AMPs have no specific consensus amino acid sequence but most of
them have an amphipathic character and are positively charged under physiological pH
conditions. AMPs have been detected across a wide spectrum of different species. In
February 2015, the Antimicrobial Peptide Database (APD; http://aps.unmc.edu/AP/
main.php) lists 2493 AMPs: 244 from bacteria (bacteriocins), 2 from archaea, 7 from
protists, 13 from fungi, 312 from plants, and 1874 from animals. In human, 108 differ-
ent AMPs have been described to date (Wang 2014). According to their secondary
structure, AMPs can be classified into four groups: (i) AMPs with linear α-helical
domains (e.g., human cathelicidin LL-37), (ii) AMPs with β-sheets and disulfide bonds
(e.g., α- and β-defensins), (iii) AMPs with extended structures (e.g., indolicidin), and
(iv) AMPs with loop structures (cyclic defensins, bactenecin) (Lai and Gallo 2009).
Various different human AMPs with activity against bacteria, fungi, and viruses
have been described and characterized (Wang 2014). Figure 2.1 provides an over-
view about known AMPs at the ocular surface and lacrimal structures.
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 21
Glands of Moll
hBD-1,-2 Lacrimal gland
LL-37 hBD-1,-2,-3
Psoriasin
Cornea
epithelium
hBD-1,-2,-3,-9
LL-37
RNase-7
Psoriasin
LEAP1/Hepcidin
Tears
HNP 1-3
LL-37,
hBD-2,-3
Histatin 5
Dermcidin
Psoriasin
Conjunctiva
epithelium
hBD-1,-2,-3,-9
LL-37
RNase-7
Psoriasin
Meibomian
glands
Psoriasin
Fig. 2.1 Expression of antimicrobial peptides with antibacterial and antiviral activity at the ocular
surface structures, in the lacrimal gland, and in tears (Partly modified from Kolar and Mcdermott
(2011))
Human defensins are small in size (29–45 amino acids), are cationic, and are char-
acterized by the presence of six conserved cysteine residues. Based on the distribu-
tion of the cysteines and the linkage of disulfide bonds, defensins are classified into
three subfamilies, referred to α-, β-, and θ-defensins (theta or minidefensins).
Humans express six α-defensins and up to 31 β-defensins. α-Defensins and
β-defensins differ by their organization of the three disulfide bonds (Hazlett and Wu
2011). θ-Defensins are not translated in humans due to premature stop codons pre-
venting their expression (Lehrer et al. 2012). Interestingly, pseudogenes of
22 F. Garreis et al.
θ-defensins, named retrocyclins, show antimicrobial activity against the gut bacte-
rium E. coli as well as against HIV-1 (Tran et al. 2008; Wang et al. 2004). Human
α-defensins have been divided into human neutrophil peptides (HNP) 1–4 and
human defensin (HD)-5 and human defensin-6. HNP1–HNP4 were identified in
particularly high concentrations in azurophil granules of neutrophils but also in
other immune cells. In contrast, HD-5 and HD-6 are expressed by Paneth cells in the
small intestine as well as different epithelial cells (Lehrer et al. 2012). Human
β-defensins (hBD) 1–3 are expressed by epithelial cells and also in different immune
cells (Semple and Dorin 2012).
At the ocular surface, α-defensins and hBD-1 are constitutively expressed
whereas hBD-2 and hBD-3 show a low basal expression at the ocular surface but
enhanced expression after stimulation with different pro-inflammatory stimuli,
including cytokines and bacterial compounds (Garreis et al. 2010). In addition,
hBD-9 (DEFB109) has been identified at the ocular surface. Interestingly, hBD-9
shows a reduced ex vivo gene expression in patients with keratitis and also a down-
regulation in human corneal limbal epithelial cells after cocultivation with
Acanthamoeba spp. and bacteria from keratitis patients (Abedin et al. 2008; Otri
et al. 2012).
Both α- and β-defensins have broad activity against different microbes, including
various bacteria and viruses in vitro (Gwyer Findlay et al. 2013; Klotman and Chang
2006; Wilson et al. 2013). In vivo, only α-defensin concentrations are generally
within the range of direct antimicrobial activity whereas the concentrations of con-
stitutively expressed β-defensins often do not reach levels sufficient for a direct
antimicrobial activity (Wilson et al. 2013). Interestingly, various studies indicate
that the local β-defensin concentration can be increased markedly by various inflam-
matory and (patho)physiological processes. HBD-1 becomes a potent antimicrobial
peptide after reduction of disulfide bridges against gut pathogens (Schroeder et al.
2011). In addition, all human defensins have also several immunomodulatory prop-
erties (see below).
Beside defensins and cathelicidins, also other AMPs and other antimicrobial com-
pounds have been demonstrated to show antibacterial and antiviral activity at the ocu-
lar surface. Thus, a group of human inhibitors of neutrophil serine proteases known as
SLPI (secretory leukocyte protease inhibitor) and elafin (also known as peptidase
inhibitor 3 or skin-derived antileucoprotease [SKALP]) have been associated with
anti-HIV activity of vaginal fluid (Ma et al. 2004; Pillay et al. 2001). The antiviral
activity does not depend on inhibition of serine proteases. It is associated with binding
of host cell membrane-associated proteins such as scramblase and/or annexin II (Ma
et al. 2004; Tseng and Tseng 2000). The antifungal peptide histatins (histidine-rich
cationic peptides) are present in high amounts in both human saliva and tears and are
also discussed for their antiviral activity. A histatin-5 derivate affects HIV-1 replica-
tion by promoting host cell entry (Groot et al. 2006). Psoriasin (S100A7), a highly
potent AMP against Escherichia coli, was detected in elevated concentrations in
human tears and in tissues of the ocular surface and lacrimal apparatus (Garreis et al.
2011). Two RNases from eosinophils, eosinophil-derived neurotoxin (RNase 2/EDN),
and eosinophil-cationic protein (RNase 3/ECP) have been shown to inhibit respira-
tory syncytial viruses (RSV) and HIV infection in vitro (Domachowske et al. 1998).
RNase 2 antiviral activity depends on ribonucleolytic activity. In addition, RNase 2
functions as chemoattractant and can activate immune cells via Toll-like receptor
(TLR)-2 and myeloid differentiation factor 88 (Myd88) signaling. RNase 3 is able to
permeabilize microbial membranes and stimulate histamine release from mast cells
(Wiesner and Vilcinskas 2010). In addition, also RNase 5 (angiogenin) acts antivirally
against HIV with a yet unsolved mechanism (Cocchi et al. 2012). In contrast to
RNases 2 and 3, which have not yet been analyzed at the ocular surface and in tears,
tear fluid contains very high amounts of RNase 5 (Sack et al. 2005). However, neither
the cellular source and whether and how it is functionally active at the ocular surface
is unclear yet. A further RNase has been described at the ocular surface: RNase 7.
However, RNase 7 has not yet been associated with antiviral activity (Boix and
Nogues 2007). It plays an important role as antibacterial factor in cutaneous defense
(Simanski et al. 2012) and its expression in corneal epithelial cells, which is regulated
by tears and specific microRNAs, suggests that RNase 7 may also participate to pro-
tect the ocular surface against bacterial infection (Mun et al. 2013). The liver-expressed
antimicrobial peptide (LEAP), 1/hepcidin, is expressed at the ocular surface and
shows increased expression in viral keratitis (Mohammed et al. 2011).
AMPs are an essential part of the innate immune defense at the ocular surface, with
both direct antimicrobial activity and immunomodulatory functions. AMPs demon-
strate direct antimicrobial activity against a broad spectrum of ocular pathogens,
including Gram-positive and Gram-negative bacteria, fungi, and various viruses.
24 F. Garreis et al.
Several AMPs have an antiviral activity against different viruses including human
immunodeficiency virus (HIV), herpes simplex virus (HSV), influenza A virus
(IAV), and non-enveloped viruses, like adenovirus (AV) and human papillomavirus
(HPV). Figure 2.2 shows various antiviral mechanisms by AMPs. These include:
• Direct interaction (“virolysis”)
• Blockage of host cell surface receptors
• Inhibition of viral fusion to host cells
• Aggregation of viruses
• Inhibition of viral replication
• Activation of adaptive immune response
Most of the antiviral properties of AMPs are based on direct interaction between
cationic AMPs and different components of the virus. The major compound of
enveloped viruses is the negatively charged lipid bilayer. Cationic AMPs interact
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 25
Fig. 2.2 Overview of the main antiviral mechanisms of antimicrobial peptides (AMPs). AMPs
show activity against various cell surface targets (virus and/or host cell) as well as intracellular
targets. In vitro, cationic AMPs have been shown to decrease viral infection by following mecha-
nism: 1 direct interaction with envelope virus (“virolysis”), 2 blocking of viral entry into host cell
by binding of viral and/or host cell receptors, 3 suppressing viral fusion with host cell, 4 extracel-
lular aggregation of viruses, 5 activation of adaptive immune response, and 6 inhibition of viral
replication. Arrow indicates blockage of viral transcription
with negatively charged phospholipids of the viral envelope. This leads to a desta-
bilization and neutralization of the virus (“virolysis”) and additionally to an inhibi-
tion of viral fusion with host cells (Wilson et al. 2013). Furthermore, AMPs interact
with viral attachment proteins (glycoproteins as well as capsid proteins) and endog-
enous host cell receptors. These blocking mechanisms inhibit the viral binding to
host cells and inhibit virus infection (Wilson et al. 2013). A critical step in virus
infection is the penetration of the virus (genome) into the host cell. Enveloped
viruses have to fuse their lipid bilayer with the cell membrane of the host cell. Non-
enveloped viruses penetrate cell membranes by specific viral capsid proteins.
Several studies demonstrate inhibition of the viral fusion process by AMPs through
preventing penetration of host cell membranes (Wilson et al. 2013). In addition,
AMPs initiate accumulation and aggregation of viruses in vitro. It has not been
26 F. Garreis et al.
2.4 Keratitis
Various studies have investigated AMP activity at the ocular surface related to bac-
terial infection. Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus
pneumonia, and Pseudomonas aeruginosa are reported to be the most common
pathogens associated with bacterial keratitis (Karsten et al. 2012). P. aeruginosa
infection is the most common cause for bacterial keratitis in contact lens wearers.
As described before, human defensins and LL-37 are expressed at the ocular surface
and show varying levels of antibacterial activity against common ocular bacteria in
vitro. Antimicrobial activity occurs in a low micromolar range, and AMPs show
large variability in their efficiency in killing specific pathogens (McDermott 2013).
Generally, the concentrations of AMPs in tears are too low for direct antibacterial
activity. Furthermore, some AMPs show a reduced antibacterial activity in the pres-
ence of tear-specific salt concentration as well as in contact with ocular mucins.
However, studies have also shown an elevated secretion of AMPs under inflamma-
tory conditions or after bacterial infection. In addition, synergistic and/or additive
interactions between different AMPs and other antimicrobial compounds of the tear
film have been described. These effects may compensate the low AMP concentra-
tion in tears (McDermott 2013). Animal models of bacterial keratitis have demon-
strated that CRAMP (mouse homolog of LL-37) and mouse beta-defensins 2 and
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 27
Herpes simplex virus type 1 (HSV-1) is the most common cause of viral keratitis
(Fig. 2.3) and also the most common cause of irreversible cornea-derived blindness
in developed nations (Karsten et al. 2012). Worldwide, an estimated ten million
persons suffer from HSV keratitis, with about two million individuals left with
impaired vision (Rowe et al. 2013). The incidence lies between 5.9 and 20.7/105 of
the population per year and with a prevalence of 149/105 in the developed countries
(for review, see Kaye and Choudhary 2006). The initial (not necessarily primary)
sites of herpetic eye involvement usually manifest as a blepharitis (infection of the
lid rim), conjunctivitis, or corneal epithelial keratitis. More often, a younger age
group is infected and tends to be more severely infected, especially in the develop-
ing world, where malnutrition and several diseases as well as the lack of access to
treatment may be present.
As already mentioned, defensins are beside LL-37 the most abundant group of
AMPs in human. Their antiviral activity already was defined in the mid-1980s.
Studies of AMPs interacting with HSV-1 revealed direct inactivation of the virus by
HNP1 produced by neutrophils (Daher et al. 1986). Since that time, the antiviral
activity of various AMPs against HSV infection has been followed up by an
With regard to other frequent viral ocular surface infections such as adenovirus or
vaccinia virus infection and in addition to other viruses, no data exist in a context
with AMPs.
Here, the reader is referred to a very recent review by Curtis R. Brandt (Brandt
2014). This review presents an overview about antimicrobial drugs available for the
treatment of ocular surface infections. In that review, it is also discussed that given
the nature of peptides, topical applications are the most likely use to be successful
for treating keratitis. Such peptides would be effective against drug-resistant patho-
gens and might act synergistically if used in combination therapy. Although hun-
dreds of peptides with antimicrobial properties have been isolated or synthesized,
only a handful have been tested so far against ocular pathogens and even fewer have
been tested in animal models. The review summarizes the currently available infor-
mation on the use of these peptides to treat keratitis, outlines some of the problems
that have been identified, and discusses future studies that will be needed. As out-
lined in the review, most of the peptides that have been tested so far have shown
activity at concentrations that do not warrant further development, but nevertheless,
there are promising candidates such as, for example, RC2, as mentioned above, or
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 29
the coupling of acyclovir (ACV) to the 3-OS heparan sulfate (a co-receptor for
HSV-1 glycoprotein) binding G2 (a M13 phage peptide) raising the possibility that
peptides and peptide-drug conjugates can be developed to treat viral keratitis.
A further strategy to prevent microbial keratitis is the attachment of modified AMPs
to contact lenses or contact lens cases. Contact lens-mediated microbial keratitis, in
particular by P. aeruginosa, Fusarium spp., and Acanthamoeba (an ocular parasite), is
associated with microbial adherence and biofilm formation. In the last decade, various
antimicrobial strategies, such as cationic metals and peptides, selenium, quorum-sens-
ing inhibitors, and various biocidal and non-biocidal agents, have been developed and
tested with promising results in various animal models (Dutta and Willcox 2014).
Covalent binding of cationic selenium to contact lenses revealed inhibition of bacterial
colonization as well as reduction of acute red eye formation and bacterial ulceration in
a rabbit model (Mathews et al. 2006). Fimbrolide-coated lenses (a quorum-sensing
inhibitor) reduced bacterial and Acanthamoeba adhesion without negative ocular
responses in a 1-month animal model and in an overnight human trial (Zhu et al. 2008).
Moreover, contact lenses coated with melamine, a synthetic cationic hybrid AMP pre-
pared by combining active regions of the AMPs protamine (from salmon sperm) and
melittin (from bee venom), reduced adhesion to contact lenses and prevented coloniza-
tion against a broad spectrum of ocular pathogens (Dutta et al. 2013). In addition,
melamine-coated contact lenses showed reduced ocular inflammation and corneal
infiltrates and less epithelial defects in some animal keratitis models (Cole et al. 2010).
Acknowledgment We would like to thank Jörg Pekarsky, Department of Anatomy II, FAU
Erlangen, Germany, for drawing Figures 2.1 + 2.2 and Gerd Geerling, Director and Head,
Department of Ophthalmology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany,
for providing us with Figure 2.3. Supported by Deutsche Forschungsgemeinschaft (DFG) grant
PA738/9-2.
References
Daher KA, Selsted ME, Lehrer RI (1986) Direct inactivation of viruses by human granulocyte
defensins. J Virol 60:1068–1074
Di Luca M, Maccari G, Nifosi R (2014) Treatment of microbial biofilms in the post-antibiotic era:
prophylactic and therapeutic use of antimicrobial peptides and their design by bioinformatics
tools. Pathog Dis 70:257–270
Diler E, Schicht M, Rabung A et al (2014) The novel surfactant protein SP-H enhances the phago-
cytosis efficiency of macrophage-like cell lines U937 and MH-S. BMC Res Notes 7:851
Domachowske JB, Bonville CA, Dyer KD et al (1998) Evolution of antiviral activity in the ribo-
nuclease A gene superfamily: evidence for a specific interaction between eosinophil-derived
neurotoxin (EDN/RNase 2) and respiratory syncytial virus. Nucleic Acids Res 26:5327–5332
Domingues MM, Lopes SC, Santos NC et al (2009) Fold-unfold transitions in the selectivity and
mechanism of action of the N-terminal fragment of the bactericidal/permeability-increasing
protein (rBPI(21)). Biophys J 96:987–996
Dutta D, Willcox MD (2014) Antimicrobial contact lenses and lens cases: a review. Eye Contact
Lens 40:312–324
Dutta D, Cole N, Kumar N et al (2013) Broad spectrum antimicrobial activity of melimine cova-
lently bound to contact lenses. Invest Ophthalmol Vis Sci 54:175–182
Garreis F, Schlorf T, Worlitzsch D et al (2010) Roles of human beta-defensins in innate immune
defense at the ocular surface: arming and alarming corneal and conjunctival epithelial cells.
Histochem Cell Biol 134:59–73
Garreis F, Gottschalt M, Schlorf T et al (2011) Expression and regulation of antimicrobial peptide
psoriasin (S100A7) at the ocular surface and in the lacrimal apparatus. Invest Ophthalmol Vis
Sci 52:4914–4922
Gayton JL (2009) Etiology, prevalence, and treatment of dry eye disease. Clin Ophthalmol 3:405–412
Groot F, Sanders RW, Ter Brake O et al (2006) Histatin 5-derived peptide with improved fungicidal
properties enhances human immunodeficiency virus type 1 replication by promoting viral
entry. J Virol 80:9236–9243
Gwyer Findlay E, Currie SM, Davidson DJ (2013) Cationic host defence peptides: potential as
antiviral therapeutics. BioDrugs Clin Immunother Biopharm Gene Ther 27:479–493
Harrod KS, Trapnell BC, Otake K et al (1999) SP-A enhances viral clearance and inhibits inflam-
mation after pulmonary adenoviral infection. Am J Physiol 277:L580–L588
Hazlett L, Wu M (2011) Defensins in innate immunity. Cell Tissue Res 343:175–188
Hazrati E, Galen B, Lu W et al (2006) Human alpha- and beta-defensins block multiple steps in
herpes simplex virus infection. J Immunol 177:8658–8666
Huang LC, Reins RY, Gallo RL et al (2007) Cathelicidin-deficient (Cnlp -/-) mice show increased
susceptibility to Pseudomonas aeruginosa keratitis. Invest Ophthalmol Vis Sci 48:4498–4508
Jang SA, Kim H, Lee JY et al (2012) Mechanism of action and specificity of antimicrobial pep-
tides designed based on buforin IIb. Peptides 34:283–289
Karsten E, Watson SL, Foster LJ (2012) Diversity of microbial species implicated in keratitis: a
review. Open ophthalmol J 6:110–124
Kaye S, Choudhary A (2006) Herpes simplex keratitis. Prog Retin Eye Res 25:355–380
Kishore U, Greenhough TJ, Waters P et al (2006) Surfactant proteins SP-A and SP-D: structure,
function and receptors. Mol Immunol 43:1293–1315
Klotman ME, Chang TL (2006) Defensins in innate antiviral immunity. Nat Rev Immunol 6:
447–456
Kolar SS, Mcdermott AM (2011) Role of host-defence peptides in eye diseases. Cell Mol Life Sci
CMLS 68:2201–2213
Kragol G, Lovas S, Varadi G et al (2001) The antibacterial peptide pyrrhocoricin inhibits the
ATPase actions of DnaK and prevents chaperone-assisted protein folding. Biochemistry 40:
3016–3026
Lai Y, Gallo RL (2009) AMPed up immunity: how antimicrobial peptides have multiple roles in
immune defense. Trends Immunol 30:131–141
Lawson WE, Crossno PF, Polosukhin VV et al (2008) Endoplasmic reticulum stress in alveolar
epithelial cells is prominent in IPF: association with altered surfactant protein processing and
herpesvirus infection. Am J Physiol Lung Cell Mol Physiol 294:L1119–L1126
2 Antimicrobial Peptides as Endogenous Antibacterials and Antivirals 31
Lehrer RI, Cole AM, Selsted ME (2012) theta-Defensins: cyclic peptides with endless potential.
J Biol Chem 287:27014–27019
Levine AM, Whitsett JA, Hartshorn KL et al (2001) Surfactant protein D enhances clearance of
influenza A virus from the lung in vivo. J Immunol 167:5868–5873
Lobo DS, Pereira IB, Fragel-Madeira L et al (2007) Antifungal Pisum sativum defensin 1 interacts
with Neurospora crassa cyclin F related to the cell cycle. Biochemistry 46:987–996
Ma G, Greenwell-Wild T, Lei K et al (2004) Secretory leukocyte protease inhibitor binds to
annexin II, a cofactor for macrophage HIV-1 infection. J Exp Med 200:1337–1346
Mathews SM, Spallholz JE, Grimson MJ et al (2006) Prevention of bacterial colonization of con-
tact lenses with covalently attached selenium and effects on the rabbit cornea. Cornea 25:
806–814
McDermott AM (2013) Antimicrobial compounds in tears. Exp Eye Res 117:53–61
Mohammed I, Abedin A, Tsintzas K et al (2011) Increased expression of hepcidin and toll-like
receptors 8 and 10 in viral keratitis. Cornea 30:899–904
Mun J, Tam C, Chan G et al (2013) MicroRNA-762 is upregulated in human corneal epithelial
cells in response to tear fluid and Pseudomonas aeruginosa antigens and negatively regulates
the expression of host defense genes encoding RNase7 and ST2. PLoS One 8, e57850
Otri AM, Mohammed I, Al-Aqaba MA et al (2012) Variable expression of human Beta defensins 3
and 9 at the human ocular surface in infectious keratitis. Invest Ophthalmol Vis Sci 53:757–761
Paulsen FP, Berry MS (2006) Mucins and TFF peptides of the tear film and lacrimal apparatus.
Prog Histochem Cytochem 41:1–53
Peuravuori H, Aho VV, Aho HJ et al (2006) Bactericidal/permeability-increasing protein in lacri-
mal gland and in tears of healthy subjects. Graefe’s archive for clinical and experimental oph-
thalmology = Albrecht von Graefes Archiv fur klinische und experimentelle. Ophthalmologie
244:143–148
Pillay K, Coutsoudis A, Agadzi-Naqvi AK et al (2001) Secretory leukocyte protease inhibitor in
vaginal fluids and perinatal human immunodeficiency virus type 1 transmission. J Infect Dis
183:653–656
Qu XD, Lehrer RI (1998) Secretory phospholipase A2 is the principal bactericide for staphylo-
cocci and other gram-positive bacteria in human tears. Infect Immun 66:2791–2797
Rowe AM, St Leger AJ, Jeon S et al (2013) Herpes keratitis. Prog Retin Eye Res 32:88–101
Sack RA, Conradi L, Krumholz D et al (2005) Membrane array characterization of 80 chemokines,
cytokines, and growth factors in open- and closed-eye tears: angiogenin and other defense
system constituents. Invest Ophthalmol Vis Sci 46:1228–1238
Sallenave JM (2010) Secretory leukocyte protease inhibitor and elafin/trappin-2: versatile mucosal
antimicrobials and regulators of immunity. Am J Respir Cell Mol Biol 42:635–643
Sathe S, Sakata M, Beaton AR et al (1998) Identification, origins and the diurnal role of the prin-
cipal serine protease inhibitors in human tear fluid. Curr Eye Res 17:348–362
Schicht M (2012) Humane Surfactant Proteine: Detektion und Charakterisierung. Halle, Univ.,
Naturwissenschaftliche Fakultät I, Diss
Schicht M, Posa A, Paulsen F et al (2010) The ocular surfactant system and its relevance in the dry
eye. Klin Monbl Augenheilkd 227:864–870
Schroeder BO, Wu Z, Nuding S et al (2011) Reduction of disulphide bonds unmasks potent anti-
microbial activity of human beta-defensin 1. Nature 469:419–423
Semple F, Dorin JR (2012) beta-Defensins: multifunctional modulators of infection, inflammation
and more? J Innate Immun 4:337–348
Simanski M, Koten B, Schroder JM et al (2012) Antimicrobial RNases in cutaneous defense.
J Innate Immun 4:241–247
Stern ME, Beuerman RW, Fox RI et al (1998) The pathology of dry eye: the interaction between
the ocular surface and lacrimal glands. Cornea 17:584–589
Tiffany JM (2008) The normal tear film. Dev Ophthalmol 41:1–20
Tran D, Tran P, Roberts K et al (2008) Microbicidal properties and cytocidal selectivity of rhesus
macaque theta defensins. Antimicrob Agents Chemother 52:944–953
Tseng CC, Tseng CP (2000) Identification of a novel secretory leukocyte protease inhibitor-
binding protein involved in membrane phospholipid movement. FEBS Lett 475:232–236
32 F. Garreis et al.
Abstract The innate immune system of the lung is a complex network of different
cellular and noncellular components protecting the lung from inhaled pathogens.
Antimicrobial peptides (AMP) are produced by epithelial and myeloid cells as part
of this system. AMPs, such as defensins and cathelicidin, are small cationic peptides
with a broad microbicidal activity against respiratory bacteria, viruses, and fungi.
However, their functions go beyond antimicrobial activity and include modulation
of the innate and adaptive immune response to infection as well as lung repair after
injury. Thus, AMPs are involved in pathophysiological processes of many lung dis-
eases, such as acute and chronic lung infection, chronic obstructive pulmonary dis-
ease, cystic fibrosis, and lung cancer.
Its function within the innate immune system exceeds well-known mechanisms
like barrier formation and mucociliary clearance. The epithelium of the respiratory
tract is an immunologically active tissue, which exerts functions such as pathogen
recognition, pathogen neutralization, and activation of further immune mecha-
nisms. These include resident alveolar macrophages as well as inflowing neutro-
phils. Those professional immune cells have a high capability to neutralize
microbial pathogens. In addition, the release of immunomodulatory mediators
from epithelial and myeloid cells induces activation of the adaptive immune
response.
Small cationic antimicrobial peptides (AMPs) from the families of defensins and
cathelicidins play a key role within the innate immune network of the lung.
AMPs are expressed by various cell types present in the lung during health and
disease. The main AMPs expressed in the lung are α-defensins, β-defensins, and
cathelicidins. Table 3.1 gives an overview on AMPs of the human lung.
3.2.1 Defensins
3.2.1.1 α-Defensins
α-Defensins comprise the human neutrophil peptides (HNP) 1–4 and the epithelial
α-defensins HD-5 and HD-6 (Doss et al. 2010). Different to other epithelial tissues
of the human body, the airway epithelium hardly produces α-defensins; only HD-5
has been detected in airway epithelial cells in small amounts (Frye et al. 2000). The
main sources of α-defensins in the human lung are neutrophilic granulocytes. They
constitutively express α-defensins, which are stored in azurophilic granula and rep-
resent their main protein content. Upon infection, activation of innate immunity
triggers the influx of neutrophils into the lung, where they neutralize pathogens
through phagocytosis and degranulation of their azurophilic granula (Ganz et al.
1985; Selsted and Ouellette 2005).
In human alveolar macrophages, α-defensins are virtually absent; only rabbits
have been found to express α-defensins in their alveolar macrophages (Ganz et al.
1985; Selsted and Ouellette 2005).
3.2.1.2 β-Defensins
The principal β-defensins found in the human lung are human β-defensin (hBD) 1–4
(Doss et al. 2010). Their main sources are epithelial cells and submucous glands
(Kao et al. 2003; Bals et al. 1998a). However, β-defensins (hBD-1 and hBD-2) are
also released from myeloid cells such as alveolar macrophages and dendritic cells
(Duits et al. 2002). The expression of β-defensins shows a gene-specific behavior:
hBD-1 is constitutively expressed by the epithelium, ensuring a constant basic anti-
microbial activity of the airway surface liquid (McCray and Bentley 1997; Singh
et al. 1998). In contrast, hBD-2, hBD-3, and hBD-4 are not released by default, but
only if required; high levels can only be detected in the case of infection or inflam-
mation (Hess et al. 2010; Harder et al. 2001; Yanagi et al. 2005; Scharf et al. 2010a,
2012; Benincasa et al. 2009). Compared to hBD-3 and hBD-4, which are predomi-
nantly expressed in other epithelial tissues, hBD-2 is mainly expressed in the respi-
ratory tract (Bals et al. 1998a).
3.2.2 Cathelicidin
3.3.1 Defensins
2007). hBD-2 expression is also stimulated by IL-17A (Kao et al. 2004), which is
derived from Th17 cells during lung infection (Ye et al. 2001), and airway epithelial
cell-derived IL-17C (Kusagaya et al. 2014). Upon viral infection, interferon-gamma
release is triggered, for example, by TLR3 (Akira 2009) and leads to increased
hBD-3 expression (Albanesi et al. 2007).
Stimulation of TLRs and cytokine receptors leads to activation of intracellular
signaling processes like mentioned above. Therefore, TLR and cytokine receptors
induce β-defensin expression in an NF-κB- or MAP kinase-dependent manner
(McCray and Bentley 1997; Scharf et al. 2010b). In fact, hBD-2 has been shown to
be a direct target gene of the transcription factors NF-κB and the MAP kinase target
AP-1 (Wehkamp et al. 2004; O’Neil et al. 1999). With a vitamin D response element
within its promoter sequence, hBD-2 further is a target gene of vitamin D; however,
vitamin D reactivity of the hBD-2 gene is not as strong as it is described for cathe-
licidin (Wang et al. 2004).
3.3.2 Cathelicidin
Within the innate immune network of the lung, AMPs are one of the first defense
mechanisms to face invading pathogens. Apart from their antimicrobial activity,
AMPs have many immunomodulatory features that are similar to those of classic
cytokines. For example, AMPs act as chemoattractants for different immune cells,
recruiting them to sites of inflammation. α-Defensins have been shown as chemo-
tactic factors for monocytes, dendritic cells, and T cells (Yang et al. 2000a, 2001;
Chertov et al. 1996). β-Defensins chemoattract monocytes, macrophages, and neu-
trophils via the chemokine receptor CCR2, immature dendritic cells and T cells via
CCR6, and mast cells in a phospholipase C-dependent manner (Soruri et al. 2007;
Yang et al. 1999; Röhrl et al. 2010; Niyonsaba 2002). Finally, cathelicidin is capable
of recruiting neutrophils, monocytes, and T cells via FPRL1 (Yang et al. 2000b;
Kurosaka et al. 2005), and mast cells via phospholipase C (Niyonsaba et al. 2002),
and takes part in the regulation of dendritic cell maturation and differentiation
(Davidson et al. 2004; Kandler et al. 2006).
In addition to their chemokine nature, AMPs are able to act as endogenous TLR
agonists. For example, mouse β-defensin 2 activates TLR4 (Biragyn et al. 2002),
and β-defensin 3 is capable of activating TLR1 and TLR2 (Funderburg et al. 2007).
By that, AMPs can induce cytokine expression and activation of immune cells:
β-defensins induce IL-6, IL-8, IL-10, and MCP-1 in monocytes (Boniotto et al.
2006) and IL-1β and IL-6 in dendritic cells (Biragyn et al. 2002). In myeloid and
epithelial cells, IL-8 expression is upregulated by α-defensins (Khine et al. 2006).
β-Defensin 2 leads to dendritic cell maturation in mice (Biragyn et al. 2002), and
β-defensin 3 activates monocytes and neutrophils (Funderburg et al. 2007).
Cathelicidin has been shown to indirectly enhance TLR signaling (Lai et al. 2011).
Cathelicidin further induces IL-8 and MCP-1 in a MAPK-dependent manner (Scott
et al. 2002; Tjabringa et al. 2003; Bowdish et al. 2004; Yu et al. 2007). Mast cell
degranulation and histamine release are mediated by β-defensins and cathelicidin
(Schiemann et al. 2009; Niyonsaba et al. 2001).
Excessive activity of the immune system can cause self-damage of the host by
immunopathological processes. Therefore, balancing of pro- and anti-inflammatory
signals is crucial. Above immunostimulation, AMPs also take part in the fine-tuning
of the immune response by additionally regulating anti-inflammatory mechanisms.
Activation of the complement system, which can be induced in the lung (Varsano
et al. 2000), is inhibited by β-defensin 2 (Bhat et al. 2007). β-Defensin 3 has been
shown to inhibit ERK and, via downmodulation of TRIF and MyD88, TLR signal-
ing, leading to a decreased production of proinflammatory cytokines in macrophages
and dendritic cells (Semple et al. 2010, 2011; Pingel et al. 2008). α-Defensins have
an anti-inflammatory effect on macrophages, reducing production of reactive oxy-
gen species and inhibiting the expression of proinflammatory cytokines (Miles et al.
2009). However, compared to PAM3CSK4, another TLR1/2 agonist, β-defensin
3-derived TLR1/2 activation induces a pro- rather than an anti-inflammatory cyto-
kine pattern in monocytes (Funderburg et al. 2011).
40 F. Seiler et al.
pro- and anti-inflammatory cytokines, the keratinocyte growth factor (KGF), and
the epidermal growth factor (EGF) (Tjabringa et al. 2003; Campbell et al. 2011;
Puddicombe et al. 2000; Michelson et al. 1999). AMPs qualify as mediators of
wound healing and tissue repair as the expression of AMPs by inflammatory and
epithelial cells is increased under infectious and inflammatory conditions and epi-
thelial injury induced by infectious agents leads to an increased risk that pathogens
invade the host (Steinstraesser et al. 2008; Lai and Gallo 2009). Indeed, there is
strong evidence that AMPs are involved in epithelial repair processes, such as cell
proliferation, epithelial regeneration, and reepithelialization (Lai and Gallo 2009;
Heilborn et al. 2003; Tokumaru et al. 2005; Carretero et al. 2008; Beisswenger and
Bals 2005; Murphy et al. 1993; Aarbiou et al. 2004).
The function of AMPs in the repair of epithelial injury has been particularly stud-
ied in wound healing of the skin. Cathelicidin, for instance, induces reepithelializa-
tion after skin injury via transactivation of the EGF receptor (Heilborn et al. 2003;
Tokumaru et al. 2005; Carretero et al. 2008). Cathelicidin also plays an important role
in angiogenesis and thereby contributes to wound repair (Koczulla et al. 2003). As
cathelicidins and defensins induce proliferation of diverse airway epithelial cell lines
and primary airway epithelial cells in vitro, it is suggested that AMPs also mediate
wound healing in the lung via the activation of pro-proliferative signaling cascades
(Beisswenger and Bals 2005; Murphy et al. 1993). Cathelicidin activates airway epi-
thelial cell via metalloproteinase-mediated cleavage of membrane-anchored EGF
receptor ligands as well as MAP kinase-dependent signaling pathways (Tjabringa
et al. 2003) and stimulates airway epithelial wound closure in an EGF receptor-, G
protein-coupled receptor-, and MAP kinase-dependent manner (Shaykhiev et al.
2005). A function of cathelicidins in tissue repair is further supported by a mouse
study which associates deficiency of the mouse cathelicidin CRAMP with increased
lung injury during infection with gram-negative bacteria (Kovach et al. 2012).
Like cathelicidin, α-defensins from human neutrophils induce proliferation of
airway epithelial cell lines via activation of MAP kinase signaling cascades (Aarbiou
et al. 2002) and enhance wound closure in an EGF-dependent manner (Aarbiou
et al. 2004). α-Defensins also increase proliferation of lung fibroblasts and collagen
synthesis (Han et al. 2009).
However, in vitro studies also suggest that cathelicidins and defensins induce the
release of inflammatory mediators by airway epithelial cells and that AMPs are
cytotoxic at higher concentrations (Tjabringa et al. 2003; Shaykhiev et al. 2005;
Sakamoto et al. 2005). Therefore, the proinflammatory, cytotoxic, and pro-
proliferative functions of AMPs may also have adverse consequences by perpetuating
inflammation and potentially contribute to epithelial injury in lung diseases
(Shaykhiev et al. 2005; Sakamoto et al. 2005). Moreover, concentrations of AMPs
have been found to be increased in severe lung diseases. Plasma concentrations of
α-defensins, for instance, are increased in pulmonary fibrosis (Mukae et al. 2002)
and β-defensins are increased in bronchoalveolar lavage fluids of patients with
diffuse panbronchiolitis and in bronchiolitis obliterans syndrome after lung
transplantation (Ross et al. 2004; Hiratsuka et al. 2003). Cathelicidin associates
with bronchial inflammation in cystic fibrosis lung disease (Chen et al. 2004).
42 F. Seiler et al.
Respiratory tract infections are very common. Especially pneumonia is a major cause
of global morbidity and mortality in children as well as in adults (Klugman and Garau
2009). Due to their microbicidal properties (“endogenous antibiotics”), the role of
AMPs in acute respiratory tract infection has drawn much attention. AMP levels are
increased in patients with pneumonia. High levels of β-defensins and cathelicidin can
be detected in plasma or sputum of patient with acute pneumonia (Hiratsuka et al.
1998; Herr et al. 2009; Schaller-Bals et al. 2002; Ishimoto et al. 2006).
The important role of AMPs in respiratory tract infection is especially underlined
by studies in animal models. Cathelicidin treatment has been shown to reduce pro-
inflammatory cytokine release during MRSA pneumonia and improves the histo-
pathological outcome (Hou et al. 2013). In cathelicidin-deficient mice, the immune
response to gram-negative pneumonia with K. pneumoniae is delayed, eventually
resulting in more inflammation and increased lung injury (Kovach et al. 2012).
During gram-negative pneumonia with P. aeruginosa, overexpression of β-defensin
2 in rats reduces bacterial load in the lung as well as inflammation and lung injury
and increases survival rate (Shu et al. 2006; Hu et al. 2010). In β-defensin 1-defi-
cient mice, clearance of H. influenzae in the lung is impaired (Moser et al. 2002).
The effect of AMPs is also modulated by host-microbe interactions. The S. aureus
virulence factor staphylokinase acts synergistically with cathelicidin to promote
fibrinolysis during mouse pneumonia, which leads to a more invasive infection (Braff
et al. 2007). Staphylokinase is also able to inactivate α-defensins (Jin et al. 2004).
While there are natural limitations in evaluation of the in vivo impact of AMPs in
human pneumonia, the high AMP levels found in patients with pneumonia together
with the studies mentioned above suggest a crucial role of AMPs in the response to
lung infection.
Cigarette smoke is the major risk factor for the development of the chronic obstruc-
tive pulmonary disease (COPD). COPD is characterized by chronic inflammation of
the lung leading to tissue destruction, emphysema, and loss of pulmonary function
(Sethi and Murphy 2008; Sethi 2010). Stable COPD patients are frequently colo-
nized with bacterial pathogens (e.g., H. influenzae); recurrent infections of the
3 Function of Antimicrobial Peptides in Lung Innate Immunity 43
respiratory tract and chronic bronchitis are common in COPD (Sethi and Murphy
2008; Sethi 2010; Moghaddam et al. 2011; Garmendia et al. 2012). Microbial infec-
tions of the lung evoke harmful inflammatory responses that contribute to clinical
deterioration of COPD patients (Sethi 2010). Even in healthy individuals, smoking
suppresses pulmonary host defense making the lung more susceptible for microbial
colonization and infection (Herr et al. 2009; Sethi 2010; Mehta et al. 2008). Ex vivo
and in vitro studies showed that the expression of AMPs is inhibited by cigarette
smoke and that COPD is associated with skewed AMP expression in the respiratory
tract. Analyses of human samples showed that current or former smoking is associ-
ated with significantly reduced levels of the β-defensin hBD-2 in pharyngeal wash-
ing fluids and sputum from patients with acute pneumonia (Herr et al. 2009). In
vitro studies demonstrated that exposure to cigarette smoke and reactive oxygen
species inhibit the expression of hBD-2 in respiratory epithelial cells in response to
inflammatory mediators or bacteria (Herr et al. 2009). The effects of cigarette smoke
on β-defensin expression are likely due to the suppression of cellular signaling cas-
cades by cigarette smoke, such as NF-κB and AP-1 signaling (Pace et al. 2012;
Kulkarni et al. 2010; Laan et al. 2004). In currently smoking COPD patients, hBD-2
expression is decreased in central but not in distal airways, inversely correlating
with cigarette smoke exposure (Pace et al. 2012). This has been underlined by the
finding that hBD-2 mRNA expression in peripheral lung samples has been found to
be increased in COPD patients and was associated with higher IL-8 expression and
poor lung function (Liao et al. 2012). Cathelicidin has also been shown to be
increased in small airways of COPD patients, where it may contribute to airway
remodeling (Sun et al. 2014). In addition, a proteomic study showed that the
α-defensins are increased in bronchoalveolar lavage fluids of COPD patients
(Merkel et al. 2005). In summary, the expression pattern of AMP seems to be
deranged in COPD with low antimicrobial activity in the central airways. Altered
AMP expression may lead to airway colonization with pathogens in smokers and
patient with COPD, providing reservoirs for recurrent infection. Enhanced suscep-
tibility to recurrent respiratory tract infections subsequently leads to chronic inflam-
mation, aberrant expression of AMPs, and disease progression (Herr et al. 2009;
Sethi 2010).
Studies also suggest that skewed expression of AMPs and inhibition of the bacte-
ricidal activity of AMPs play a role in cystic fibrosis (CF). AMPs are detectable in
sputum or bronchoalveolar lavage fluid of CF patients with α-defensins and catheli-
cidin being present at high levels (Felgentreff et al. 2006; Chen et al. 2004; Soong
et al. 1997). Moreover, enhanced levels of cathelicidin correlate with severity of CF
lung disease (Chen et al. 2004). β-Defensin levels do not seem to correlate with the
severity of inflammation, but decreased levels of hBD-2 are associated with severity
of CF (Chen et al. 2004; Dauletbaev et al. 2002; Bals et al. 2001). In vitro, the bac-
tericidal activity of cationic AMPs is salt sensitive. Thus, it is suggested that increased
salt concentrations in airway fluids of CF patients contribute to chronic lung infec-
tion by inhibiting bactericidal functions of AMPs (Bals et al. 1998a). In line with this
hypothesis, bactericidal activity of hBD-1 has been shown to be decreased in air-
ways of CF patients and CF airway fluids fail to kill S. aureus and P. aeruginosa
(Goldman et al. 1997; Bals et al. 1999). However, in a human bronchial xenograft
44 F. Seiler et al.
For several tumor types such as ovarian cancer and malignant melanoma, adverse
effects of AMPs on tumor development and progression have been shown. In lung
cancer, overexpression of different AMPs was found. Cathelicidin is overexpressed
in up to 25 % of NSCLC samples (Von Haussen et al. 2008). It is also released by
myeloid cells in the tumor microenvironment (Li et al. 2014). Cathelicidin has been
shown to transactivate EGFR in airway epithelial cells (Tjabringa et al. 2003).
EGFR signaling is one of the critical pathways in lung carcinogenesis (Mitsudomi
and Yatabe 2010). Through interaction with the EGFR pathway, cathelicidin pro-
motes tumor growth in vitro and in vivo (Von Haussen et al. 2008), which is further
induced by cigarette smoke (Li et al. 2014).
As for β-defensins, hBD-1 and hBD-2 are overexpressed in about 50–60 % of
NSCLC tissue samples (Shestakova et al. 2008). In serum of patients with lung
cancer, concentrations of hBD-1 and hBD-2 are significantly higher as compared to
healthy controls and even patients with pneumonia. Therefore, hBD-1 and hBD-2
have even been proposed as lung tumor markers (Arimura et al. 2004).
AMPs take part in the orchestration of important cellular functions that contrib-
ute to oncogenesis and tumor promotion, as they modulate proliferation, cell
migration, and angiogenesis. Thus, given their central role in chronic lung disease,
AMPs might link chronic airway inflammation to the development of lung cancer.
In contrast to cathelicidin and β-defensins, the α-defensin HNP-1 has been shown
to act as an inhibitor of tumor growth. HNP-1 is cytotoxic to tumor cells (Van
Wetering et al. 1997; Okrent et al. 1990) and induces apoptosis in lung cancer and
other cells (Xu et al. 2008; Lichtenstein 1991). This even led to successful gene
therapy approaches with direct intratumoral administration of plasmid DNA encod-
ing HNP-1 in a murine xenograft tumor model (Xu et al. 2008).
3.7 Conclusions
In the human lung, AMPs are abundantly expressed by different cell types, includ-
ing epithelial and immune cells. The expression of defensins is largely triggered
upon stimulation with PAMPs and inflammatory mediators, whereas cathelicidin
3 Function of Antimicrobial Peptides in Lung Innate Immunity 45
References
Aarbiou J, Ertmann M, van Wetering S et al (2002) Human neutrophil defensins induce lung epi-
thelial cell proliferation in vitro. J Leukoc Biol 72:167–174
Aarbiou J, Verhoosel RM, Van Wetering S et al (2004) Neutrophil defensins enhance lung epithe-
lial wound closure and mucin gene expression in vitro. Am J Respir Cell Mol Biol 30:
193–201
Aerts AM, François IEJA, Cammue BPA, Thevissen K (2008) The mode of antifungal action of
plant, insect and human defensins. Cell Mol Life Sci 65:2069–2079
Agerberth B, Grunewald J, Castaños-Velez E et al (1999) Antibacterial components in bronchoal-
veolar lavage fluid from healthy individuals and sarcoidosis patients. Am J Respir Crit Care
Med 160:283–290
Akira S (2009) Pathogen recognition by innate immunity and its signaling. Proc Jpn Acad Ser B
Phys Biol Sci 85:143–156
Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124:
783–801
Alalwani SM, Sierigk J, Herr C et al (2010) The antimicrobial peptide LL-37 modulates the
inflammatory and host defense response of human neutrophils. Eur J Immunol 40:1118–1126
Albanesi C, Fairchild HR, Madonna S et al (2007) IL-4 and IL-13 negatively regulate TNF-alpha-
and IFN-gamma-induced beta-defensin expression through STAT-6, suppressor of cytokine
signaling (SOCS)-1, and SOCS-3. J Immunol 179:984–992
Alekseeva L, Huet D, Féménia F et al (2009) Inducible expression of beta defensins by human
respiratory epithelial cells exposed to Aspergillus fumigatus organisms. BMC Microbiol 9:33
Arimura Y, Ashitani J, Yanagi S et al (2004) Elevated serum beta-defensins concentrations in
patients with lung cancer. Anticancer Res 24:4051–4057
Bals R, Wang X, Wu Z et al (1998a) Human beta-defensin 2 is a salt-sensitive peptide antibiotic
expressed in human lung. J Clin Invest 102:874–880
Bals R, Wang X, Zasloff M, Wilson JM (1998b) The peptide antibiotic LL-37/hCAP-18 is
expressed in epithelia of the human lung where it has broad antimicrobial activity at the airway
surface. Proc Natl Acad Sci U S A 95:9541–9546
Bals R, Weiner DJ, Meegalla RL, Wilson JM (1999) Transfer of a cathelicidin peptide antibiotic
gene restores bacterial killing in a cystic fibrosis xenograft model. J Clin Invest 103:
1113–1117
46 F. Seiler et al.
Hou M, Zhang N, Yang J et al (2013) Antimicrobial peptide LL-37 and IDR-1 ameliorate MRSA
pneumonia in vivo. Cell Physiol Biochem 32:614–623
Hu Q, Zuo P, Shao B et al (2010) Administration of nonviral gene vector encoding rat beta-
defensin-2 ameliorates chronic Pseudomonas aeruginosa lung infection in rats. J Gene Med
12:276–286
Ishimoto H, Mukae H, Date Y et al (2006) Identification of hBD-3 in respiratory tract and serum:
the increase in pneumonia. Eur Respir J 27:253–260
Iwase T, Uehara Y, Shinji H et al (2010) Staphylococcus epidermidis Esp inhibits Staphylococcus
aureus biofilm formation and nasal colonization. Nature 465:346–349
Jin T, Bokarewa M, Foster T et al (2004) Staphylococcus aureus resists human defensins by pro-
duction of staphylokinase, a novel bacterial evasion mechanism. J Immunol 172:1169–1176
Kandler K, Shaykhiev R, Kleemann P et al (2006) The anti-microbial peptide LL-37 inhibits the
activation of dendritic cells by TLR ligands. Int Immunol 18:1729–1736
Kao CY, Chen Y, Zhao YH, Wu R (2003) ORFeome-based search of airway epithelial cell-specific
novel human [beta]-defensin genes. Am J Respir Cell Mol Biol 29:71–80
Kao C-Y, Chen Y, Thai P et al (2004) IL-17 markedly up-regulates beta-defensin-2 expression in
human airway epithelium via JAK and NF-kappaB signaling pathways. J Immunol 173:
3482–3491
Kawasaki T, Kawai T (2014) Toll-like receptor signaling pathways. Front Immunol 5:464
Khine AA, Del Sorbo L, Vaschetto R et al (2006) Human neutrophil peptides induce interleukin-8
production through the P2Y6 signaling pathway. Blood 107:2936–2942
Kirikae T, Hirata M, Yamasu H et al (1998) Protective effects of a human 18-kilodalton cationic
antimicrobial protein (CAP18)-derived peptide against murine endotoxemia. Infect Immun
66:1861–1868
Klotman ME, Chang TL (2006) Defensins in innate antiviral immunity. Nat Rev Immunol 6:
447–456
Klugman KP, Garau J (2009) A preventable killer: pneumonia. Clin Microbiol Infect 15:989–990
Koczulla R, von Degenfeld G, Kupatt C et al (2003) An angiogenic role for the human peptide
antibiotic LL-37/hCAP-18. J Clin Invest 111:1665–1672
Koeffler HP, Reichel H, Bishop JE, Norman AW (1985) gamma-Interferon stimulates production
of 1,25-dihydroxyvitamin D3 by normal human macrophages. Biochem Biophys Res Commun
127:596–603
Kota S, Sabbah A, Chang TH et al (2008) Role of human beta-defensin-2 during tumor necrosis
factor-alpha/NF-kappaB-mediated innate antiviral response against human respiratory syncy-
tial virus. J Biol Chem 283:22417–22429
Kovach MA, Standiford TJ (2011) Toll like receptors in diseases of the lung. Int Immunopharmacol
11:1399–1406
Kovach MA, Ballinger MN, Newstead MW et al (2012) Cathelicidin-related antimicrobial peptide
is required for effective lung mucosal immunity in Gram-negative bacterial pneumonia.
J Immunol 189:304–311
Kulkarni R, Rampersaud R, Aguilar JL et al (2010) Cigarette smoke inhibits airway epithelial cell
innate immune responses to bacteria. Infect Immun 78:2146–2152
Kurosaka K, Chen Q, Yarovinsky F et al (2005) Mouse cathelin-related antimicrobial peptide
chemoattracts leukocytes using formyl peptide receptor-like 1/mouse formyl peptide receptor-
like 2 as the receptor and acts as an immune adjuvant. J Immunol 174:6257–6265
Kusagaya H, Fujisawa T, Yamanaka K et al (2014) Toll-like receptor-mediated airway IL-17C
enhances epithelial host defense in an autocrine/paracrine manner. Am J Respir Cell Mol Biol
50:30–39
Laan M, Bozinovski S, Anderson GP (2004) Cigarette smoke inhibits lipopolysaccharide-induced
production of inflammatory cytokines by suppressing the activation of activator protein-1 in
bronchial epithelial cells. J Immunol 173:4164–4170
Lai Y, Gallo RL (2009) AMPed up immunity: how antimicrobial peptides have multiple roles in
immune defense. Trends Immunol 30:131–141
Lai Y, Adhikarakunnathu S, Bhardwaj K et al (2011) LL37 and cationic peptides enhance TLR3
signaling by viral double-stranded RNAs. PLoS One 6, e26632
3 Function of Antimicrobial Peptides in Lung Innate Immunity 49
Pace E, Ferraro M, Minervini MI et al (2012) Beta defensin-2 is reduced in central but not in distal
airways of smoker COPD patients. PLoS One 7, e33601
Parker D, Prince A (2011) Innate immunity in the respiratory epithelium. Am J Respir Cell Mol
Biol 45:189–201
Pingel LC, Kohlgraf KG, Hansen CJ et al (2008) Human beta-defensin 3 binds to hemagglutinin
B (rHagB), a non-fimbrial adhesin from Porphyromonas gingivalis, and attenuates a pro-
inflammatory cytokine response. Immunol Cell Biol 86:643–649
Pistolic J, Cosseau C, Li Y et al (2009) Host defence peptide LL-37 induces IL-6 expression in
human bronchial epithelial cells by activation of the NF-kappaB signaling pathway. J Innate
Immun 1:254–267
Platz J, Beisswenger C, Dalpke A et al (2004) Microbial DNA induces a host defense reaction of
human respiratory epithelial cells. J Immunol 173:1219–1223
Proud D, Sanders SP, Wiehler S (2004) Human rhinovirus infection induces airway epithelial cell
production of human beta-defensin 2 both in vitro and in vivo. J Immunol 172:4637–4645
Puddicombe SM, Polosa R, Richter A et al (2000) Involvement of the epidermal growth factor
receptor in epithelial repair in asthma. FASEB J 14:1362–1374
Rivas-Santiago B, Schwander SK, Sarabia C et al (2005) Human {beta}-defensin 2 is expressed
and associated with Mycobacterium tuberculosis during infection of human alveolar epithelial
cells. Infect Immun 73:4505–4511
Rivas-Santiago B, Hernandez-Pando R, Carranza C et al (2008) Expression of cathelicidin LL-37
during Mycobacterium tuberculosis infection in human alveolar macrophages, monocytes,
neutrophils, and epithelial cells. Infect Immun 76:935–941
Röhrl J, Yang D, Oppenheim JJ, Hehlgans T (2010) Human beta-defensin 2 and 3 and their mouse
orthologs induce chemotaxis through interaction with CCR2. J Immunol 184:6688–6694
Ross DJ, Cole AM, Yoshioka D et al (2004) Increased bronchoalveolar lavage human beta-
defensin type 2 in bronchiolitis obliterans syndrome after lung transplantation. Transplantation
78:1222–1224
Saiman L, Tabibi S, Starner TD et al (2001) Cathelicidin peptides inhibit multiply antibiotic-resistant
pathogens from patients with cystic fibrosis. Antimicrob Agents Chemother 45:2838–2844
Sakamoto N, Mukae H, Fujii T et al (2005) Differential effects of alpha- and beta-defensin on
cytokine production by cultured human bronchial epithelial cells. Am J Physiol Lung Cell Mol
Physiol 288:L508–L513
Schaller-Bals S, Schulze A, Bals R (2002) Increased levels of antimicrobial peptides in tracheal
aspirates of newborn infants during infection. Am J Respir Crit Care Med 165:992–995
Scharf S, Vardarova K, Lang F et al (2010a) Legionella pneumophila induces human beta defen-
sin-3 in pulmonary cells. Respir Res 11:93
Scharf S, Hippenstiel S, Flieger A et al (2010b) Induction of human β-defensin-2 in pulmonary
epithelial cells by Legionella pneumophila: involvement of TLR2 and TLR5, p38 MAPK,
JNK, NF-κB, and AP-1. Am J Physiol Lung Cell Mol Physiol 298:L687–L695
Scharf S, Zahlten J, Szymanski K et al (2012) Streptococcus pneumoniae induces human
β-defensin-2 and -3 in human lung epithelium. Exp Lung Res 38:100–110
Schauber J, Dorschner RA, Coda AB et al (2007a) Injury enhances TLR2 function and antimicro-
bial peptide expression through a vitamin D-dependent mechanism. J Clin Invest 117:803–811
Schauber J, Oda Y, Büchau AS et al (2007b) Histone acetylation in keratinocytes enables control
of the expression of cathelicidin and CD14 by 1,25-dihydroxyvitamin D3. J Invest Dermatol
128:816–824
Schiemann F, Brandt E, Gross R et al (2009) The cathelicidin LL-37 activates human mast cells
and is degraded by mast cell tryptase: counter-regulation by CXCL4. J Immunol 183:
2223–2231
Scott MG, Gold MR, Hancock RE (1999a) Interaction of cationic peptides with lipoteichoic acid
and gram-positive bacteria. Infect Immun 67:6445–6453
Scott MG, Yan H, Hancock REW (1999b) Biological properties of structurally related alpha -heli-
cal cationic antimicrobial peptides. Infect Immun 67:2005–2009
3 Function of Antimicrobial Peptides in Lung Innate Immunity 51
Scott MG, Vreugdenhil AC, Buurman WA et al (2000) Cutting edge: cationic antimicrobial pep-
tides block the binding of lipopolysaccharide (LPS) to LPS binding protein. J Immunol
164:549–553
Scott MG, Davidson DJ, Gold MR et al (2002) The human antimicrobial peptide LL-37 is a mul-
tifunctional modulator of innate immune responses. J Immunol 169:3883–3891
Scott A, Weldon S, Buchanan PJ et al (2011) Evaluation of the ability of LL-37 to neutralise LPS
in vitro and ex vivo. PLoS One 6, e26525
Seiler F, Hellberg J, Lepper PM et al (2013) FOXO transcription factors regulate innate immune
mechanisms in respiratory epithelial cells. J Immunol 190:1603–1613
Selsted ME, Ouellette AJ (2005) Mammalian defensins in the antimicrobial immune response. Nat
Immunol 6:551–557
Semple F, Webb S, Li H-N et al (2010) Human beta-defensin 3 has immunosuppressive activity
in vitro and in vivo. Eur J Immunol 40:1073–1078
Semple F, MacPherson H, Webb S et al (2011) Human β-defensin 3 affects the activity of pro-
inflammatory pathways associated with MyD88 and TRIF. Eur J Immunol 41:3291–3300
Sethi S (2010) Infection as a comorbidity of COPD. Eur Respir J 35:1209–1215
Sethi S, Murphy TF (2008) Infection in the pathogenesis and course of chronic obstructive pulmo-
nary disease. N Engl J Med 359:2355–2365
Shaykhiev R, Beisswenger C, Kändler K et al (2005) Human endogenous antibiotic LL-37 stimu-
lates airway epithelial cell proliferation and wound closure. Am J Physiol Lung Cell Mol
Physiol 289:L842–L848
Shaykhiev R, Sierigk J, Herr C et al (2010) The antimicrobial peptide cathelicidin enhances activa-
tion of lung epithelial cells by LPS. FASEB J 24:4756–4766
Shestakova T, Zhuravel E, Bolgova L et al (2008) Expression of human beta-defensins-1, 2 and 4
mRNA in human lung tumor tissue: a pilot study. Exp Oncol 30:153–156
Shu Q, Shi Z, Zhao Z et al (2006) Protection against Pseudomonas aeruginosa pneumonia and
sepsis-induced lung injury by overexpression of beta-defensin-2 in rats. Shock 26:365–371
Singh PK, Jia HP, Wiles K et al (1998) Production of beta-defensins by human airway epithelia.
Proc Natl Acad Sci U S A 95:14961–14966
Soong LB, Ganz T, Ellison A, Caughey GH (1997) Purification and characterization of defensins
from cystic fibrosis sputum. Inflamm Res 46:98–102
Soruri A, Grigat J, Forssmann U et al (2007) beta-Defensins chemoattract macrophages and mast
cells but not lymphocytes and dendritic cells: CCR6 is not involved. Eur J Immunol 37:
2474–2486
Steinstraesser L, Koehler T, Jacobsen F et al (2008) Host defense peptides in wound healing. Mol
Med 14:528–537
Sun C, Zhu M, Yang Z et al (2014) LL-37 secreted by epithelium promotes fibroblast collagen
production: a potential mechanism of small airway remodeling in chronic obstructive pulmo-
nary disease. Lab Invest 94:991–1002
Tjabringa GS, Aarbiou J, Ninaber DK et al (2003) The antimicrobial peptide LL-37 activates
innate immunity at the airway epithelial surface by transactivation of the epidermal growth
factor receptor. J Immunol 171:6690–6696
Tokumaru S, Sayama K, Shirakata Y et al (2005) Induction of keratinocyte migration via transac-
tivation of the epidermal growth factor receptor by the antimicrobial peptide LL-37. J Immunol
175:4662–4668
Tripathi S, Tecle T, Verma A et al (2013) The human cathelicidin LL-37 inhibits influenza A
viruses through a mechanism distinct from that of surfactant protein D or defensins. J Gen
Virol 94:40–49
Van Wetering S, Mannesse-Lazeroms SP, Dijkman JH, Hiemstra PS (1997) Effect of neutrophil
serine proteinases and defensins on lung epithelial cells: modulation of cytotoxicity and IL-8
production. J Leukoc Biol 62:217–226
Vandamme D, Landuyt B, Luyten W, Schoofs L (2012) A comprehensive summary of LL-37, the
factotum human cathelicidin peptide. Cell Immunol 280:22–35
52 F. Seiler et al.
Abstract Due to its close proximity to the gastrointestinal tract, the normally ster-
ile urinary tract is constantly challenged by microbial invasion. To counter this
microbial assault, the urinary tract has developed a highly effective antimicrobial
“shield” that can rapidly eliminate invading pathogens or prevent their growth.
During recent years, considerable advances have been made in our understanding of
the immune mechanisms that contribute to urinary tract sterility. Recent evidence
indicates that cationic antimicrobial peptides contribute to the innate host defense of
the urinary tract. This chapter reviews the published literature on the role(s) of anti-
microbial peptides (AMPs) in maintaining urinary tract sterility.
4.1 Introduction
Urinary tract infections (UTI) are one of the most common and serious bacterial
infections encountered by physicians (Bachur and Harper 2001; Foxman et al. 2000).
Nearly half of all women develop a UTI requiring antimicrobial therapy during their
lifetime (Foxman et al. 2000). In 2013, aggregate hospital charges for inpatient UTI
management exceeded $640 million US dollars (Spencer et al. 2011a). Specific sub-
populations have increased UTI risk, including pregnant women, the elderly, patients
with diabetes or multiple sclerosis, patients with acquired immunodeficiency dis-
eases, patients with urologic anomalies, and those having urinary tract intervention
such as catheter insertion. Although UTI is not typically associated with significant
morbidities, UTI does increase the risk of premature delivery and fetal mortality
among pregnant women (Foxman et al. 2000). Long-term complications of UTI
include renal insufficiency, renal scarring, hypertension, and chronic kidney disease.
UTI refers to the presence of microbial pathogens within the urinary tract. The site
of infection classifies UTI—infection localized to the bladder is referred to as “cys-
titis” and infection in the kidney is “pyelonephritis.” Cystitis typically presents with
lower urinary tract symptoms—including dysuria, urgency, and urinary frequency.
Pyelonephritis is often associated with more severe or systemic symptoms includ-
ing fever, back/flank pain, and vomiting. Ascending infection may result in bactere-
mia and present as the systemic inflammatory response syndrome or overt septic
shock (i.e., urosepsis).
Escherichia coli (E. coli) is the most frequent bacterial pathogen responsible for
UTI—accounting for 85–90 % of cases. Uropathogenic E. coli (UPEC) are thought to
originate from the fecal flora, spread across the perineum, and enter the bladder through
the urethra. Before invading the urothelium, UPEC must overcome several intrinsic
characteristics of the urinary tract. Proposed functional mechanisms contributing to
urinary tract sterility include barrier formation by uroepithelial cells, unidirectional
flow of urine, regular bladder emptying, mucous production, the urinary microbiome,
and alterations in the urinary ionic composition (Spencer et al. 2014a; Sobel 1997).
The microbial virulence of UPEC has been linked to many factors (Ragnarsdottir
and Svanborg 2012; Mulvey et al. 1998, 2000). The most prominent is type I fim-
briae, which are filamentous bacterial appendages that are capped by FimH, a
mannose-binding adhesion protein. Type I fimbriae promote tight bacterial binding
to a matrix of uroplakin complexes on the surface of superficial bladder epithelial
cells (Mulvey et al. 2000). After binding, UPEC invades the bladder uroepithelium
where it may establish a state of commensalism or cause a severe, symptomatic
infection characterized by a rapid innate host response with cytokine secretion and
recruitment of leukocytes to the infection source (Weichhart et al. 2008).
In the kidney, UPEC binds to the apical surfaces of the kidney’s collect duct. Within
the collecting duct, UPEC specifically targets the intercalated cells (Chassin et al.
2011; Paragas et al. 2014). The signaling pathways activated by UPEC have been
investigated in primary cultures of medullary collecting duct cells dissected from the
kidneys of LPS-sensitive C3H/HeOuJ mice (Chassin et al. 2011). Signaling pathway
analysis demonstrates that UPEC stimulates the expression of pro-inflammatory
mediators in the medullary collecting ducts via TLR4-mediated, MyD88-dependent,
TRIF-independent NF-kB and MAPK-activated pathways and also via a TLR4-
independent, MyD88-independent pathway. The TLR4-independent pathway results
from activation of the TNF receptor-associated factor-2 (TRAF2) and apoptosis sig-
nal-regulatory kinase 1 (ASK1)-JNK pathway (Chassin et al. 2011; Vandewalle 2008).
4 Antimicrobial Peptides: Maintaining Sterility of the Urinary Tract 55
Proximal Tubule
Ureter LL-37
HD5
LL37
RNase7
Urethra
Bladder Loop of Henle
HD5
HD5 HBD1
RNase7 HBD2 Collecting Tubule
HD5 HBD1
HBD2
HD5
RNase7
Fig. 4.1 Defined AMP in the kidney and urinary tract. Left panel: AMP identified in the human
lower and upper urinary tract. Right panel: AMPs identified in the nephron and collecting tubule
of human kidney (Image reused with permission from Spencer et al. (2014a))
4.3 Cathelicidin
The human cathelicidin LL-37 is an amphipathic, α-helical AMP expressed on all epi-
thelial surfaces, myeloid bone marrow cells, and circulating neutrophils. Cathelicidin
possesses antimicrobial activity against viruses and Gram-positive and Gram-negative
bacteria. Moreover, it acts as a chemoattractant for neutrophils and tissue-derived
monocytes by interacting with their fMLP receptors. The optimal chemoattractant
56 B. Becknell and J.D. Spencer
4.4 Defensins
Defensins, characterized by a 15–20 amino acid sequence including six cysteine resi-
dues, are one of the most studied families of AMPs. Defensins typically have broad-
spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria,
viruses, fungi, and protozoa (Lehrer et al. 1993). Along with their direct antimicrobial
properties, defensins play a role in cell-mediated immunity as chemoattractants for
immature dendritic cells (Zasloff 2007). Defensins are initially synthesized as
4 Antimicrobial Peptides: Maintaining Sterility of the Urinary Tract 57
4.5 α-Defensins
The α-defensins HNP1–HNP4 are primarily found in neutrophils where they pro-
vide non-oxidative microbicidal activity. HNPs encounter pathogens after they are
secreted onto the cell’s surface via degranulation or after a pathogen undergoes
phagocytosis and the phagocytic vacuole fuses with the neutrophilic granule (Ganz
2003). Regarding the urinary tract, Ihi et al. demonstrated that urinary levels of
HNP1–HNP3 significantly increased in the setting of UPEC and Enterococcus fae-
calis UTI (6.5 ± 1.1 pg/μL to 29 ± 5.7 pg/μL) (Ihi et al. 1997). Similarly, Tikhonov
et al. demonstrated that urinary HNP1 increased eightfold in patients with chronic
pyelonephritis compared to control patients and patients with glomerulonephritis.
Urinary HNP1 levels correlated with urinary IL-8 levels as well as leukocyte count,
suggesting that urinary HNP1 may reflect neutrophil recruitment to the infection
site (Tikhonov et al. 1997). No microbiological information has been published in
regard to the HNPs and UTI.
The expression and function of epithelial human defensin HD5 have been
reported mostly in the small intestine where it is secreted by Paneth cells into the
intestinal crypts. HD5 has also been described in the male and female reproductive
tracts, with evidence suggesting that it is inducible and important in eradicating
infection (Porter et al. 2005; Quayle et al. 1998). Recombinant mature HD5 has
been shown to have bactericidal activity against uropathogenic bacteria, including
Pseudomonas aeruginosa, Klebsiella pneumoniae, Staphylococcus epidermidis,
Enterococcus faecium, and UPEC (Wang et al. 2010).
In the urinary tract, HD5 has been localized to the uroepithelium of the kidney,
ureter, and bladder (Spencer et al. 2012). Schwaderer et al. demonstrate that HD5
gene and protein expression were significantly greater in human kidney biopsy
specimens from patients with pyelonephritis compared to noninfected controls.
Their data also show that secreted HD5 was not routinely detected in culture-
negative human urine samples; however, HD5 levels significantly increased in urine
samples infected with UPEC (300–670 ng HD5/mg urine creatinine). Although uri-
nary HD5 did not reach concentrations likely to be directly antimicrobial against
common uropathogens, the mucosal surface concentrations may be higher (Spencer
et al. 2012). Urinary HD5 levels are higher in patients who have undergone ileal
neobladder reconstruction and ileal conduit urinary diversion (Porter et al. 1998;
Townes et al. 2011).
58 B. Becknell and J.D. Spencer
4.6 β-Defensins
The human β-defensins, encoded by over 28 genes, are widely expressed in human
epithelia and certain family members exhibit antimicrobial activity toward Gram-
positive and Gram-negative bacteria (Ali et al. 2009; Schutte et al. 2002). Of the
β-defensins, human β-defensin-1 (HBD1, encoded by the DEFB1 gene) and human
β-defensin-2 (HBD2, encoded by DEFB4) have been described in the human urinary
tract. DEFB1 mRNA is constitutively expressed by the epithelial lining of the kid-
ney’s loop of Henle, the distal tubule, and the collecting duct (Valore et al. 1998).
HBD1 is translated as a 68 amino acid pro-peptide and undergoes variable amino-
terminal processing to a 36–47 amino acid (Valore et al. 1998; Zucht et al. 1998).
Mature HBD1 is constitutively detected in sterile urine (10–100 μg/L) and levels
increase up to threefold in patients with UTI (Valore et al. 1998; Hiratsuka et al.
2000). Although these urinary levels of HBD1 are insufficient to kill invading bacte-
ria, HBD1 may provide a fast-acting antimicrobial coating of urothelium and prevent
infection by inhibiting bacterial attachment to the urothelium (Valore et al. 1998).
Defb1 encoding mouse BD1 (mBD1) has been considered orthologous to HBD1
based on conserved gene structure, expression pattern, and antimicrobial activity,
though the amino acid sequences of mBD1 and HBD1 are only 51 % identical
(Huttner et al. 1997; Bals et al. 1998; Morrison et al. 1998). Defb1 is transcribed
within lower urinary tract and the kidney’s distal tubules and collecting ducts (Bals
et al. 1998; Becknell et al. 2013). The mature mBD1 peptide exhibits salt-sensitive
antimicrobial activity toward Gram-positive and Gram-negative bacteria (Bals et al.
1998; Morrison et al. 1998). Mice lacking both copies of Defb1 (Defb1−/−) exhibit
increased incidence of spontaneous bacteriuria compared to Defb1+/− and wild-type
controls, with Staphylococcus species predominating among urine isolates
(Morrison et al. 2002). Wild-type mice undergoing transurethral inoculation of
UPEC exhibit significant reduction in bladder Defb1 transcript levels within 2 h of
infection that persists up to 2 days. These results suggest that successful UPEC
colonization of the urinary tract is achieved through local inhibition of Defb1
expression. However, when Defb1−/− animals were challenged with UPEC, no dif-
ference in upper or lower tract bacterial burden was observed relative to wild-type
controls (Becknell et al. 2013).
Additional Defb family members are detectable at the mRNA level in the murine
urinary tract. Defb3 (encoding mBD3) and Defb14 (encoding mBD14) mRNA are
enriched in mouse bladder, compared to ureter and kidney (Becknell et al. 2013).
Both mBD3 and mBD14 exhibit bactericidal activity toward UPEC in the low-
micromolar range. Other Defb transcripts, such as Defb2, Defb28, Defb29, and
Defb42, are enriched or exclusively expressed in murine ureter and kidney, compared
to bladder (Becknell et al. 2013). The lack of available antibodies has limited the
study of the levels and distribution of these mBD peptides in the urinary tract. The
establishment of mouse strains with deletions in one or more of Defb family mem-
bers will facilitate studies that determine their biological relevance within the uri-
nary tract (Zhou et al. 2013; Navid et al. 2012).
4 Antimicrobial Peptides: Maintaining Sterility of the Urinary Tract 59
4.7 Hepcidin
Lactoferrin inhibits bacterial growth through free iron chelation or the effects of
lactoferricin, a bactericidal protein generated by the proteolytic cleavage of lacto-
ferrin (Bellamy et al. 1992). Lactoferrin is detectable in human urine; however,
these levels are relatively low (14–145 ng/mL). In the human kidney, lactoferrin is
expressed in the cells lining the distal collecting ducts of the medulla (Abrink et al.
2000). Lipocalin 2 (LCN2) is a member of the large lipocalin protein family, which
has a wide range of biological functions. Several reports indicate that lipocalin 2
(LCN2) mediates a bacteriostatic effect by sequestering iron (Berger et al. 2006; Flo
et al. 2004; Holmes et al. 2005). Specifically, LCN2 binds the secreted siderophore
enterochelin (Ent), which UPEC and other pathogens release into the extracellular
milieu to acquire essential iron. Upon complexing with Ent:Fe3+, LCN2 reroutes
Ent:Fe3+ for degradation and prevents iron transfer to bacteria. Without iron, uro-
pathogens are unable to grow, facilitating immune clearance (Paragas et al. 2014).
Thus, Lcn2-deficient mice demonstrate increased susceptibility to bacterial infec-
tions when challenged with intraperitoneal E. coli (Berger et al. 2006).
Recently, Paragas et al. observed a significant induction of Lcn2 in the urine of
mice with UTI compared with uninfected animals. The degree of Lcn2 upregulation
correlated with the number of infecting bacteria in the urine and reduction of bacte-
rial load with antibiotics resulted in decreased Lcn2 production. A similar correla-
tion between bacterial load and LCN2 was also seen in the clinical setting—with
urinary levels increasing tenfold in patients with UTI (30–300 ng/mL) compared to
60 B. Becknell and J.D. Spencer
healthy controls (7.4–36.3 ng/mL). These results provide clinical support for LCN2
production in immune defense of the urinary tract (Paragas et al. 2014). Additionally,
using an Lcn2 global knockout mouse, Paragas et al. demonstrate that after transure-
thral UPEC inoculation, Lcn2−/− mice have higher bladder bacterial burden than
their littermates and take longer to clear the infection. Both wild-type and Lcn2−/−
mice were comparably infected with an Ent mutant UPEC strain, indicating that the
antimicrobial actions of Lcn2 on uropathogens require iron acquisition (Paragas
et al. 2014).
Using an Lcn2-Luc2-mCherry reporter mouse, this same research group demon-
strates that Lcn2 is constitutively produced by the thick ascending limbs of the loop
of Henle and the α-intercalated cells of the renal collecting tubule (Paragas et al.
2011, 2014). During experimental UTI, GFP-labeled UPEC bound the kidney’s
α-intercalated cells and induced Lcn2 expression in a TLR-4- and NF-κB-sensitive
fashion (Paragas et al. 2014; van Adelsberg et al. 1994; Xie et al. 2008).
the renal collecting tubule. When urinary RNase 7 is neutralized in human urine
specimens in vitro, urinary bacterial growth increases (Spencer et al. 2011b, 2014b).
During UTI, urine from children with UPEC infection contained about twice the
concentration of RNase 7 as the urine of noninfected controls. Similarly, human
kidney biopsies from patients with pyelonephritis demonstrate that tissue concen-
trations of RNase 7 protein are about twice what was measured in normal controls.
What distinguishes the expression of RNase 7 within the urinary tract from the other
previously described AMPs is the considerably greater amount secreted. Whereas
median urinary concentrations of cathelicidin and HBD1 are 1.6 × 10−5 μM and
2.5 × 10−5 μM, respectively, RNase 7 is present at around a 1000-fold greater con-
centrations (Spencer et al. 2011b, 2013; Zasloff 2013). These results suggest that
RNase 7 provides a front-line antimicrobial shield that permits uropathogenic
organisms from invading the urothelium (Zasloff 2013). If this barrier is breached
during microbial assault, other defenses like RNase 6 or other AMPs are activated
to combat infection.
THP or uromodulin is the most abundant protein in human urine, and mounting
evidence points to its role in maintaining urinary tract sterility. Mice deficient in
Thp (Thp−/−) exhibit delayed clearance of UPEC and higher UPEC recovery from
urine and bladders following experimental UTI (Bates et al. 2004). This was con-
firmed independently by Mo and colleagues, who further demonstrated that this
protective effect of Thp existed specifically toward type 1-fimbriated UPEC (Mo
et al. 2004). This may be explained by the observation that type 1-fimbriated UPEC
interacts with THP’s mannose residues and relies on these structures for attachment
to uroplakin plaques on bladder urothelium (Pak et al. 2001). While THP does not
possess bactericidal activity in vitro, it is capable of inhibiting bacterial attachment
to cultured kidney epithelial cells (Hawthorn et al. 1991).
The therapeutic potential of AMPs to prevent or treat UTI has been given added
incentive as antibiotic resistance toward uropathogenic bacteria has been increasing.
The use of AMPs does require some degree of caution as AMPs are powerful bio-
molecules that can elicit native cell death if applied in high concentration (Ali et al.
2009). The most straightforward method of AMP UTI therapy is direct application to
the source of infection (i.e., through bladder washes) or oral therapy. The use of oral
lactoferrin B effectively decreased infection and inflammation in the mouse urinary
tract after UPEC infection (Haversen et al. 2000). An alternative possibility for using
AMPs therapeutically would be to induce or augment their natural production using
62 B. Becknell and J.D. Spencer
References
Abrink M, Larsson E, Gobl A, Hellman L (2000) Expression of lactoferrin in the kidney: implica-
tions for innate immunity and iron metabolism. Kidney Int 57:2004–2010
Ali AS, Townes CL, Hall J, Pickard RS (2009) Maintaining a sterile urinary tract: the role of anti-
microbial peptides. J Urol 182:21–28
Bachur RG, Harper MB (2001) Predictive model for serious bacterial infections among infants
younger than 3 months of age. Pediatrics 108:311–316
Bals R, Goldman MJ, Wilson JM (1998) Mouse beta-defensin 1 is a salt-sensitive antimicrobial
peptide present in epithelia of the lung and urogenital tract. Infect Immun 66:1225–1232
Bates JM, Raffi HM, Prasadan K, Mascarenhas R, Laszik Z, Maeda N, Hultgren SJ, Kumar S
(2004) Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: rapid
communication. Kidney Int 65:791–797
Becknell B, Spencer JD, Carpenter AR, Chen X, Singh A, Ploeger S, Kline J, Ellsworth P, Li B,
Proksch E, Schwaderer AL, Hains DS, Justice SS, McHugh KM (2013) Expression and anti-
microbial function of beta-defensin 1 in the lower urinary tract. PLoS One 8:e77714
Becknell B, Eichler TE, Beceiro S, Li B, Easterling RS, Carpenter AR, James CL, McHugh KM,
Hains DS, Partida-Sanchez S, Spencer JD (2015) Ribonucleases 6 and 7 have antimicrobial
function in the human and murine urinary tract. Kidney Int 87(1):151–161
Bellamy W, Takase M, Wakabayashi H, Kawase K, Tomita M (1992) Antibacterial spectrum of
lactoferricin B, a potent bactericidal peptide derived from the N-terminal region of bovine
lactoferrin. J Appl Bacteriol 73:472–479
Berger T, Togawa A, Duncan GS, Elia AJ, You-Ten A, Wakeham A, Fong HE, Cheung CC, Mak
TW (2006) Lipocalin 2-deficient mice exhibit increased sensitivity to Escherichia coli infection
but not to ischemia-reperfusion injury. Proc Natl Acad Sci U S A 103:1834–1839
Boix E, Nogues MV (2007) Mammalian antimicrobial proteins and peptides: overview on the
RNase A superfamily members involved in innate host defence. Mol Biosyst 3:317–335
Chassin C, Tourneur E, Bens M, Vandewalle A (2011) A role for collecting duct epithelial cells in
renal antibacterial defences. Cell Microbiol 13:1107–1113
Chromek M (2015) The role of the antimicrobial peptide cathelicidin in renal diseases. Pediatr
Nephrol 30(8):1225–1232
Chromek M, Slamova Z, Bergman P, Kovacs L, Podracka L, Ehren I, Hokfelt T, Gudmundsson
GH, Gallo RL, Agerberth B, Brauner A (2006) The antimicrobial peptide cathelicidin protects
the urinary tract against invasive bacterial infection. Nat Med 12:636–641
Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira S, Aderem A (2004)
Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron.
Nature 432:917–921
4 Antimicrobial Peptides: Maintaining Sterility of the Urinary Tract 63
Foxman B, Barlow R, D’Arcy H, Gillespie B, Sobel JD (2000) Urinary tract infection: self-reported
incidence and associated costs. Ann Epidemiol 10:509–515
Ganz T (2003) Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol 3:710–720
Harder J, Schroder JM (2002) RNase 7, a novel innate immune defense antimicrobial protein of
healthy human skin. J Biol Chem 277:46779–46784
Haversen LA, Engberg I, Baltzer L, Dolphin G, Hanson LA, Mattsby-Baltzer I (2000) Human
lactoferrin and peptides derived from a surface-exposed helical region reduce experimental
Escherichia coli urinary tract infection in mice. Infect Immun 68:5816–5823
Hawthorn LA, Bruce AW, Reid G (1991) Ability of uropathogens to bind to Tamm Horsfall
protein-coated renal tubular cells. Urol Res 19:301–304
Hertting O, Holm A, Luthje P, Brauner H, Dyrdak R, Jonasson AF, Wiklund P, Chromek M,
Brauner A (2010) Vitamin D induction of the human antimicrobial Peptide cathelicidin in the
urinary bladder. PLoS One 5:e15580
Hiratsuka T, Nakazato M, Ihi T, Minematsu T, Chino N, Nakanishi T, Shimizu A, Kangawa K,
Matsukura S (2000) Structural analysis of human beta-defensin-1 and its significance in uri-
nary tract infection. Nephron 85:34–40
Holmes MA, Paulsene W, Jide X, Ratledge C, Strong RK (2005) Siderocalin (Lcn 2) also binds
carboxymycobactins, potentially defending against mycobacterial infections through iron
sequestration. Structure 13:29–41
Huang YC, Lin YM, Chang TW, Wu SJ, Lee YS, Chang MD, Chen C, Wu SH, Liao YD (2007)
The flexible and clustered lysine residues of human ribonuclease 7 are critical for membrane
permeability and antimicrobial activity. J Biol Chem 282:4626–4633
Huttner KM, Kozak CA, Bevins CL (1997) The mouse genome encodes a single homolog of the
antimicrobial peptide human beta-defensin 1. FEBS Lett 413:45–49
Ihi T, Nakazato M, Mukae H, Matsukura S (1997) Elevated concentrations of human neutrophil
peptides in plasma, blood, and body fluids from patients with infections. Clin Infect Dis 25:
1134–1140
Krause A, Neitz S, Magert HJ, Schulz A, Forssmann WG, Schulz-Knappe P, Adermann K (2000)
LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS
Lett 480:147–150
Lehrer RI, Lichtenstein AK, Ganz T (1993) Defensins: antimicrobial and cytotoxic peptides of
mammalian cells. Annu Rev Immunol 11:105–128
Liu L, Zhao C, Heng HH, Ganz T (1997) The human beta-defensin-1 and alpha-defensins are
encoded by adjacent genes: two peptide families with differing disulfide topology share a com-
mon ancestry. Genomics 43:316–320
Mo L, Zhu XH, Huang HY, Shapiro E, Hasty DL, Wu XR (2004) Ablation of the Tamm-Horsfall
protein gene increases susceptibility of mice to bladder colonization by type 1-fimbriated
Escherichia coli. Am J Physiol Renal Physiol 286:F795–F802
Morrison GM, Davidson DJ, Kilanowski FM, Borthwick DW, Crook K, Maxwell AI, Govan JR,
Dorin JR (1998) Mouse beta defensin-1 is a functional homolog of human beta defensin-1.
Mamm Genome 9:453–457
Morrison G, Kilanowski F, Davidson D, Dorin J (2002) Characterization of the mouse beta defen-
sin 1, Defb1, mutant mouse model. Infect Immun 70:3053–3060
Mulvey MA, Lopez-Boado YS, Wilson CL, Roth R, Parks WC, Heuser J, Hultgren SJ (1998)
Induction and evasion of host defenses by type 1-piliated uropathogenic Escherichia coli.
Science 282:1494–1497
Mulvey MA, Schilling JD, Martinez JJ, Hultgren SJ (2000) Bad bugs and beleaguered bladders:
interplay between uropathogenic Escherichia coli and innate host defenses. Proc Natl Acad Sci
U S A 97:8829–8835
Navid F, Boniotto M, Walker C, Ahrens K, Proksch E, Sparwasser T, Muller W, Schwarz T,
Schwarz A (2012) Induction of regulatory T cells by a murine beta-defensin. J Immunol 188:
735–743
Pak J, Pu Y, Zhang ZT, Hasty DL, Wu XR (2001) Tamm-Horsfall protein binds to type 1 fimbriated
Escherichia coli and prevents E. coli from binding to uroplakin Ia and Ib receptors. J Biol Chem
276:9924–9930
64 B. Becknell and J.D. Spencer
Paragas N, Qiu A, Zhang Q, Samstein B, Deng SX, Schmidt-Ott KM, Viltard M, Yu W, Forster CS,
Gong G, Liu Y, Kulkarni R, Mori K, Kalandadze A, Ratner AJ, Devarajan P, Landry DW,
D’Agati V, Lin CS, Barasch J (2011) The Ngal reporter mouse detects the response of the kid-
ney to injury in real time. Nat Med 17:216–222
Paragas N, Kulkarni R, Werth M, Schmidt-Ott KM, Forster C, Deng R, Zhang Q, Singer E, Klose
AD, Shen TH, Francis KP, Ray S, Vijayakumar S, Seward S, Bovino ME, Xu K, Takabe Y,
Amaral FE, Mohan S, Wax R, Corbin K, Sanna-Cherchi S, Mori K, Johnson L, Nickolas T,
D’Agati V, Lin CS, Qiu A, Al-Awqati Q, Ratner AJ, Barasch J (2014) alpha-Intercalated cells
defend the urinary system from bacterial infection. J Clin Invest 124:2963–2976
Park CH, Valore EV, Waring AJ, Ganz T (2001) Hepcidin, a urinary antimicrobial peptide synthe-
sized in the liver. J Biol Chem 276:7806–7810
Porter EM, Poles MA, Lee JS, Naitoh J, Bevins CL, Ganz T (1998) Isolation of human intestinal
defensins from ileal neobladder urine. FEBS Lett 434:272–276
Porter E, Yang H, Yavagal S, Preza GC, Murillo O, Lima H, Greene S, Mahoozi L, Klein-Patel M,
Diamond G, Gulati S, Ganz T, Rice PA, Quayle AJ (2005) Distinct defensin profiles in Neisseria
gonorrhoeae and Chlamydia trachomatis urethritis reveal novel epithelial cell-neutrophil inter-
actions. Infect Immun 73:4823–4833
Quayle AJ, Porter EM, Nussbaum AA, Wang YM, Brabec C, Yip KP, Mok SC (1998) Gene
expression, immunolocalization, and secretion of human defensin-5 in human female repro-
ductive tract. Am J Pathol 152:1247–1258
Ragnarsdottir B, Svanborg C (2012) Susceptibility to acute pyelonephritis or asymptomatic bacte-
riuria: host-pathogen interaction in urinary tract infections. Pediatr Nephrol 27:2017–2029
Rosenberg HF (2008) RNase A ribonucleases and host defense: an evolving story. J Leukoc Biol
83:1079–1087
Schutte BC, Mitros JP, Bartlett JA, Walters JD, Jia HP, Welsh MJ, Casavant TL, McCray PB Jr
(2002) Discovery of five conserved beta -defensin gene clusters using a computational search
strategy. Proc Natl Acad Sci U S A 99:2129–2133
Simanski M, Dressel S, Glaser R, Harder J (2010) RNase 7 protects healthy skin from
Staphylococcus aureus colonization. J Invest Dermatol 130:2836–2838
Sobel JD (1997) Pathogenesis of urinary tract infection. Role of host defenses. Infect Dis Clin
North Am 11:531–549
Spencer JD, Schwaderer A, McHugh K, Vanderbrink B, Becknell B, Hains DS (2011a) The demo-
graphics and costs of inpatient vesicoureteral reflux management in the USA. Pediatr Nephrol
26(11):1995–2001
Spencer JD, Schwaderer AL, Dirosario JD, McHugh KM, McGillivary G, Justice SS, Carpenter
AR, Baker PB, Harder J, Hains DS (2011b) Ribonuclease 7 is a potent antimicrobial peptide
within the human urinary tract. Kidney Int 80(2):174–180
Spencer JD, Hains DS, Porter E, Bevins CL, DiRosario J, Becknell B, Wang H, Schwaderer AL
(2012) Human alpha defensin 5 expression in the human kidney and urinary tract. PLoS One
7:e31712
Spencer JD, Schwaderer AL, Wang H, Bartz J, Kline J, Eichler T, Desouza KR, Sims-Lucas S,
Baker P, Hains DS (2013) Ribonuclease 7, an antimicrobial peptide upregulated during infec-
tion, contributes to microbial defense of the human urinary tract. Kidney Int 83(4):615–625
Spencer JD, Schwaderer AL, Becknell B, Watson J, Hains DS (2014a) The innate immune
response during urinary tract infection and pyelonephritis. Pediatr Nephrol 29(7):1139–1149
Spencer JD, Schwaderer AL, Eichler T, Wang H, Kline J, Justice SS, Cohen DM, Hains DS
(2014b) An endogenous ribonuclease inhibitor regulates the antimicrobial activity of ribonu-
clease 7 in the human urinary tract. Kidney Int 85(5):1179–1191
Tikhonov I, Rebenok A, Chyzh A (1997) A study of interleukin-8 and defensins in urine and
plasma of patients with pyelonephritis and glomerulonephritis. Nephrol Dial Transplant
12:2557–2561
Townes CL, Ali A, Robson W, Pickard R, Hall J (2011) Tolerance of bacteriuria after urinary
diversion is linked to antimicrobial peptide activity. Urology 77(509):e501–e508
Valore EV, Park CH, Quayle AJ, Wiles KR, McCray PB Jr, Ganz T (1998) Human beta-defensin-1:
an antimicrobial peptide of urogenital tissues. J Clin Invest 101:1633–1642
4 Antimicrobial Peptides: Maintaining Sterility of the Urinary Tract 65
van Adelsberg J, Edwards JC, Takito J, Kiss B, al-Awqati Q (1994) An induced extracellular
matrix protein reverses the polarity of band 3 in intercalated epithelial cells. Cell 76:
1053–1061
Vandewalle A (2008) Toll-like receptors and renal bacterial infections. Chang Gung Med J 31:
525–537
Wang AP, Su YP, Shen MQ, Chen F, Chen M, Wang JP (2010) Antibacterial activity and mecha-
nism of recombinant human alpha defensin 5 against clinical antibiotic-resistant strains. Afr
J Microbiol Res 4:626–633
Wang H, Schwaderer AL, Kline J, Spencer JD, Kline D, Hains DS (2013) Contribution of struc-
tural domains to ribonuclease 7’s activity against uropathogenic bacteria. Antimicrob Agents
Chemother 57(2):766–774
Weichhart T, Haidinger M, Horl WH, Saemann MD (2008) Current concepts of molecular defence
mechanisms operative during urinary tract infection. Eur J Clin Invest 38(Suppl 2):29–38
Weinstein DA, Roy CN, Fleming MD, Loda MF, Wolfsdorf JI, Andrews NC (2002) Inappropriate
expression of hepcidin is associated with iron refractory anemia: implications for the anemia of
chronic disease. Blood 100:3776–3781
Xie Y, Deng S, Thomas CJ, Liu Y, Zhang YQ, Rinderspacher A, Huang W, Gong G, Wyler M,
Cayanis E, Aulner N, Tobben U, Chung C, Pampou S, Southall N, Vidovic D, Schurer S,
Branden L, Davis RE, Staudt LM, Inglese J, Austin CP, Landry DW, Smith DH, Auld DS
(2008) Identification of N-(quinolin-8-yl)benzenesulfonamides as agents capable of down-
regulating NFkappaB activity within two separate high-throughput screens of NFkappaB acti-
vation. Bioorg Med Chem Lett 18:329–335
Yang D, Liu ZH, Tewary P, Chen Q, de la Rosa G, Oppenheim JJ (2007) Defensin participation in
innate and adaptive immunity. Curr Pharm Des 13:3131–3139
Zasloff M (2007) Antimicrobial peptides, innate immunity, and the normally sterile urinary tract.
J Am Soc Nephrol 18:2810–2816
Zasloff M (2013) The antibacterial shield of the human urinary tract. Kidney Int 83:548–550
Zhou YS, Webb S, Lettice L, Tardif S, Kilanowski F, Tyrrell C, Macpherson H, Semple F, Tennant
P, Baker T, Hart A, Devenney P, Perry P, Davey T, Barran P, Barratt CL, Dorin JR (2013) Partial
deletion of chromosome 8 beta-defensin cluster confers sperm dysfunction and infertility in
male mice. PLoS Genet 9:e1003826
Zucht HD, Grabowsky J, Schrader M, Liepke C, Jurgens M, Schulz-Knappe P, Forssmann WG
(1998) Human beta-defensin-1: a urinary peptide present in variant molecular forms and its
putative functional implication. Eur J Med Res 3:315–323
Chapter 5
Antimicrobial Peptides in the Gut
Abstract The gut represents a unique interface toward our environment. It not only
facilitates digestion and resorption but also battles ingested pathogens, while also
controlling an immense community of commensal microorganisms. To aid with the
latter, it produces a wide range of innate immune mediators, such as antimicrobial
peptides (AMPs), which can combat viruses, bacteria, and fungi. Gut AMPs have
differing activity ranges and modes of action, so their expression varies depending
on the present conditions and threats. The most famous examples for site-specific
AMPs are probably the two α-defensins HD5 and HD6. In a homeostatic state, they
are exclusive to the Paneth cells of the small intestine. Since the importance of gut
microbiota has become more and more evident, research on AMPs has also
increased. This is particularly obvious in the case of inflammatory bowel diseases,
but also noticeable in other disorders. Defects in the AMP machinery have been
linked to increased susceptibility to infections, chronic inflammation, and distur-
bances in commensal composition. Recently, even a role in colon cancer has been
proposed. The gut provides a complex and challenging environment for the study of
interactions between AMPs and microbes; and while we are now widely aware of
their crucial role in keeping us healthy, more research is needed to fully uncover the
involved multi-level crosstalk of their actions. Such investigations might one day
help us in fully understanding the mechanisms of various diseases. Even more, they
might aid in developing new anti-infectious, antiinflammatory, and maybe even
antitumorigenic drugs.
M.J. Ostaff
Dr. Margarete Fischer Bosch Institute of clinical pharmacology and University of Tübingen
E.F. Stange
Department of Gastroenterology, Robert Bosch Hospital, Stuttgart, Germany
J. Wehkamp (*)
Department of Internal Medicine I, University Hospital Tuebingen,
Otfried-Müller-Straße 10, Tuebingen 72076, Germany
e-mail: [email protected]
5.1 Introduction
The human gut has always been a fascinating topic for scientists and laypersons
alike. We tend to trust our proverbial gut instinct and sometimes wallow in hard to
digest life lessons. We all have experienced moments when our gut gravely interfered
with daily routines, whether it was an exam-related nervous stomach, a bad onset of
abdominal influenza, food poisoning, travelers’ diarrhea, or indigestion after enjoy-
ing food that was simply not good for us. A particularly recent advance in the field
involves an increasing awareness of the importance of commensal microbiota. While
early medical research focused primarily on pathogens, contemporary studies often
favor analyses on the role of regular microbes as drivers of gut health or disease.
Since AMPs are tightly involved in regulating the composition of these gut inhabit-
ants, studies on their expression and functional role have also experienced somewhat
of a renaissance. The following paragraphs will now aim at discussing this excep-
tional host microbe situation in the gut and the role of AMP in mediating health and
disease in this context.
Our gastrointestinal (GI) tract includes the mouth, esophagus, stomach, as well as
the small and large intestine ending with the rectum and anus. All these parts per-
form specific tasks aimed at an optimized uptake of nutrients, while the prevention
of inflammation and infection by food-borne and resident microbes has to be
ensured. The GI system consequently represents a particularly tightly controlled
host–microbe interface and is characterized by distinct expression patterns of vari-
ous antimicrobial peptides. In the mouth, bacteria can form a structurally and func-
tionally organized biofilm designated as dental plaque. While distinct interindividual
differences in the microbial composition among healthy individuals exist, within
one person, the oral microbiota can remain relatively stable over time. Thereby,
Firmicutes, Proteobacteria, Bacteroidetes, and Actinobacteria seem to be the most
abundant phyla (Bik et al. 2010). When however a shift in one of the critical param-
eters controlling this ecological homeostasis occurs, disease can follow. One of the
most prevalent oral diseases is caries, and clinical studies have associated it with an
increase in acid-forming or acid-tolerating bacteria, especially streptococci and
lactobacilli, which can demineralize enamel (Marsh 2006). The adjacent GI com-
ponent, the esophagus, forms an exception in the gut, as it is not meant to retain
food. However, it is nonetheless characterized by a distinct microbial community.
While it’s most abundant phyla, Firmicutes, Proteobacteria, Bacteroidetes, and
Actinobacteria, are shared with the oral cavity, and it also displays a similar level
of complexity, the esophagus also shows specific features, e.g., significantly higher
5 Antimicrobial Peptides in the Gut 69
amounts of Prevotella, whereas Neisseria appear less frequent (Fillon et al. 2012).
The stomach represents the first GI component which holds ingested food for some-
what longer periods of time. It should consequently provide sound conditions for
microbial growth. However, the high acidity in gastric juices generates a unique
ecological environment which limits bacterial survival. While early on it was
thought that the gastric acid kills indeed all entering bacteria, leaving it unsuitable
for colonization, later studies unveiled large numbers of acid-resistant bacteria,
stemming from the transient oral and food-borne microbiota, including Streptococci,
Neisseria, and Lactobacilli (Wang and Yang 2013). A 2006 study by Bik et al. had
already detected 128 different phylotypes among 8 bacterial phyla, dominated by
Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria, suggesting a much
greater diversity within this ecosystem than previously described (Bik et al. 2006).
Different from the stomach, the presence and importance of colonizing microbiota
in the small and large intestines has been acknowledged early on. A role in, for
instance, the development of a proper intestinal morphology and function was
already accepted in the 1970s (Thompson and Trexler 1971) based on evidence
from different germ-free or gnotobiotic animal models. An influence of bacteria has
also been implicated in epithelial cell renewal rates (Heitman et al. 1980; Sun et al.
2004) and the development of Peyer’s patches (ileum-specific immune tissue) and
other gut-associated lymphoid tissue (Hooper and Gordon 2001). Intestinal bacteria
are involved in optimal food utilization, by degrading nutrients, aiding in digestion,
absorption, and vitamin synthesis while also inhibiting pathogen growth (Saulnier
et al. 2009; Hooper and Gordon 2001). In return, the intestine offers its commensals
an ideal habitat with constant temperatures and a never ending nutritional supply.
The generation of a beneficial microbial symbiosis is essential for the health in the
intestinal tract, but it can also influence peripheral bodily functions and even impact
on the brain (Sommer and Backhed 2013; Stilling et al. 2014). Most intestinal bac-
teria belong to the phyla Bacteroidetes or Firmicutes, but their numbers and com-
position vary locally and are in large parts dependent on the present local immune
defense strategies. While the ileum, for example, contains a somewhat fairly large
amount of aerobic and anaerobic bacteria such as Enterobacter and Lactobacilli,
the most densely populated intestinal region is found in the colon with numbers of
up to 1012 bacteria per gram of luminal content and predominately anaerobic repre-
sentatives such as Bacteroides, Bifidobacteria, Fusobacteria, Clostridium, and
Peptostreptococci (Sears 2005).
Many recent studies try to map and understand the relationship between the host
and its intestinal microbiota. Similar to what has been found in studies on oral plaque,
their composition is varying between individuals, especially when comparing groups
from different geographical locations, but it is also shifting among one individual,
depending on age (Yatsunenko et al. 2012). Additional environmental factors, such as
a specific nutritional lifestyle, or certain medications, can further impact on microbial
composition (David et al. 2014; Ji et al. 2014; Voreades et al. 2014). Due to its high
variability and dynamic, defining an “ideal gut microbiota” is challenging.
Nonetheless, many diseases, including cancer, obesity, autism, and, above all,
70 M.J. Ostaff et al.
inflammatory bowel diseases (IBD), have been associated with more or less distinct
alterations (Uronis et al. 2009; Sartor 2014; Turnbaugh et al. 2009; Sekirov et al.
2010). The reliability of some of these associations seems however greatly dependent
on the effect scope, study design, sampling, and analysis (Hanage 2014). Changes in
the microbiota in IBD patients however show a high effect size with robust observa-
tions reported by several independent groups (Frank et al. 2011; Gophna et al. 2006;
Walker et al. 2011; Sartor 2014). While mostly a reduced overall diversity and an
increase in Proteobacteria with a parallel decrease in Firmicutes are seen, more stud-
ies are indispensable until microbiota might be considered a possible diagnostic tool
or a promising therapeutic target in the treatment of this disorder (Matsuoka and
Kanai 2015).
In general, even in health, the GI tract is constantly challenged by a remarkable
and diverse community of commensals. It is therefore surprising that infections or
bacterial translocations are rather rare and limited to highly pathogenic strains or
predisposing disease states. This is in big parts to the effective generation of a highly
competent arsenal of antimicrobial peptides (AMP) with activity against bacteria,
viruses, protozoa, and fungi, reflecting the different threats and challenges met at GI
mucosae. Most of these AMP, such as β-defensins, are thereby generated by gut
epithelial cells themselves, but some are also contributed by specialized resident
immune cells.
Many members of the gastrointestinal AMP arsenal are at least in part dependent
on so-called pattern recognition receptors (PRR) for either their transcriptional
induction or their secretion. PRRs include, among others, the outer membrane Toll-
like receptors (TLRs) and the intracellular NOD-like receptors (NLR) and detect
highly conserved microbial structures, called pathogen- (or microbial)-associated
molecular pattern (PAMP or MAMP). Upon binding their ligand, PRRs can activate
innate immune reactions, which often occur via MyD88 and NF-κB or MAP kinases
(Abreu et al. 2005). These events then lead to a proinflammatory response but also
the release of stored AMP and/or their de novo synthesis (Takeda and Akira 2004;
Bevins 2003). A relatively low expression of some TLRs in the gut, as well as a
basolateral location upon epithelial cells of others, allows an effective recognition
of invading pathogens, but also tolerance toward commensals without generating
constant inflammatory conditions (Abreu 2010; Cario 2010). The following chap-
ters will take a closer look on the different AMPs found in the gut and how their
expression and function is related to different gastrointestinal disorders.
The upper gastrointestinal tract, for all intents and purposes, includes the esopha-
gus, the stomach, and the duodenum. However, in the framework of this chapter,
when speaking of the upper GI tract, it is referred to the mouth, esophagus, and
stomach. The duodenum is included below when discussing the situation present in
the small intestine.
5 Antimicrobial Peptides in the Gut 71
Starting with the oral cavity, major AMPs, which control the microbial environ-
ment in this compartment, include human β-defensins (hBDs), histatins, the cathe-
licidin LL-37, the antifungal and antibacterial lactoferrin, as well as lysozyme. As
a whole, they not only limit an overgrowth of commensal microorganisms but also
prevent colonization by pathogens (Melino et al. 2014; van’t Hof et al. 2014; Hans
and Madaan 2014). While the β-defensin hBD-4 has not been reported to be
expressed in oral epithelial cells, the constitutively expressed hBD-1, as well as
the inducible hBD-2 and hBD-3, can be found in oral mucosa, gingiva, and tongue
epithelia as well as in salivary glands (Hans and Madaan 2014). The latter two
hBDs are thereby upregulated during infection, as it can occur in gingivitis and
periodontitis, and are also involved in response to regular plaque-forming bacteria.
A recent study by Wang et al. could, for instance, show that the bacteria found in
oral biofilms from healthy individuals have the potential to induce antibacterial or
immunomodulatory reactions in oral epithelial cells during early stages of bacte-
ria–host interactions in a distinct species-dependent way. This includes the asso-
ciation of a transcriptional upregulation of hBD-2 and hBD-3 with an abundance
of Prevotella and the absence of species of the Streptococcus cluster (Wang et al.
2015). One mechanism by which oral bacteria have been reported to mediate an
upregulation of inducible β-defensins is based on cytokine-mediated pathway acti-
vation, e.g., via IL-17 signaling, which has an important role in defending the oral
cavity against Candida albicans infections (Trautwein-Weidner et al. 2015). C.
albicans can cause oral candidiasis in immune-deficient patients, as it is, for
instance, the case in human immunodeficiency virus (HIV) carriers (Samaranayake
et al. 2009). Aside from a functional involvement of β-defensins, which has been
demonstrated in mouse models (Tomalka et al. 2015), lysozyme and lactoferrin
can also play an important role in limiting oral C. albicans growth (Samaranayake
et al. 2001). Similar reports also exist for histatins, which are secreted by the
parotid and submandibular salivary glands and are found in saliva in high concen-
trations. Their antimicrobial function is based on multiple modes of actions includ-
ing the induction of a non-lytic ATP loss from actively respiring cells, the
disruption of the cell, as well as the generation of reactive oxygen species, which
can all lead to cell death (Kavanagh and Dowd 2004). Interestingly, C. albicans
however does not necessarily submit to oral AMPs without a fight. Several resis-
tance mechanisms have been reported which can deter antifungal activities. One is
the secretion of a Msb2 glycoprotein, that can function as a broad-range anti-AMP
protectant; another is the use of efflux transporters to shuttle AMP out of the cell,
while an adaptive modification of the fungal membrane can also convey resistance
against membrane-interacting AMPs such as hBD-2 (Swidergall and Ernst 2014).
In addition to oral manifestations, Candida infections can also target the
esophagus.
Here again, a role for β-defensins, in particular the inducible hBD-2 and hBD-3,
in response to an autocrine IL-1β loop and an activation of the epidermal growth
factor receptor (EGFR) by endogenous transforming growth factor-α (TGF-α) have
been reported (Pahl et al. 2011). Their upregulation might thereby help in protecting
the host against invasive candidiasis by limiting the infection to the mucosal surface
72 M.J. Ostaff et al.
Regarding its antimicrobial peptide defense, the small intestine holds an exceptional
position. This metabolically most active GI compartment is characterized by a
largely puckered surface which contains invaginations, called crypts of Lieberkühn,
and protrusions, known as villi; both are thereby crucial in enlarging the mucosal
surface to optimize nutrient resorption but also offer up a particularly large host–
microbe interface. While the villi are more leaflike in the duodenum, they appear in
fingerlike structures in the ileum (Helander and Fandriks 2014). It is important to
acknowledge that the bacterial density rises steadily from the proximal duodenum,
over the jejunum in the middle, to the distal ileum. This is accompanied by an
increasing number of Paneth cells toward the ileum. Paneth cells are important
secretory specialists and specific to the small intestine, where they are found at the
bottom of crypts. They generate a wide arsenal of antimicrobial factors making
them crucial players in the regulation of gut microbiota while also mediating defense
5 Antimicrobial Peptides in the Gut 73
against pathogens (Clevers and Bevins 2013). Expressing the human α-defensins
HD5 and HD6, as well as lysozyme, the lectin Reg3γ and phospholipase A2 group
IIA (sPLA2, also known as PLA2G2A), among others, the Paneth cell represents
the main supplier of small intestinal AMP. To a smaller extent, other small intestinal
epithelial cells deliver additional mediators like hBD-1, hBD-2, hBD-3, elafin, and
LL-37.
Paneth cells store AMPs in vesicles and secrete them mainly upon bacterial
stimulation, allowing enteric defensin concentrations of up to 15–100 mg/ml in the
crypt of mice models (Ayabe et al. 2000; Selsted and Ouellette 2005) or 50–250 μg/
ml of HD5 alone, in human studies (Ghosh et al. 2002). This outreaches an effec-
tive antimicrobial concentration by far and ensures a sterile crypt microenviron-
ment (Ericksen et al. 2005; Ouellette and Bevins 2001). The highly conserved
Paneth cell α-defensin genes include two exons and are produced as prepro-defen-
sins, meaning they initially include a signaling sequence and a pro-segment, which
is promptly separated. The still inactive propeptide is stored in granules and acti-
vated upon release by targeted cleavage (Bevins and Salzman 2011). With respect
to their genetic organization, HD5 and HD6 exhibit a relatively similar 5′- putative
promoter region, sharing several similar transcription factor binding sites, includ-
ing multiple Wnt response elements (Beisner et al. 2014). This might explain the
almost parallel regulation of HD5 and HD6 in many setups. It was indeed shown
by several studies that the Wnt pathway can directly impact on HD5 and HD6
expression, while it is interestingly also directly involved in Paneth cell maturation
(Ostaff et al. 2013; Andreu et al. 2008). Even though they are transcriptionally
regulated in a similar way, the mature Paneth cell α-defensin peptides differ greatly
in their antimicrobial function. HD5 is a very potent defense mediator with high
activity at low concentrations against several bacteria, fungi, and viruses (Bevins
and Salzman 2011). It furthermore possesses chemotactic properties (Grigat et al.
2007). Using transgenic HD5 mice, a direct influence on the composition of intes-
tinal microbiota and a direct effect against invading Salmonella could be demon-
strated (Salzman et al. 2003, 2010). Less is however known about its mode of
action. It was shown to form dimers, and, depending on its amphipathic structure,
it likely kills bacteria by disrupting their cell wall (Clevers and Bevins 2013).
Moreover, HD5 is able to inactivate bacterial toxins, probably by their unfolding,
making them susceptible to proteolysis (Kudryashova et al. 2014). The second
enteric α-defensin, HD6, has not yet been as rigorously studied. Although it was
shown to exhibit antiviral properties (Doss et al. 2009), its antimicrobial activity
has long been evasive. Quite recently, Bevins and his colleagues discovered that
HD6 can form nanonets in vivo, which actively trap bacteria (Chu et al. 2012), a
strategy similar to what has been reported on neutrophil defensins (Brinkmann and
Zychlinsky 2007). Furthermore, it was shown that under reducing conditions – as
present in the gut – HD6 also possesses antimicrobial properties aimed at commen-
sals (Schroeder et al. 2015).
The study of mice models allowed the elucidation of essential functions of
Paneth cells in fending of commensal but also pathogenic threats. A lack of
Paneth cell α-defensin (termed cryptdins in mice)-activating enzyme promotes a
74 M.J. Ostaff et al.
Related to microbes, the large intestine represents the most densely populated
region in the human body. It is therefore not surprising that it possesses a thick,
complex mucus barrier formed by a high amount of resident goblet cells that helps
to shield its epithelium. Different from a somewhat thin and much looser layer
found in the small intestine, it is made up by a dual system. The two colonic mucus
layers encompass an adherent film as well as a mobile coating, which is continu-
ously shuffled distally, ensuring optimized passage of luminal content, potential
harmful substances, but also microorganisms which might colonize it. Of note, the
inner colon mucus layer is thereby rapidly renewed and converted into the outer one
by a host controlled endogenous proteolytic process (Johansson et al. 2011). The
glycoprotein Muc2 is the most important protein component of these layers, and a
genetic ablation of this mucin leads to a spontaneous and chronic inflammation in
mice which resembles ulcerative colitis in humans (Wenzel et al. 2014). An impor-
tant function of colonic mucus is the retention of AMPs, which ensures comparably
high concentrations of these important innate immune mediators in close proximity
to the epithelial surface (Antoni et al. 2013).
Colonic epithelia lack the villus structures found in the small intestine, but crypts
are present, which normally do not sport Paneth cells. Hence, a multitude of non-
Paneth cell epithelial antimicrobials play a front role in ensuring a tight control of
colonic commensals. Among these are LL-37, elafin, as well as another antiprote-
ase, the secretory leukocyte protease inhibitor SLPI, and ß-defensins. β-defensins,
while sharing common properties, such as most AMPs (positive charge, disulfide
bonds) (Boman 2003), exhibit vast differences regarding their antimicrobial effec-
tiveness and in their modes of action. They share this apparent paradox with HD5
and HD6, which, as discussed above, also display quite differing functions. A strik-
ing example within the family of ß-defensins is provided by hBD-1. hBD-1 has
previously shown to exhibit only weak antimicrobial activity as compared to hBD-2
or others. The reason for this perceived lack of function became evident in the work
of Schroeder et al. Thereby, a quite strong activity against various commensal gut
bacteria and the facultative pathogenic fungus C. albicans only emerged after a
biochemical activation in a reducing environment, as it is, for instance, present in
the colon (Schroeder et al. 2011). In addition, a physiological activation via the
thioredoxin system has also been demonstrated using the colonic epithelial cell line
Caco-2 (Jaeger et al. 2013).
The important role of colonic AMPs has been demonstrated in mouse models.
An example for a direct link between antimicrobial function and colonic health
could be demonstrated in mice lacking cathelicidin (mCRAMP in mice, LL-37 in
humans), which is normally upregulated during colitis, but also in ileitis, depending
on the level of inflammation (Kubler et al. 2009; Schauber et al. 2006; Koon et al.
2011). The cathelicidin knockout model strikingly showed more severe symptoms
and mucosal disruption in a dextran sodium sulfate (DSS) chemically induced coli-
tis approach (Koon et al. 2011). Interestingly, the different abnormalities in this
76 M.J. Ostaff et al.
as it is normally found in a healthy gut (Fig. 5.1), as the main disease driver (Ostaff
et al. 2013). Intestinal epithelia of IBD patients consequently display mucosal adher-
ent bacteria, activated T cells, and antibodies aimed not toward pathogens but regular
gut inhabitants (MacPherson et al. 1996; Sartor 2001; Duchmann et al. 1995).
Regarding to genetics, the intracellular PRR NOD2, also known as CARD15, a
receptor for muramyl dipeptide, a minimal bioactive peptidoglycan motif common to
all bacteria, represents the foremost CD susceptibility factor (Hugot et al. 2001;
Ogura et al. 2001; Girardin et al. 2003). Its association with the disease was the first
major genetic mechanism underlining the importance of innate defenses and a crucial
role of bacteria–host interactions. NOD2 mutations were later specifically associated
with the clinical phenotype of small intestinal (or ileal) Crohn’s disease. Since NOD2
is mainly expressed in Paneth cells (Lala et al. 2003) and, as discussed above,
involved in Paneth cell-mediated small intestinal antimicrobial defense, this location-
specific association appears plausible.
The pathogenesis of small intestinal (or more specific ileal) CD is, to a substan-
tial extent, based on defects in the Paneth cell. When comparing ileal CD patients to
healthy individuals, but also patients with colonic CD or UC, both Paneth cell
α-defensins show reduced expression levels. In contrast, other Paneth cell products,
such as lysozyme and sPLA2, seem to be unchanged (Wehkamp et al. 2005). The
specific reduction of Paneth cell HD5 and HD6 appears to be particularly marked in
CD patients with a NOD2 frameshift mutation, while other NOD2 SNPs seem to be
inconsequential in this regard (Bevins et al. 2009; Wehkamp et al. 2004, 2005).
Another functional single nucleotide polymorphism (SNP) has also been associated
with particularly low Paneth cell defensin levels. It is present in the Wnt coreceptor
LRP6, which showed a genetic association with early-onset ileal CD (Koslowski
et al. 2012). The intensified decrease in both cases, NOD2 and LRP6, has been
found to be independent from current inflammation. Additional mechanisms under-
lying the reduced α-defensin expression also affect the Wnt pathway. Patients with
ileal CD show diminished mRNA expression of the transcription factor TCF-4, the
Wnt coreceptor LRP6, and also the Wnt pathway transcription factor TCF1
(Koslowski et al. 2012; Beisner et al. 2014; Wehkamp et al. 2007). While also an
SNP in the TCF4 promoter showed an association with ileal CD (Koslowski et al.
2009), a causal relationship between Wnt/TCF-4 and α-defensin levels could be
underlined in a knockout mouse model. In these animals, a 50 % reduction of TCF-4
mRNA promoted a significant drop of Paneth cell cryptdins and subsequent dimin-
ished antibacterial activity (Wehkamp et al. 2007). Aside from controlling the
expression of various Paneth cell genes, among them HD5 and HD6, Wnt also
plays, as mentioned, an essential role in the maturation of these specialized secre-
tory cells, while also governing the proliferation of intestinal epithelia. An involve-
ment of important Wnt pathway components in ileal CD supports the idea that a
disrupted Paneth cell differentiation and a disturbed epithelial regenerative homeo-
stasis might potentially be relevant in disease pathogenesis. Aside from NOD2,
TCF-4, and LRP6, the autophagy factor ATG16L1 (autophagy-related protein 16-1)
is another genetically associated factor with an important role for Paneth cell func-
tion. ATG16L1 is involved in cellular recycling processes which are important for
cell and tissue homeostasis. In a genome-wide association study, an SNP in
ATG16L1 was associated with CD and preceded changes in Paneth cell granules in
homozygous carriers. Similar aberrations were thereby also present in ATG16L1
hypomorphic mice (Cadwell et al. 2008). An additional study in pediatric patients
furthermore found activation of autophagy in Paneth cells, irrespective of inflamma-
tion and independent of the disease-associated variants (Thachil et al. 2012). A
deregulated autophagy might therefore be an additional and general characteristic in
ileal CD with a particular impact on Paneth cell biology.
In small intestinal CD, the involvement of Paneth cell defects clearly highlights
a role for antimicrobial defense defects in patients. The colonic manifestation of
the disease also shows distinct AMP-related aberrations, such as an attenuated
induction of hBD-2, as well as an inflammation-related decrease in hBD-1,
although the respective mechanisms are generally less well understood. While in
the subgroup of UC, colonic expression levels of hBD-2 are readily induced on a
high extent during inflammation, CD patients show a much lower upregulation of
this important defense mediator in active disease (Wehkamp et al. 2003). This has
originally been linked to lower copy numbers of the hBD-2 gene (Fellermann et al.
2006), but multiple reports could not confirm or rather contradicted this association
(Bentley et al. 2009; Aldhous et al. 2009). However, based on functional studies,
investigating the antimicrobial killing activity of mucosal protein extracts against
E. coli, Nuding et al. could clearly show that, while antibacterial activity was
majorly enhanced in UC, colonic CD patients have a severely inhibited killing
80 M.J. Ostaff et al.
capacity against this gut bacterium (Nuding et al. 2007). Additional inducible
AMPs associated with a lower inducibility in CD are the cathelicidin LL-37 and
the antimicrobial serine protease inhibitors elafin and SLPI (secretory leukocyte
protease inhibitor). The constitutively expressed hBD-1 on the other hand shows
reduced levels in the colon of IBD in general, which is however especially pro-
nounced in colonic CD (Peyrin-Biroulet et al. 2010). Interestingly, this might partly
be explained by a functional promoter SNP that has previously been associated
with protection from colonic CD in a European cohort (Peyrin-Biroulet et al. 2010).
Furthermore, thioredoxin, an enzyme involved in the redox activation of hBD-1,
also exhibits a reduction in active colonic CD, but this can however also be seen in
UC patients (Jaeger et al. 2013).
While as mentioned, general mucosal production of antimicrobial factors seems
to be enhanced in UC (Nuding et al. 2007), patients are nonetheless characterized
by a diminished epithelial barrier protection. This is related to distinct aberrations
in the mucus layer. As previously discussed, one important function of colonic
mucus is the retention of AMPs, keeping them close to the mucosa to ensure that
commensal bacteria cannot attach to and/or translocate over the intestinal barrier.
Specific detrimental changes of the mucus structure, regarding a reduction in thick-
ness and continuity, have been repeatedly described in UC patients (Johansson et al.
2013). It is easy to imagine how such aberrations can considerably weaken the
mucosal barrier. Acute inflamed areas have even been described to be sometimes
completely depleted of mucus. In contrast, patients with CD show a comparable or
even thicker mucus layer than what is seen in healthy controls. Additional changes
in UC patients related to mucus promote a simplification of mucin glycosylation
patterns, potentially allowing it to be easily degradable by bacterial enzymes and
also include alterations in the content of phosphatidylcholine and lysophosphatidyl-
choline (Antoni et al. 2014). Whether such changes are primary or secondary, with
regard to the presence of inflammation, has not yet been definitively determined.
While some studies have shown an inflammation dependency of mucus-related
alterations in active versus non-active UC, genetic links also highlight that the
observed effects are likely not an exclusive result of continuous inflammatory pro-
cesses. A higher susceptibility for UC was, for instance, found in carriers of rare
alleles in the MUC3 gene (Kyo et al. 1999), and an association of MUC4 and
MUC13 variants with ulcerative colitis has also been described (Moehle et al.
2006). It was furthermore demonstrated that UC patients show an impairment in the
inducibility of certain secretory and especially goblet cell differentiation factors
when compared with active CD. A significant inflammation-related upregulation of
the secretory differentiation factors Hath1 and the goblet cell differentiation factor
KLF4, as it can be seen in active CD, is absent in ulcerative colitis (Gersemann et al.
2009).
Taken together, in UC a diminished mucus layer provides a less effective reser-
voir for AMPs and a reduced protection against bacterial invasion, while in CD,
distinct impairments related to the production of AMPs disturb the host–microbe
balance at the epithelial barrier (Fig. 5.2). The underlying mechanisms of IBD-
related barrier defects are various and very complex. We are certainly only begin-
5 Antimicrobial Peptides in the Gut 81
Fig. 5.2 An impaired barrier, as it, for instance, occurs when AMP expression is diminished, and
a dysbiosis of commensals can lead to an imbalanced host–microbe interaction. This can promote
the activation of ongoing and over-shooting inflammatory immune responses, which characterize
inflammatory bowel diseases
ning to understand all the detailed aspects, so the study of interactions between the
different factors and their control, as well as environmental influences, is sure to
provide long-ranging potential for scientific research. A better understanding of the
mucosal barrier, and in particular the role of AMPs, will however be the key to open
up new therapeutic avenues.
References
Abreu MT (2010) Toll-like receptor signalling in the intestinal epithelium: how bacterial recogni-
tion shapes intestinal function. Nat Rev Immunol 10:131–144
Abreu MT, Fukata M, Arditi M (2005) TLR signaling in the gut in health and disease. J Immunol
174:4453–4460
Aldhous MC, Noble CL, Satsangi J (2009) Dysregulation of human beta-defensin-2 protein in
inflammatory bowel disease. PLoS One 4:e6285
Amendola A, Butera A, Sanchez M, Strober W, Boirivant M (2014) Nod2 deficiency is associated
with an increased mucosal immunoregulatory response to commensal microorganisms.
Mucosal Immunol 7:391–404
Ananthakrishnan AN (2015) Epidemiology and risk factors for IBD. Nat Rev Gastroenterol
Hepatol 12(4):205–217
Andreu P, Peignon G, Slomianny C, Taketo MM, Colnot S, Robine S, Lamarque D, Laurent-Puig
P, Perret C, Romagnolo B (2008) A genetic study of the role of the Wnt/beta-catenin signalling
in Paneth cell differentiation. Dev Biol 324:288–296
Antoni L, Nuding S, Wehkamp J, Stange EF (2014) Intestinal barrier in inflammatory bowel dis-
ease. World J Gastroenterol 20:1165–1179
Antoni L, Nuding S, Weller D, Gersemann M, Ott G, Wehkamp J, Stange EF (2013) Human
colonic mucus is a reservoir for antimicrobial peptides. J Crohns Colitis 7:e652–e664
Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ,
Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO,
Fodor AA, Jobin C (2012) Intestinal inflammation targets cancer-inducing activity of the
microbiota. Science 338:120–123
82 M.J. Ostaff et al.
Ayabe T, Satchell DP, Wilson CL, Parks WC, Selsted ME, Ouellette AJ (2000) Secretion of micro-
bicidal alpha-defensins by intestinal Paneth cell response to bacteria. Nat Immunol 1(2):
113–118
Bauer B, Wex T, Kuester D, Meyer T, Malfertheiner P (2013) Differential expression of human
beta defensin 2 and 3 in gastric mucosa of Helicobacter pylori-infected individuals. Helicobacter
18:6–12
Beisner J, Teltschik Z, Ostaff MJ, Tiemessen MM, Staal FJ, Wang G, Gersemann M, Perminow G,
Vatn MH, Schwab M, Stange EF, Wehkamp J (2014) TCF-1 mediated Wnt Signaling regulates
Paneth cell innate immune defense effectors HD-5 and −6: implications for Crohn’s disease.
Am J Physiol Gastrointest Liver Physiol 307(5):G487–G498
Bentley RW, Pearson J, Gearry RB, Barclay ML, McKinney C, Merriman TR, Roberts RL (2009)
Association of higher DEFB4 genomic copy number with Crohn’s disease. Am J Gastroenterol
105(2):354–359
Bevins CL (2003) Antimicrobial peptides as effector molecules of mammalian host defense.
Contrib Microbiol 10:106–148
Bevins CL, Salzman NH (2011) Paneth cells, antimicrobial peptides and maintenance of intestinal
homeostasis. Nat Rev Microbiol 9:356–368
Bevins CL, Stange EF, Wehkamp J (2009) Decreased Paneth cell defensin expression in ileal
Crohn’s disease is independent of inflammation, but linked to the NOD2 1007fs genotype. Gut
58:882–883
Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, Perez-Perez G, Blaser MJ,
Relman DA (2006) Molecular analysis of the bacterial microbiota in the human stomach. Proc
Natl Acad Sci U S A 103:732–737
Bik EM, Long CD, Armitage GC, Loomer P, Emerson J, Mongodin EF, Nelson KE, Gill SR,
Fraser-Liggett CM, Relman DA (2010) Bacterial diversity in the oral cavity of 10 healthy indi-
viduals. ISME J 4:962–974
Boman HG (2003) Antibacterial peptides: basic facts and emerging concepts. J Intern Med 254:
197–215
Brinkmann V, Zychlinsky A (2007) Beneficial suicide: why neutrophils die to make NETs. Nat
Rev Microbiol 5:577–582
Cadwell K, Liu JY, Brown SL, Miyoshi H, Loh J, Lennerz JK, Kishi C, Kc W, Carrero JA, Hunt S,
Stone CD, Brunt EM, Xavier RJ, Sleckman BP, Li E, Mizushima N, Stappenbeck TS, Virgin
HW (2008) A key role for autophagy and the autophagy gene Atg16l1 in mouse and human
intestinal Paneth cells. Nature 456:259–263
Cario E (2010) Toll-like receptors in inflammatory bowel diseases: a decade later. Inflamm Bowel
Dis 16:1583–1597
Celli JP, Turner BS, Afdhal NH, Keates S, Ghiran I, Kelly CP, Ewoldt RH, McKinley GH, So P,
Erramilli S, Bansil R (2009) Helicobacter pylori moves through mucus by reducing mucin
viscoelasticity. Proc Natl Acad Sci U S A 106:14321–14326
Chu H, Pazgier M, Jung G, Nuccio SP, Castillo PA, de Jong MF, Winter MG, Winter SE, Wehkamp
J, Shen B, Salzman NH, Underwood MA, Tsolis RM, Young GM, Lu W, Lehrer RI, Baumler
AJ, Bevins CL (2012) Human alpha-defensin 6 promotes mucosal innate immunity through
self-assembled peptide nanonets. Science 337:477–481
Clevers HC, Bevins CL (2013) Paneth cells: maestros of the small intestinal crypts. Annu Rev
Physiol 75:289–311
David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS,
Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ (2014) Diet rapidly and
reproducibly alters the human gut microbiome. Nature 505:559–563
Dixon LJ, Kabi A, Nickerson KP, McDonald C (2015) Combinatorial effects of diet and genetics
on inflammatory bowel disease pathogenesis. Inflamm Bowel Dis 21(4):912–922
Doss M, White MR, Tecle T, Gantz D, Crouch EC, Jung G, Ruchala P, Waring AJ, Lehrer RI,
Hartshorn KL (2009) Interactions of alpha-, beta-, and theta-defensins with influenza A virus
and surfactant protein D. J Immunol 182:7878–7887
5 Antimicrobial Peptides in the Gut 83
Hodin CM, Lenaerts K, Grootjans J, de Haan JJ, Hadfoune M, Verheyen FK, Kiyama H, Heineman
E, Buurman WA (2011) Starvation compromises Paneth cells. Am J Pathol 179:2885–2893
Hooper LV, Gordon JI (2001) Commensal host-bacterial relationships in the gut. Science
292(5519):1115–1118
Hosaka Y, Koslowski M, Nuding S, Wang G, Schlee M, Schafer C, Saigenji K, Stange EF,
Wehkamp J (2008) Antimicrobial host defense in the upper gastrointestinal tract. Eur
J Gastroenterol Hepatol 20:1151–1158
Hugot J-P, Chamaillard C, Zouali H, Lesage S, Cezard J-P, Belaiche J, Almer S, Tysk C, O’Morain
CA, Gassull M, Binder V, Finkel Y, Cortot A, Modigliani R, Laurent-Puig P, Gower-Rousseau
C, Macry J, Colombel J-F, Sahbatou M, Thomas G (2001) Association of NOD2 leucine-rich
repeat variants with susceptibility to Crohn’s disease. Nature 411:599–603
Jaeger SU, Schroeder BO, Meyer-Hoffert U, Courth L, Fehr SN, Gersemann M, Stange EF,
Wehkamp J (2013) Cell-mediated reduction of human beta-defensin 1: a major role for muco-
sal thioredoxin. Mucosal Immunol 6(6):1179–1190
Ji Y, Sun S, Goodrich JK, Kim H, Poole AC, Duhamel GE, Ley RE, Qi L (2014) Diet-induced
alterations in gut microflora contribute to lethal pulmonary damage in TLR2/TLR4-deficient
mice. Cell Rep 8:137–149
Johansson ME, Larsson JM, Hansson GC (2011) The two mucus layers of colon are organized by
the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proc
Natl Acad Sci U S A 108(Suppl 1):4659–4665
Johansson ME, Sjovall H, Hansson GC (2013) The gastrointestinal mucus system in health and
disease. Nat Rev Gastroenterol Hepatol 10:352–361
Kavanagh K, Dowd S (2004) Histatins: antimicrobial peptides with therapeutic potential. J Pharm
Pharmacol 56:285–289
Kiehne K, Brunke G, Meyer D, Harder J, Herzig KH (2005) Oesophageal defensin expression
during Candida infection and reflux disease. Scand J Gastroenterol 40:501–507
Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nunez G, Flavell RA (2005)
Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science
307:731–734
Koon HW, Shih DQ, Chen J, Bakirtzi K, Hing TC, Law I, Ho S, Ichikawa R, Zhao D, Xu H, Gallo
R, Dempsey P, Cheng G, Targan SR, Pothoulakis C (2011) Cathelicidin signaling via the Toll-
like receptor protects against colitis in mice. Gastroenterology 141:1852–1863
Koprivnjak T, Peschel A (2011) Bacterial resistance mechanisms against host defense peptides.
Cell Mol Life Sci 68:2243–2254
Koslowski MJ, Kubler I, Chamaillard M, Schaeffeler E, Reinisch W, Wang G, Beisner J, Teml A,
Peyrin-Biroulet L, Winter S, Herrlinger KR, Rutgeerts P, Vermeire S, Cooney R, Fellermann K,
Jewell D, Bevins CL, Schwab M, Stange EF, Wehkamp J (2009) Genetic variants of Wnt tran-
scription factor TCF-4 (TCF7L2) putative promoter region are associated with small intestinal
Crohn’s disease. PLoS One 4:e4496
Koslowski MJ, Teltschik Z, Beisner J, Schaeffeler E, Wang G, Kubler I, Gersemann M, Cooney R,
Jewell D, Reinisch W, Vermeire S, Rutgeerts P, Schwab M, Stange EF, Wehkamp J (2012)
Association of a functional variant in the Wnt co-receptor LRP6 with early onset ileal Crohn’s
disease. PLoS Genet 8:e1002523
Kubler I, Koslowski MJ, Gersemann M, Fellermann K, Beisner J, Becker S, Rothfuss K, Herrlinger
KR, Stange EF, Wehkamp J (2009) Influence of standard treatment on ileal and colonic antimi-
crobial defensin expression in active Crohn’s disease. Aliment Pharmacol Ther
30(6):621–633
Kudryashova E, Quintyn R, Seveau S, Lu W, Wysocki VH, Kudryashov DS (2014) Human defen-
sins facilitate local unfolding of thermodynamically unstable regions of bacterial protein tox-
ins. Immunity 41:709–721
Kyo K, Parkes M, Takei Y, Nishimori H, Vyas P, Satsangi J, Simmons J, Nagawa H, Baba S, Jewell
D, Muto T, Lathrop GM, Nakamura Y (1999) Association of ulcerative colitis with rare VNTR
alleles of the human intestinal mucin gene, MUC3. Hum Mol Genet 8:307–311
Lala S, Ogura Y, Osborne C, Hor SY, Bromfield A, Davies S, Ogunbiyi O, Nunez G, Keshav S (2003)
Crohn’s disease and the NOD2 gene: a role for paneth cells. Gastroenterology 125:47–57
5 Antimicrobial Peptides in the Gut 85
Shen B, Porter EM, Reynoso E, Shen C, Ghosh D, Connor JT, Drazba J, Rho HK, Gramlich TL,
Li R, Ormsby AH, Sy MS, Ganz T, Bevins CL (2005) Human defensin 5 expression in intesti-
nal metaplasia of the upper gastrointestinal tract. J Clin Pathol 58:687–694
Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Van Tran NJ,
Furet JP (2011) Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One 6:e16393
Sommer F, Backhed F (2013) The gut microbiota–masters of host development and physiology.
Nat Rev Microbiol 11:227–238
Stilling RM, Bordenstein SR, Dinan TG, Cryan JF (2014) Friends with social benefits: host-
microbe interactions as a driver of brain evolution and development? Front Cell Infect
Microbiol 4:147
Sun J, Hobert ME, Rao AS, Neish AS, Madara JL (2004) Bacterial activation of beta-catenin sig-
naling in human epithelia. Am J Physiol Gastrointest Liver Physiol 287:G220–G227
Swidergall M, Ernst JF (2014) Interplay between Candida albicans and the antimicrobial peptide
armory. Eukaryot Cell 13:950–957
Tai EK, Wu WK, Wang XJ, Wong HP, Yu L, Li ZJ, Lee CW, Wong CC, Yu J, Sung JJ, Gallo RL,
Cho CH (2013) Intrarectal administration of mCRAMP-encoding plasmid reverses exacer-
bated colitis in Cnlp(−/−) mice. Gene Ther 20:187–193
Takeda K, Akira S (2004) TLR signaling pathways. Semin Immunol 16:3–9
Tan G, Li RH, Li C, Wu F, Zhao XM, Ma JY, Lei S, Zhang WD, Zhi FC (2015a) Down-regulation
of human enteric antimicrobial peptides by NOD2 during differentiation of the paneth cell
lineage. Sci Rep 5:8383
Tan G, Zeng B, Zhi FC (2015b) Regulation of human enteric alpha-defensins by NOD2 in the
Paneth cell lineage. Eur J Cell Biol 94:60–66
Teltschik Z, Wiest R, Beisner J, Nuding S, Hofmann C, Schoelmerich J, Bevins CL, Stange EF,
Wehkamp J (2012) Intestinal bacterial translocation in rats with cirrhosis is related to compro-
mised paneth cell antimicrobial host defense. Hepatology 55(4):1154–1163
Thachil E, Hugot JP, Arbeille B, Paris R, Grodet A, Peuchmaur M, Codogno P, Barreau F, Ogier-
Denis E, Berrebi D, Viala J (2012) Abnormal activation of autophagy-induced crinophagy in
paneth cells from patients with Crohn’s disease. Gastroenterology 142:1097–1099
Thompson GR, Trexler PC (1971) Gastrointestinal structure and function in germ-free or gnotobi-
otic animals. Gut 12:230–235
Tomalka J, Azodi E, Narra HP, Patel K, O’Neill S, Cardwell C, Hall BA, Wilson JM, Hise AG
(2015) Beta-defensin 1 plays a role in acute mucosal defense against Candida albicans.
J Immunol 194:1788–1795
Trautwein-Weidner K, Gladiator A, Nur S, Diethelm P, LeibundGut-Landmann S (2015) IL-17-
mediated antifungal defense in the oral mucosa is independent of neutrophils. Mucosal
Immunol 8:221–231
Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ,
Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI (2009) A core
gut microbiome in obese and lean twins. Nature 457:480–484
Uraki S, Sugimoto K, Shiraki K, Tameda M, Inagaki Y, Ogura S, Kasai C, Nojiri K, Yoneda M,
Yamamoto N, Takei Y, Nobori T, Ito M (2014) Human beta-defensin-3 inhibits migration of
colon cancer cells via downregulation of metastasis-associated 1 family, member 2 expression.
Int J Oncol 45:1059–1064
Uronis JM, Muhlbauer M, Herfarth HH, Rubinas TC, Jones GS, Jobin C (2009) Modulation of the
intestinal microbiota alters colitis-associated colorectal cancer susceptibility. PLoS One
4:e6026
van ’t Hof W, Veerman EC, Nieuw Amerongen AV, Ligtenberg AJ (2014) Antimicrobial defense
systems in saliva. Monogr Oral Sci 24:40–51
Voreades N, Kozil A, Weir TL (2014) Diet and the development of the human intestinal microbi-
ome. Front Microbiol 5:494
88 M.J. Ostaff et al.
Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, Rayment N, Brostoff J, Parkhill
J, Dougan G, Petrovska L (2011) High-throughput clone library analysis of the mucosa-
associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed
regions of the intestine in inflammatory bowel disease. BMC Microbiol 11:7
Wang P, Duan D, Zhou X, Li X, Yang J, Deng M, Xu Y (2015) Relationship between expression
of human gingival beta-defensins and levels of periodontopathogens in subgingival plaque.
J Periodontal Res 50:113–122
Wang ZK, Yang YS (2013) Upper gastrointestinal microbiota and digestive diseases. World
J Gastroenterol 19:1541–1550
Wehkamp J, Harder J, Weichenthal M, Mueller O, Herrlinger KR, Fellermann K, Schroeder JM,
Stange EF (2003) Inducible and constitutive beta-defensins are differentially expressed in
Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis 9:215–223
Wehkamp J, Harder J, Weichenthal M, Schwab M, Schaffeler E, Schlee M, Herrlinger KR,
Stallmach A, Noack F, Fritz P, Schroder JM, Bevins CL, Fellermann K, Stange EF (2004)
NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-
defensin expression. Gut 53:1658–1664
Wehkamp J, Salzman NH, Porter E, Nuding S, Weichenthal M, Petras RE, Shen B, Schaeffeler E,
Schwab M, Linzmeier R, Feathers RW, Chu H, Lima H Jr, Fellermann K, Ganz T, Stange EF,
Bevins CL (2005) Reduced Paneth cell alpha-defensins in ileal Crohn’s disease. Proc Natl
Acad Sci U S A 102:18129–18134
Wehkamp J, Schwind B, Herrlinger KR, Baxmann S, Schmidt K, Duchrow M, Wohlschlager C,
Feller AC, Stange EF, Fellermann K (2002) Innate immunity and colonic inflammation:
enhanced expression of epithelial alpha-defensins. Dig Dis Sci 47:1349–1355
Wehkamp J, Wang G, Kubler I, Nuding S, Gregorieff A, Schnabel A, Kays RJ, Fellermann K, Burk
O, Schwab M, Clevers H, Bevins CL, Stange EF (2007) The Paneth cell alpha-defensin defi-
ciency of ileal Crohn’s disease is linked to Wnt/Tcf-4. J Immunol 179:3109–3118
Wenzel UA, Magnusson MK, Rydstrom A, Jonstrand C, Hengst J, Johansson ME, Velcich A,
Ohman L, Strid H, Sjovall H, Hansson GC, Wick MJ (2014) Spontaneous colitis in Muc2-
deficient mice reflects clinical and cellular features of active ulcerative colitis. PLoS One
9:e100217
Wilson CL, Ouellette AJ, Satchell DP, Ayabe T, Lopez-Boado YS, Stratman JL, Hultgren SJ,
Matrisian LM, Parks WC (1999) Regulation of intestinal alpha-defensin activation by the
metalloproteinase matrilysin in innate host defense. Science 286:113–117
Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M,
Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J,
Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI (2012)
Human gut microbiome viewed across age and geography. Nature 486:222–227
Chapter 6
Metal Sequestration: An Important
Contribution of Antimicrobial Peptides
to Nutritional Immunity
Abstract First-row transition elements are essential for all forms of life. During
infection invading microbes must obtain these nutrients from their host. Vertebrates
take advantage of this fact to combat invaders by sequestering essential nutrients, a
defense known as nutritional immunity. The most well-characterized aspect of this
defense is the iron-withholding response. Advances in elemental imaging have
revealed that zinc and manganese are also sequestered during infection. The impor-
tance of nutritional immunity to host defense is emphasized by the increased sus-
ceptibility to infection when levels of these metals are elevated. This chapter will
discuss iron, zinc, and manganese availability during infection, the impact of with-
holding these metals from invading pathogens, and the antimicrobial peptides uti-
lized by the host to restrict the availability of these essential nutrients.
6.1 Introduction
Transition elements such as iron (Fe), zinc (Zn), and manganese (Mn) are essential
for all forms of life. This essentiality stems from their numerous structural and cata-
lytic roles. The importance of these metals to life is further emphasized by analyses
of protein databases, which suggest that 30 % of all proteins utilize a metal cofactor
(Andreini et al. 2008). Fe is highly versatile and is utilized by bacteria in numerous
ways including in mononuclear enzymes, such as ribonucleotide reductase and
superoxide dismutase; in Fe-S-containing enzymes, such as succinate dehydroge-
nase; and in heme-containing enzymes, such as cytochrome oxidase (Andreini et al.
2008; Py and Barras 2010; Mayfield et al. 2011). In bacteria, 4–6 % of all proteins
S. Damo
Department of Life and Physical Sciences, Fisk University, Nashville, TN, USA
T.E. Kehl-Fie, PhD ()
Department of Microbiology, University of Illinois Urbana-Champaign,
601 South Goodwin Avenue, MC-110, Urbana, IL 61801, USA
e-mail: [email protected]
Zn Zn
Epithelial Cells
Fe
Granules Fe
Fe
Fe
Fe Virulence Factors
Cytoplasm Zn
Zn Essential Processes
Mn
Zn
Neutrophils Pathogens
Inhibited
S100A7 S100A15
Processes
Calprotectin Transferrin
S100A12 Lactoferrin
Fig. 6.1 Antimicrobial peptides sequester iron, zinc, and manganese during infection
and hemopexin, respectively. Bacteria also express receptors that allow them to
utilize host Fe scavenging proteins including transferrin, haptoglobin, and hemo-
pexin directly as nutrient sources. To acquire Fe, bacteria also produce siderophores,
which have a higher affinity for Fe than transferrin. More recently it has become
apparent that the host produces proteins that bind bacterial siderophores, such as
lipocalin-2, preventing them from facilitating bacterial Fe acquisition. Lipocalin-2
is capable of binding Enterobactin and other similar siderophores (Goetz et al.
2002). An eight-stranded beta barrel protein, lipocalin-2, binds siderophores with
subnanomolar affinity using a highly specific binding site comprised of lysine and
arginine residues that interact tightly with the negatively charged siderophores
(Goetz et al. 2002). To counter this defense, pathogens express “stealth sidero-
phores,” which cannot be bound by lipocalin-2 (Abergel et al. 2006; Raffatellu et al.
2009). The numerous strategies employed by host and pathogen reinforce the impact
that wining the struggle for nutrient metals can have on the outcome of infection.
The observation that NRAMP1 removes Mn from the phagolysosome and contrib-
utes to controlling infection provided some of the first evidence that restricting Mn
availability contributes to host defense (Papp-Wallace and Maguire 2006). The use
of LA-ICP-MS has significantly expanded our knowledge regarding the scope of
Mn sequestration during infection. Similar to Zn, LA-ICP-MS revealed that the
staphylococcal abscess, but not the surrounding tissue, is rendered devoid of Mn
(Corbin et al. 2008; Kehl-Fie et al. 2013). The localized Mn sequestration does not
appear to impact total Mn levels in the organ (Kehl-Fie et al. 2013). Further investi-
gation revealed that calprotectin (CP) contributes to this restriction, as CP-deficient
mice do not remove Mn from staphylococcal liver abscesses (Corbin et al. 2008)
6 Antimicrobial Peptides and Nutritional Immunity 93
(Fig. 6.1). In addition to binding Zn, CP also binds Mn tightly, and the ability to
sequester this metal contributes to the antimicrobial activity of the protein (Damo
et al. 2013; Kehl-Fie et al. 2011). Mice that lack CP are more susceptible to bacterial
and fungal pathogens, including S. aureus, A. baumannii, Klebsiella pneumoniae,
and Candida albicans (Corbin et al. 2008; Kehl-Fie et al. 2013; Hood et al. 2012;
Achouiti et al. 2012). CP is not the only host protein that contributes to Mn seques-
tration, as CP-deficient mice still remove Mn from staphylococcal kidney abscesses
(Kehl-Fie et al. 2013). Similar to Fe and Zn, loss of Mn uptake systems in S.
typhimurium, B. abortus, Y. pestis, S. aureus, Streptococcus pneumoniae, and S.
pyogenes results in reduced virulence (Dintilhac et al. 1997; Berry and Paton 1996;
Anderson et al. 2009; Bearden and Perry 1999; Horsburgh et al. 2002; Janulczyk
et al. 2003; Sun et al. 2009; Kehl-Fie et al. 2013; Janakiraman and Slauch 2000).
The importance of sequestering Mn is highlighted by the observation that a staphy-
lococcal strain lacking Mn importers has a virulence defect in wild-type mice but is
able to grow as well as the parental strain in the livers of CP-deficient mice (Kehl-
Fie et al. 2013).
6.4.1 Lactoferrin
The S100 subclass of EF hand family of calcium (Ca)-binding proteins are small,
acidic, alpha helical proteins that are found in vertebrates. Their expression is tissue
specific, and their regulation has been correlated with a wide range of diseases
including autoimmune disorders, cancer, and neurological disorders (Heizmann
et al. 2002). S100 proteins are approximately 100 amino acids long and typically
form homodimers arranged in an antiparallel fashion. Each subunit contains two EF
hand Ca-binding motifs connected by a hinge region.
The first crystal structure of a Zn-bound S100 protein was determined for
S100A7 (Brodersen et al. 1999). This structure revealed the presence of two identi-
cal Zn-binding sites located at opposite ends of the dimer interface containing three
histidines and one aspartic acid. In the S100A7 dimer, each binding site is com-
prised of H86 and H90, arranged in an HXXXH motif from one subunit and H17
and D24 from the other. These binding sites are distinct from the Ca-binding sites,
which are characteristic of S100 proteins. While the binding sites are distinct, the
presence of either Ca or a transition metal increases the affinity for the other (Brophy
et al. 2012; Hayden et al. 2013; Moroz et al. 2011). This three His Asp site is the
canonical transition metal-binding site in S100 proteins. Zn-bound structures have
also been determined for S100B, S100A12, and S100A15 and suggest that the
canonical binding site is largely conserved; however, there is some variation (Murray
et al. 2012; Ostendorp et al. 2011; Moroz et al. 2009, 2011). For example, while
S100A15 shares 93 % sequence identity with S100A7, originally it was thought to
not bind Zn, as the conserved aspartate in the canonical binding site has been
replaced by glycine. However, the crystal structure of Zn-bound S100A15 reveals a
unique binding motif where a chloride ion acts as the fourth ligand, replacing the
conserved aspartate (Murray et al. 2012).
Of the S100 proteins, the metal-binding properties of CP have received consider-
able attention. This interest is due to the demonstrated contribution of CP to host
defense and the sequestration of Mn during infection (Corbin et al. 2008; Kehl-Fie
et al. 2013; Hood et al. 2012; Achouiti et al. 2012). CP comprises ~50 % of the total
protein in the neutrophil cytoplasm and can be expressed by epithelial cells when
induced by proinflammatory cytokines such as IL-22 (Blaschitz and Raffatellu 2010;
Gebhardt et al. 2006). At sites of infection, CP concentration can be in excess of 1 mg/
ml (Clohessy and Golden 1995). CP is capable of binding two Zn ions with picomolar
affinity (Kd) but only one Mn ion with nanomolar or subnanomolar affinity (Kehl-Fie
et al. 2011; Brophy et al. 2012). Consistent with CP imposing metal starvation, the
addition of excess Mn and Zn or mutation of both transition metal-binding sites elimi-
nates the antimicrobial activity of CP (Corbin et al. 2008; Kehl-Fie et al. 2011).
Due to the heterodimeric nature of CP, this protein has two nonidentical transi-
tion metal-binding sites. The first site (S1) was originally predicted to be comprised
of residues H17 and H27 from S100A8 and H91 and H95 from S100A9, with a
fourth histidine replacing the aspartate found in the canonical S100 transition metal-
binding site (Kehl-Fie et al. 2011; Korndorfer et al. 2007). However, subsequent
96 S. Damo and T.E. Kehl-Fie
investigations revealed that two additional histidines, H103 and H105 from S100A9,
also contribute to metal binding (Damo et al. 2013). The second site (S2) is com-
prised of H83 and H87 of S100A8 and H20 and D30 from S100A9 and is effectively
identical to the canonical site found in other S100 proteins (Kehl-Fie et al. 2011;
Korndorfer et al. 2007). Site 1 binds both Mn and Zn tightly, while S2 only binds Zn
tightly (Damo et al. 2013; Brophy et al. 2013). A high-resolution crystal structure
of Mn-bound CP revealed that H103 and H105 from a C-terminal extension of
S100A9 also contribute to Mn binding (Damo et al. 2013). The six histidines in S1
form an almost perfect octahedral coordination site, with the C-terminal extension
wrapping around the Mn ion. Subsequent solution studies confirmed the near-
perfect octahedral geometry and the importance of H103 and H105 to Mn binding
(Brophy et al. 2013). While loss of the two histidines disrupts the ability of S1 to
bind Mn tightly, it appears to have negligible impact on Zn binding (Damo et al.
2013). The C-terminal extension is unique to S100A9 among the S100 proteins and
provides an explanation for why none of the other S100 proteins assayed to date
bind Mn (Damo et al. 2013; Brophy et al. 2013). Similar to other S100 proteins,
there appears to be communication between the EF hands and the transition metal-
binding sites, as the presence of Ca increases the affinity of CP for Zn and Mn. In
the absence of Ca, CP has negligible affinity for Mn, and Zn binding is substantially
weaker (Brophy et al. 2012; Hayden et al. 2013). This altered affinity has been pro-
posed to serve as a switch, allowing CP to accumulate to high levels in the Ca-poor
cytosol of neutrophils without negatively impacting the cell but bind Mn and Zn
tightly when released into the Ca-rich extracellular space.
6.5 Conclusions
As invading pathogens must obtain essential nutrients from their host, nutritional
immunity is a powerful host defense mechanism. While canonically associated with
the Fe-withholding response, the application of advanced elemental analysis has
revealed that other essential metals, such as Mn and Zn, are also withheld from
invaders. The expansion of nutritional immunity to include Mn and Zn leads to
more questions. What are the host factors that restrict the availability of these metals
during infection? What are the bacterial processes that are disrupted by nutritional
immunity? How do successful pathogens overcome this host defense, allowing
them to cause disease? Answering these questions will aid in the development of
therapeutics that are intended to augment the efficacy of nutritional immunity.
References
Abergel RJ, Wilson MK, Arceneaux JE, Hoette TM, Strong RK, Byers BR, Raymond KN (2006)
Anthrax pathogen evades the mammalian immune system through stealth siderophore produc-
tion. Proc Natl Acad Sci U S A 103(49):18499–18503. doi:10.1073/pnas.0607055103
6 Antimicrobial Peptides and Nutritional Immunity 97
Achouiti A, Vogl T, Urban CF, Rohm M, Hommes TJ, van Zoelen MA, Florquin S, Roth J, van’t
Veer C, de Vos AF, van der Poll T (2012) Myeloid-related protein-14 contributes to protective
immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 8(10):e1002987.
doi:10.1371/journal.ppat.1002987
Ammendola S, Pasquali P, Pistoia C, Petrucci P, Petrarca P, Rotilio G, Battistoni A (2007) High-
affinity Zn2+ uptake system ZnuABC is required for bacterial zinc homeostasis in intracellular
environments and contributes to the virulence of Salmonella enterica. Infect Immun
75(12):5867–5876. doi:10.1128/IAI.00559-07
Anderson BF, Baker HM, Norris GE, Rumball SV, Baker EN (1990) Apolactoferrin structure
demonstrates ligand-induced conformational change in transferrins. Nature 344(6268):784–
787. doi:10.1038/344784a0
Anderson ES, Paulley JT, Gaines JM, Valderas MW, Martin DW, Menscher E, Brown TD, Burns
CS, Roop RM 2nd (2009) The manganese transporter MntH is a critical virulence determinant
for Brucella abortus 2308 in experimentally infected mice. Infect Immun 77(8):3466–3474.
doi:10.1128/IAI.00444-09
Andreini C, Banci L, Bertini I, Rosato A (2006) Zinc through the three domains of life. J Proteome
Res 5(11):3173–3178. doi:10.1021/pr0603699
Andreini C, Bertini I, Cavallaro G, Holliday GL, Thornton JM (2008) Metal ions in biological
catalysis: from enzyme databases to general principles. J Biol Inorg Chem 13(8):1205–1218.
doi:10.1007/s00775-008-0404-5
Baker HM, Baker EN (2004) Lactoferrin and iron: structural and dynamic aspects of binding and
release. Biometals Int J Role Metal Ions Biol Biochem Med 17(3):209–216
Bearden SW, Perry RD (1999) The Yfe system of Yersinia pestis transports iron and manganese
and is required for full virulence of plague. Mol Microbiol 32(2):403–414
Berry AM, Paton JC (1996) Sequence heterogeneity of PsaA, a 37-kilodalton putative adhesin
essential for virulence of Streptococcus pneumoniae. Infect Immun 64(12):5255–5262
Blaschitz C, Raffatellu M (2010) Th17 cytokines and the gut mucosal barrier. J Clin Immunol
30(2):196–203. doi:10.1007/s10875-010-9368-7
Bobrov AG, Kirillina O, Fetherston JD, Miller MC, Burlison JA, Perry RD (2014) The Yersinia
pestis siderophore, yersiniabactin, and the ZnuABC system both contribute to zinc acquisition
and the development of lethal septicaemic plague in mice. Mol Microbiol 93(4):759–775.
doi:10.1111/mmi.12693
Brodersen DE, Nyborg J, Kjeldgaard M (1999) Zinc-binding site of an S100 protein revealed. Two
crystal structures of Ca2+-bound human psoriasin (S100A7) in the Zn2+-loaded and Zn2+-free
states. Biochemistry 38(6):1695–1704. doi:10.1021/bi982483d
Brophy MB, Hayden JA, Nolan EM (2012) Calcium ion gradients modulate the zinc affinity and
antibacterial activity of human calprotectin. J Am Chem Soc 134(43):18089–18100.
doi:10.1021/ja307974e
Brophy MB, Nakashige TG, Gaillard A, Nolan EM (2013) Contributions of the S100A9 C-terminal
tail to high-affinity Mn(II) chelation by the host-defense protein human calprotectin. J Am
Chem Soc 135(47):17804–17817. doi:10.1021/ja407147d
Campoy S, Jara M, Busquets N, Perez De Rozas AM, Badiola I, Barbe J (2002) Role of the high-
affinity zinc uptake znuABC system in Salmonella enterica serovar typhimurium virulence.
Infect Immun 70(8):4721–4725
Cassat JE, Skaar EP (2013) Iron in infection and immunity. Cell Host Microbe 13(5):509–519.
doi:10.1016/j.chom.2013.04.010
Clohessy PA, Golden BE (1995) Calprotectin-mediated zinc chelation as a biostatic mechanism in
host defence. Scand J Immunol 42(5):551–556
Corbin BD, Seeley EH, Raab A, Feldmann J, Miller MR, Torres VJ, Anderson KL, Dattilo BM,
Dunman PM, Gerads R, Caprioli RM, Nacken W, Chazin WJ, Skaar EP (2008) Metal chelation
and inhibition of bacterial growth in tissue abscesses. Science 319(5865):962–965. doi:10.1126/
science.1152449
Damo SM, Kehl-Fie TE, Sugitani N, Holt ME, Rathi S, Murphy WJ, Zhang Y, Betz C, Hench L,
Fritz G, Skaar EP, Chazin WJ (2013) Molecular basis for manganese sequestration by
98 S. Damo and T.E. Kehl-Fie
calprotectin and roles in the innate immune response to invading bacterial pathogens. Proc Natl
Acad Sci U S A 110(10):3841–3846. doi:10.1073/pnas.1220341110
Davis LM, Kakuda T, DiRita VJ (2009) A Campylobacter jejuni znuA orthologue is essential for
growth in low-zinc environments and chick colonization. J Bacteriol 191(5):1631–1640.
doi:10.1128/JB.01394-08
Dintilhac A, Alloing G, Granadel C, Claverys JP (1997) Competence and virulence of Streptococcus
pneumoniae: Adc and PsaA mutants exhibit a requirement for Zn and Mn resulting from inac-
tivation of putative ABC metal permeases. Mol Microbiol 25(4):727–739
Gebhardt C, Nemeth J, Angel P, Hess J (2006) S100A8 and S100A9 in inflammation and cancer.
Biochem Pharmacol 72(11):1622–1631. doi:10.1016/j.bcp.2006.05.017
Gläser R, Harder J, Lange H, Bartels J, Christophers E, Schröder JM (2005) Antimicrobial psoria-
sin (S100A7) protects human skin from Escherichia coli infection. Nat Immunol 6(1):57–64.
doi:10.1038/ni1142, ni1142 [pii]
Goetz DH, Holmes MA, Borregaard N, Bluhm ME, Raymond KN, Strong RK (2002) The neutro-
phil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron
acquisition. Mol Cell 10(5):1033–1043
Hayden JA, Brophy MB, Cunden LS, Nolan EM (2013) High-affinity manganese coordination by
human calprotectin is calcium-dependent and requires the histidine-rich site formed at the
dimer interface. J Am Chem Soc 135(2):775–787. doi:10.1021/ja3096416
Heizmann CW, Fritz G, Schafer BW (2002) S100 proteins: structure, functions and pathology.
Front Biosci J Virtual Libr 7:d1356–d1368
Hood MI, Skaar EP (2012) Nutritional immunity: transition metals at the pathogen-host interface.
Nat Rev Microbiol 10(8):525–537. doi:10.1038/nrmicro2836
Hood MI, Mortensen BL, Moore JL, Zhang Y, Kehl-Fie TE, Sugitani N, Chazin WJ, Caprioli RM,
Skaar EP (2012) Identification of an Acinetobacter baumannii zinc acquisition system that
facilitates resistance to calprotectin-mediated zinc sequestration. PLoS Pathog 8(12):e1003068.
doi:10.1371/journal.ppat.1003068
Horsburgh MJ, Wharton SJ, Cox AG, Ingham E, Peacock S, Foster SJ (2002) MntR modulates
expression of the PerR regulon and superoxide resistance in Staphylococcus aureus through
control of manganese uptake. Mol Microbiol 44(5):1269–1286
Janakiraman A, Slauch JM (2000) The putative iron transport system SitABCD encoded on SPI1
is required for full virulence of Salmonella typhimurium. Mol Microbiol 35(5):1146–1155
Janulczyk R, Ricci S, Bjorck L (2003) MtsABC is important for manganese and iron transport,
oxidative stress resistance, and virulence of Streptococcus pyogenes. Infect Immun
71(5):2656–2664
Kehl-Fie TE, Skaar EP (2010) Nutritional immunity beyond iron: a role for manganese and zinc.
Curr Opin Chem Biol 14(2):218–224. doi:10.1016/j.cbpa.2009.11.008
Kehl-Fie TE, Chitayat S, Hood MI, Damo S, Restrepo N, Garcia C, Munro KA, Chazin WJ, Skaar
EP (2011) Nutrient metal sequestration by calprotectin inhibits bacterial superoxide defense,
enhancing neutrophil killing of Staphylococcus aureus. Cell Host Microbe 10(2):158–164.
doi:10.1016/j.chom.2011.07.004
Kehl-Fie TE, Zhang Y, Moore JL, Farrand AJ, Hood MI, Rathi S, Chazin WJ, Caprioli RM, Skaar
EP (2013) MntABC and MntH contribute to systemic Staphylococcus aureus infection by
competing with calprotectin for nutrient manganese. Infect Immun 81(9):3395–3405.
doi:10.1128/IAI.00420-13
Kehres DG, Maguire ME (2003) Emerging themes in manganese transport, biochemistry and
pathogenesis in bacteria. FEMS Microbiol Rev 27(2–3):263–290
Khan JA, Kumar P, Srinivasan A, Singh TP (2001) Protein intermediate trapped by the simultane-
ous crystallization process. Crystal structure of an iron-saturated intermediate in the Fe3+ bind-
ing pathway of camel lactoferrin at 2.7 a resolution. J Biol Chem 276(39):36817–36823.
doi:10.1074/jbc.M104343200
Kim S, Watanabe K, Shirahata T, Watarai M (2004) Zinc uptake system (znuA locus) of Brucella
abortus is essential for intracellular survival and virulence in mice. J Vet Med Sci Jpn Soc Vet
Sci 66(9):1059–1063
6 Antimicrobial Peptides and Nutritional Immunity 99
Korndorfer IP, Brueckner F, Skerra A (2007) The crystal structure of the human (S100A8/
S100A9)2 heterotetramer, calprotectin, illustrates how conformational changes of interacting
alpha-helices can determine specific association of two EF-hand proteins. J Mol Biol
370(5):887–898. doi:10.1016/j.jmb.2007.04.065
Lee KC, Eckert RL (2007) S100A7 (Psoriasin) – mechanism of antibacterial action in wounds.
J Invest Dermatol 127(4):945–957. doi:10.1038/sj.jid.5700663, 5700663 [pii]
Lee JW, Helmann JD (2007) Functional specialization within the Fur family of metalloregulators.
Biometals Int J Role Metal Ions Biol Biochem Med 20(3–4):485–499. doi:10.1007/
s10534-006-9070-7
Lewis DA, Klesney-Tait J, Lumbley SR, Ward CK, Latimer JL, Ison CA, Hansen EJ (1999)
Identification of the znuA-encoded periplasmic zinc transport protein of Haemophilus ducreyi.
Infect Immun 67(10):5060–5068
Liu JZ, Jellbauer S, Poe AJ, Ton V, Pesciaroli M, Kehl-Fie TE, Restrepo NA, Hosking MP,
Edwards RA, Battistoni A, Pasquali P, Lane TE, Chazin WJ, Vogl T, Roth J, Skaar EP, Raffatellu
M (2012) Zinc sequestration by the neutrophil protein calprotectin enhances Salmonella
growth in the inflamed gut. Cell Host Microbe 11(3):227–239. doi:10.1016/j.chom.
2012.01.017
Mayfield JA, Dehner CA, DuBois JL (2011) Recent advances in bacterial heme protein biochem-
istry. Curr Opin Chem Biol 15(2):260–266. doi:10.1016/j.cbpa.2011.02.002
Mizutani K, Toyoda M, Mikami B (2012) X-ray structures of transferrins and related proteins.
Biochim Biophys Acta 1820(3):203–211. doi:10.1016/j.bbagen.2011.08.003
Moroz OV, Blagova EV, Wilkinson AJ, Wilson KS, Bronstein IB (2009) The crystal structures of
human S100A12 in apo form and in complex with zinc: new insights into S100A12 oligomeri-
sation. J Mol Biol 391(3):536–551. doi:10.1016/j.jmb.2009.06.004
Moroz OV, Wilson KS, Bronstein IB (2011) The role of zinc in the S100 proteins: insights from
the X-ray structures. Amino Acids 41(4):761–772. doi:10.1007/s00726-010-0540-4
Murray JI, Tonkin ML, Whiting AL, Peng F, Farnell B, Cullen JT, Hof F, Boulanger MJ (2012)
Structural characterization of S100A15 reveals a novel zinc coordination site among S100
proteins and altered surface chemistry with functional implications for receptor binding. BMC
Struct Biol 12:16. doi:10.1186/1472-6807-12-16
Orsi N (2004) The antimicrobial activity of lactoferrin: current status and perspectives. Biometals
Int J Role Metal Ions Biol Biochem Med 17(3):189–196
Ostendorp T, Diez J, Heizmann CW, Fritz G (2011) The crystal structures of human S100B in the
zinc- and calcium-loaded state at three pH values reveal zinc ligand swapping. Biochim
Biophys Acta 1813(5):1083–1091. doi:10.1016/j.bbamcr.2010.10.006
Papp-Wallace KM, Maguire ME (2006) Manganese transport and the role of manganese in viru-
lence. Annu Rev Microbiol 60:187–209. doi:10.1146/annurev.micro.60.080805.142149
Py B, Barras F (2010) Building Fe-S proteins: bacterial strategies. Nat Rev Microbiol 8(6):436–
446. doi:10.1038/nrmicro2356
Raffatellu M, George MD, Akiyama Y, Hornsby MJ, Nuccio SP, Paixao TA, Butler BP, Chu H,
Santos RL, Berger T, Mak TW, Tsolis RM, Bevins CL, Solnick JV, Dandekar S, Baumler AJ
(2009) Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype
Typhimurium for growth and survival in the inflamed intestine. Cell Host Microbe 5(5):476–
486. doi:10.1016/j.chom.2009.03.011
Sabri M, Houle S, Dozois CM (2009) Roles of the extraintestinal pathogenic Escherichia coli
ZnuACB and ZupT zinc transporters during urinary tract infection. Infect Immun 77(3):1155–
1164. doi:10.1128/IAI.01082-08
Sun X, Baker HM, Ge R, Sun H, He QY, Baker EN (2009) Crystal structure and metal binding
properties of the lipoprotein MtsA, responsible for iron transport in Streptococcus pyogenes.
Biochemistry 48(26):6184–6190. doi:10.1021/bi900552c
Troxell B, Hassan HM (2013) Transcriptional regulation by Ferric Uptake Regulator (Fur) in
pathogenic bacteria. Front Cell Infect Microbiol 3:59. doi:10.3389/fcimb.2013.00059
Vallee BL, Auld DS (1990) Zinc coordination, function, and structure of zinc enzymes and other
proteins. Biochemistry 29(24):5647–5659
100 S. Damo and T.E. Kehl-Fie
Waldron KJ, Robinson NJ (2009) How do bacterial cells ensure that metalloproteins get the correct
metal? Nat Rev Microbiol 7(1):25–35. doi:10.1038/nrmicro2057
Wally J, Buchanan SK (2007) A structural comparison of human serum transferrin and human
lactoferrin. Biometals Int J Role Metal Ions Biol Biochem Med 20(3–4):249–262. doi:10.1007/
s10534-006-9062-7
Wandersman C, Delepelaire P (2004) Bacterial iron sources: from siderophores to hemophores.
Annu Rev Microbiol 58:611–647. doi:10.1146/annurev.micro.58.030603.123811
Weinberg ED (1974) Iron and susceptibility to infectious disease. Science 184(4140):952–956
Weinberg ED (2009) Iron availability and infection. Biochim Biophys Acta 1790(7):600–605.
doi:10.1016/j.bbagen.2008.07.002, S0304-4165(08)00145-1 [pii]
Weston BF, Brenot A, Caparon MG (2009) The metal homeostasis protein, Lsp, of Streptococcus
pyogenes is necessary for acquisition of zinc and virulence. Infect Immun 77(7):2840–2848.
doi:10.1128/IAI.01299-08
Chapter 7
Regulation of Antimicrobial Peptide Gene
Expression by Vitamin D
Adrian F. Gombart
7.1 Introduction
A decade ago three groups simultaneously discovered the regulation of human anti-
microbial peptide gene expression by vitamin D. In screening the human genome
for vitamin D response elements (VDREs), White and colleagues identified poten-
tial VDREs in the cathelicidin antimicrobial peptide (CAMP) and β-defensin 4
(DEFB4) genes. They demonstrated induction of these two genes by treatment of
isolated human keratinocytes, monocytes and neutrophils, and human cell lines
with 1α,25-dihydroxyvitaminD3 [1,25(OH)2D3] concomitant with increased secre-
tion of bactericidal activity from treated cells (Wang et al. 2004). Ståhle and
The most effective way to acquire vitamin D is through synthesis in the skin or
consumption of a purified supplement as diet, which is a poor source (Holick 2011).
Ultraviolet B rays provided by natural or artificial sunlight cleave the B-ring of
7-dehydrocholesterol in the skin to produce cholecalciferol or vitamin D3. This is
absorbed into the blood and hydroxylated in the liver by the cytochrome p450
enzyme CYP27A1 to calcidiol or 25(OH)D3. This form (together with 25(OH)D2;
see below) is measured in the serum as an indicator of vitamin D status (Holick
2011). 25(OH)D3 is converted to its bioactive form, calcitriol or 1,25(OH)2D3, by
the mitochondrial 1α-hydroxylase enzyme CYP27B1 in the kidney. A fungal-
derived form of vitamin D is created by the UVB exposure of ergosterol to generate
104 A.F. Gombart
normal macrophages indicated that CYP27B1 activity was induced as part of the
immune response (Koeffler et al. 1985; Reichel et al. 1986). DCs confer specific
homing properties upon T cells during the adaptive immune response, and DCs
derived from the skin are able to synthesize 1,25(OH)2D3 from vitamin D3. This, in
turn, induces expression of CC chemokine receptor 10 in T cells and suppresses
expression of gut-homing receptors which enable T cells to migrate toward the che-
mokine CCL27 that is secreted by epidermal keratinocytes. These findings demon-
strate that DCs produce locally high levels of 1,25(OH)2D3 to regulate T-cell
epidermal tropism (Sigmundsdottir et al. 2007).
The production of potentially high local levels of 1,25(OH)2D3 is most likely
important for intracrine and paracrine influences on the interactions between vita-
min D, the immune system, and pathogens (Hewison 2012). During the mid-1980s,
it was demonstrated that both 25(OH)D3 and 1,25(OH)2D3 increased the capacity of
human monocytes to control Mycobacterium tuberculosis (Mtb) growth (Davies
1985; Rook et al. 1986). Nearly 20 years later, as described above, we and others
discovered that vitamin D increased expression of the CAMP gene (Wang et al.
2004; Weber et al. 2005; Gombart et al. 2005). In addition, the human β-defensin 2
or DEFB4 gene was identified as a vitamin D inducible antimicrobial peptide gene,
but its induction by vitamin D or TLR activation is much less robust than CAMP
(Wang et al. 2004; Liu et al. 2006). These observations offered a mechanism by
which vitamin D could directly enhance killing of Mtb.
CYP27B1 activates the VDR and induces CAMP expression and antimicrobial
activity against Mtb (Krutzik et al. 2008; Montoya et al. 2014). In human mono-
cytes, Th1 cytokine IFN-γ upregulates TLR2/TLR1 induction of CYP27B1 and the
bioconversion of 25(OH)D3 to 1,25(OH)2D3 which enhances induction of CAMP
(Edfeldt et al. 2010). Further, vitamin D is required for IFN-γ-mediated activity of
human macrophages (Fabri et al. 2011). On the other hand, the Th2 cytokine IL-4
induces CYP24A1 expression which leads to the catabolism of 25(OH)D3 and
downregulation of CAMP expression (Edfeldt et al. 2010). In contrast, the Th2 cyto-
kine IL-13 enhances CAMP expression by 25(OH)D3 due to increased CYP27B1
expression and synthesis of 1,25(OH)2D3 (Schrumpf et al. 2012). No effect is
observed with IL-17 in monocytes, but in the presence of 1,25(OH)2D3, IL-17
enhances CAMP expression in human keratinocytes via activation of the Act1 and
MEK/ERK pathway (Peric et al. 2008). In addition to IL-4, other cytokines can
inhibit antimicrobial peptide (AMP) expression. In macrophages, IFN-γ-induced
vitamin D-dependent AMP expression was suppressed by IFN-β and IL-10 (Teles
et al. 2013). Similarly, in placental cells, IL-10 inhibited β-defensin and CAMP
expression, while 1,25(OH)2D3 treatment could override the suppression (Olmos-
Ortiz et al. 2015). Further, TNF-α and 1,25(OH)2D3 enhanced β-defensin, and
TNF-α reduced both basal and 1,25(OH)2D3-induced CAMP expression (Olmos-
Ortiz et al. 2015). Taken together, the differential effect of T-cell cytokines on
CAMP and DEFB4 expression represents mechanisms by which adaptive immune
responses can regulate innate immune antimicrobial peptide defenses against patho-
gens. It remains to be determined how these various signaling pathways work
together in vivo during infection.
these studies support an important role for vitamin D in modulating the immune
response to Mtb infection.
These findings have renewed interest in potentially using vitamin D to treat
tuberculosis. A review of clinical trials and case series indicates that numerous stud-
ies are methodologically flawed (Martineau et al. 2007) or an insufficient vitamin D
dose was used (Wejse et al. 2009). Two small randomized studies indicate some
benefit from vitamin supplementation of TB patients (Nursyam et al. 2006; Morcos
et al. 1998). More recently, pulmonary tuberculosis patients receiving standard ther-
apy and a 100,000 IU dose of vitamin D3 every 2 weeks showed accelerated sputum
conversion if they possessed the tt genotype of the vitamin D receptor as compared
with placebo (Martineau et al. 2011). Further, vitamin D supplementation acceler-
ated resolution of inflammation during tuberculosis treatment (Coussens et al.
2012). A recent randomized, double-blinded, multicenter, placebo-controlled clini-
cal study involving 258 patients showed that 600,000 IU vitamin D3 once per month
for 2 months led to a significant increase in average weight gain and lower residual
disease by chest x-ray as compared to placebo (Salahuddin et al. 2013).
Deficiencies in vitamin D are associated with poor outcomes in HIV-infected
individual, bacterial vaginosis in the first trimester of pregnancy, increased influ-
enza A infections, and increased respiratory tract infections (Bodnar et al. 2009;
Villamor 2006; Aloia and Li-Ng 2007; Sabetta et al. 2010). Supplementation with
vitamin D lowered the incidence of seasonal flu in school children, the elderly, and
African-American women and lowered the severity of respiratory tract infections
(Urashima et al. 2010; Avenell et al. 2007; Aloia et al. 2005; Kenny et al. 2012). In
contrast, vitamin D supplementation did not reduce the incidence and duration of
severity of upper respiratory tract infection (Li-Ng et al. 2009). In a meta-analysis
of 11 placebo-controlled studies involving 5660 patients, vitamin D showed a pro-
tective effect against respiratory tract infections with once-daily dosing being better
than bolus doses (Bergman et al. 2013). The authors noted that there was significant
heterogeneity and evidence of publication bias in the field and warned that results
should be carefully interpreted (Bergman et al. 2013). It should be noted that in all
of these studies including those with tuberculosis, the role of CAMP induction in
these outcomes is unknown. Future studies must optimize dose, dosing frequency,
and target populations that are deficient in vitamin D to detect modest effects.
patients, a positive association between 25(OH)D and hCAP18 levels was observed
in all patients (Jeng et al. 2009). In healthy individuals, a positive association
between hCAP18 and 25(OH)D levels was observed at levels of 25(OH)D below
32 ng/ml, but not above (Bhan et al. 2011; Dixon et al. 2012). In the elderly and in
atopic dermatitis patients and normal controls, a positive correlation was observed
without applying a cutoff (Alvarez-Rodriguez et al. 2012; Kanda et al. 2012). On
the other hand, in cord blood samples, patients with active TB and patients with
pneumonia, a correlation between serum 25(OH)D and hCAP18 was not observed
(Yamshchikov et al. 2010; Mandic Havelka et al. 2010; Leow et al. 2011).
Supplementation of atopic dermatitis patients with 4000 IU/day vitamin D for 3
weeks increased CAMP in skin lesions and unaffected skin, but a second study with
more patients was negative (Hata et al. 2008, 2014). Several studies using high-dose
supplementation (50,000–60,000 IU/week) did not observe increased hCAP18 in
the blood (Adams et al. 2009; Alvarez et al. 2013; Das et al. 2014). In a randomized
controlled trial in patients with severe sepsis, 1,25(OH)2D3 did not increase plasma
hCAP18 levels (Leaf et al. 2014). In a study of 15 hereditary vitamin D-resistant
rickets patients (possess a nonfunctional VDR) and 17 normal controls, it was
shown that VDR is required for induction of CAMP by vitamin D in adherent
mononuclear cells cultured for 24 h, but basal expression of CAMP in various cell
types, fluids, or tissue samples was not determined (Tiosano et al. 2013). Additional
studies are required to determine the effect of vitamin D status or treatment on in
vivo CAMP expression, particularly on the in vivo induction of CAMP in immune
cells like macrophages during infection.
The difficulty in determining the role CAMP in mediating the effects of vitamin D
on the immune response is the lack of a good animal model that replicates the path-
way as it is found in humans. As described earlier, vitamin D does not regulate
CAMP expression in mice or other mammals (Gombart et al. 2009a). In addition,
work from our own group and others revealed a striking difference in the use of
vitamin D by human versus murine macrophages. As described above, activation of
human macrophages by TLR ligands induces expression of CYP27B1 and the bio-
conversion of 25(OH)D3 to 1,25(OH)2D3. This, in turn, leads to the induction of
various VDR target genes including CAMP. In contrast, TLR activation of murine
macrophages does not induce CYP27B1 expression; thus, bioactive 1,25(OH)2D3 is
not synthesized by murine macrophages (Kapetanovic et al. 2012; Ooi et al. 2014),
and vitamin D target genes are not induced (our unpublished findings). This major
difference in the utilization of vitamin D by macrophages highlights the importance
of caution when using the mouse model to elucidate the role of vitamin D on
immune function in humans. Macrophages are very likely important for producing
locally high levels of 1,25(OH)2D3 at sites of infection in humans, but not in mice.
7 Regulation of Antimicrobial Peptide Gene Expression by Vitamin D 109
7.9 Conclusion
References
Brennan A et al (1987) Dendritic cells from human tissues express receptors for the immunoregu-
latory vitamin D3 metabolite, dihydroxycholecalciferol. Immunology 61(4):457–461
Chen S et al (2007) Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentia-
tion. J Immunol 179(3):1634–1647
Chertov O et al (1996) Identification of defensin-1, defensin-2, and CAP37/azurocidin as T-cell
chemoattractant proteins released from interleukin-8-stimulated neutrophils. J Biol Chem
271(6):2935–2940
Christakos S et al (1996) Genomic mechanisms involved in the pleiotropic actions of 1,
25-dihydroxyvitamin D3. Biochem J 316(Pt 2):361–371
Chromek M et al (2006) The antimicrobial peptide cathelicidin protects the urinary tract against
invasive bacterial infection. Nat Med 12(6):636–641
Coussens AK et al (2012) Vitamin D accelerates resolution of inflammatory responses during
tuberculosis treatment. Proc Natl Acad Sci U S A 109(38):15449–15454
Daniel C et al (2008) Immune modulatory treatment of trinitrobenzene sulfonic acid colitis with
calcitriol is associated with a change of a T helper (Th) 1/Th17 to a Th2 and regulatory T cell
profile. J Pharmacol Exp Ther 324(1):23–33
Das M et al (2014) Effect of vitamin D supplementation on cathelicidin, IFN-gamma, IL-4 and
Th1/Th2 transcription factors in young healthy females. Eur J Clin Nutr 68(3):338–343
Davidson DJ et al (2004) The cationic antimicrobial peptide LL-37 modulates dendritic cell dif-
ferentiation and dendritic cell-induced T cell polarization. J Immunol 172(2):1146–1156
Davies PD (1985) A possible link between vitamin D deficiency and impaired host defence to
Mycobacterium tuberculosis. Tubercle 66(4):301–306
Deluca HF, Cantorna MT (2001) Vitamin D: its role and uses in immunology. FASEB J 15(14):
2579–2585
Dixon BM et al (2012) Positive correlation between circulating cathelicidin antimicrobial peptide
(hCAP18/LL-37) and 25-hydroxyvitamin D levels in healthy adults. BMC Res Notes 5:575
Dowling GB (1946) New remedy for lupus. Lancet 1(6399):590
Edfeldt K et al (2010) T-cell cytokines differentially control human monocyte antimicrobial responses
by regulating vitamin D metabolism. Proc Natl Acad Sci U S A 107(52):22593–22598
Fabri M et al (2011) Vitamin D is required for IFN-gamma-mediated antimicrobial activity of
human macrophages. Sci Transl Med 3(104):104ra102
Frohm Nilsson M et al (1999) The human cationic antimicrobial protein (hCAP18), a peptide
antibiotic, is widely expressed in human squamous epithelia and colocalizes with interleukin-6.
Infect Immun 67(5):2561–2566
Gaumond E (1948) Lupus vulgaris and vitamin D. Can Med Assoc J 59(6):522–527
Gombart AF (2009) The vitamin D-antimicrobial peptide pathway and its role in protection against
infection. Future Microbiol 4(9):1151–1165
Gombart AF, Borregaard N, Koeffler HP (2005) Human cathelicidin antimicrobial peptide
(CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in
myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J 19(9):1067–1077
Gombart AF et al (2007) Regulation of the CAMP gene by 1,25(OH)2D3 in various tissues.
J Steroid Biochem Mol Biol 103(3-5):552–557
Gombart A, Saito T, Koeffler HP (2009a) Exaptation of an ancient Alu short interspersed element
provides a highly conserved vitamin D-mediated innate immune response in humans and pri-
mates. BMC Genomics 10(1):321
Gombart AF et al (2009b) Low plasma level of cathelicidin antimicrobial peptide (hCAP18) pre-
dicts increased infectious disease mortality in patients undergoing hemodialysis. Clin Infect
Dis 48(4):418–424
Grober U et al (2013) Vitamin D: update 2013: from rickets prophylaxis to general preventive
healthcare. Dermatoendocrinol 5(3):331–347
Hansdottir S et al (2008) Respiratory epithelial cells convert inactive vitamin D to its active form:
potential effects on host defense. J Immunol 181(10):7090–7099
Hata TR et al (2008) Administration of oral vitamin D induces cathelicidin production in atopic
individuals. J Allergy Clin Immunol 122(4):829–831
7 Regulation of Antimicrobial Peptide Gene Expression by Vitamin D 111
Mangelsdorf DJ et al (1995) The nuclear receptor superfamily: the second decade. Cell 83(6):
835–839
Martineau AR et al (2007) Vitamin D in the treatment of pulmonary tuberculosis. J Steroid
Biochem Mol Biol 103(3-5):793–798
Martineau AR et al (2011) High-dose vitamin D(3) during intensive-phase antimicrobial treatment
of pulmonary tuberculosis: a double-blind randomised controlled trial. Lancet 377(9761):
242–250
Mathieu C, Adorini L (2002) The coming of age of 1,25-dihydroxyvitamin D(3) analogs as immu-
nomodulatory agents. Trends Mol Med 8(4):174–179
Mily A et al (2013) Oral intake of phenylbutyrate with or without vitamin D3 upregulates the
cathelicidin LL-37 in human macrophages: a dose finding study for treatment of tuberculosis.
BMC Pulm Med 13:23
Montoya D et al (2014) IL-32 is a molecular marker of a host defense network in human tubercu-
losis. Sci Transl Med 6(250):250ra114
Morcos MM et al (1998) Vitamin D administration to tuberculous children and its value. Boll
Chim Farm 137(5):157–164
Murakami M et al (2002) Cathelicidin antimicrobial peptides are expressed in salivary glands and
saliva. J Dent Res 81(12):845–850
Nizet V et al (2001) Innate antimicrobial peptide protects the skin from invasive bacterial infec-
tion. Nature 414(6862):454–457
Nursyam EW, Amin Z, Rumende CM (2006) The effect of vitamin D as supplementary treatment
in patients with moderately advanced pulmonary tuberculous lesion. Acta Med Indones
38(1):3–5
Olmos-Ortiz A et al (2015) IL-10 inhibits while calcitriol reestablishes placental antimicrobial
peptides gene expression. J Steroid Biochem Mol Biol 148:187–193
Ooi JH et al (2014) Murine CD8+ T cells but not macrophages express the vitamin D 1alpha-
hydroxylase. J Nutr Biochem 25(1):58–65
Paz N et al (2007) Altered adenosine-to-inosine RNA editing in human cancer. Genome Res
17(11):1586–1595
Penna G, Adorini L (2000) 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation,
activation, and survival of dendritic cells leading to impaired alloreactive T cell activation.
J Immunol 164(5):2405–2411
Peric M et al (2008) IL-17A Enhances vitamin D3-induced expression of cathelicidin antimicro-
bial peptide in human keratinocytes. J Immunol 181(12):8504–8512
Provvedini D et al (1983) 1,25-dihydroxyvitamin D3 receptors in human leukocytes. Science
221(4616):1181–1183
Reichel H, Koeffler HP, Norman AW (1986) Regulation of 25-hydroxyvitamin D3 metabolism in
a human promyelocytic leukemia cell line (HL-60): 1,25-dihydroxyvitamin D3 stimulates the
synthesis of 24,25-dihydroxyvitamin D3. Arch Biochem Biophys 251(1):222–231
Rook G (1986) Vitamin D and tuberculosis. Tubercle 67(2):155–156
Rook GA et al (1986) Vitamin D3, gamma interferon, and control of proliferation of Mycobacterium
tuberculosis by human monocytes. Immunology 57(1):159–163
Sabetta JR et al (2010) Serum 25-hydroxyvitamin d and the incidence of acute viral respiratory
tract infections in healthy adults. PLoS One 5(6):e11088
Saito H et al (2003) Human fibroblast growth factor-23 mutants suppress Na + -dependent phos-
phate co-transport activity and 1alpha,25-dihydroxyvitamin D3 production. J Biol Chem
278(4):2206–2211
Salahuddin N et al (2013) Vitamin D accelerates clinical recovery from tuberculosis: results of the
SUCCINCT Study [Supplementary Cholecalciferol in recovery from tuberculosis]. A random-
ized, placebo-controlled, clinical trial of vitamin D supplementation in patients with pulmo-
nary tuberculosis’. BMC Infect Dis 13:22
Schauber J et al (2006) Control of the innate epithelial antimicrobial response is cell-type specific
and dependent on relevant microenvironmental stimuli. Immunology 118(4):509–519
7 Regulation of Antimicrobial Peptide Gene Expression by Vitamin D 113
Schauber J et al (2007) Injury enhances TLR2 function and antimicrobial peptide expression
through a vitamin D-dependent mechanism. J Clin Invest 117(3):803–811
Schauber J et al (2008) Histone acetylation in keratinocytes enables control of the expression of
cathelicidin and CD14 by 1,25-dihydroxyvitamin D3. J Invest Dermatol 128(4):816–824
Schrumpf JA et al (2012) Interleukin 13 exposure enhances vitamin D-mediated expression of the
human cathelicidin antimicrobial peptide 18/LL-37 in bronchial epithelial cells. Infect Immun
80(12):4485–4494
Sigmundsdottir H et al (2007) DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell
attraction to the epidermal chemokine CCL27. Nat Immunol 8(3):285–293
Sorensen O et al (1997) An ELISA for hCAP-18, the cathelicidin present in human neutrophils and
plasma. J Immunol Methods 206(1-2):53–59
Takahashi K et al (2002) Human neutrophils express messenger RNA of vitamin D receptor and
respond to 1alpha,25-dihydroxyvitamin D3. Immunopharmacol Immunotoxicol 24(3):335–347
Teles RM et al (2013) Type I interferon suppresses type II interferon-triggered human anti-
mycobacterial responses. Science 339(6126):1448–1453
Tiosano D et al (2013) The role of vitamin D receptor in innate and adaptive immunity: a study in
hereditary vitamin D-resistant rickets patients. J Clin Endocrinol Metab 98(4):1685–1693
Urashima M et al (2010) Randomized trial of vitamin D supplementation to prevent seasonal influ-
enza A in schoolchildren. Am J Clin Nutr 91(5):1255–1260
van der Does AM et al (2014) Vitamin D(3) and phenylbutyrate promote development of a human
dendritic cell subset displaying enhanced antimicrobial properties. J Leukoc Biol 95(6):
883–891
Verway M et al (2013) Vitamin D induces interleukin-1beta expression: paracrine macrophage
epithelial signaling controls M. tuberculosis infection. PLoS Pathog 9(6):e1003407
Villamor E (2006) A potential role for vitamin D on HIV infection? Nutr Rev 64(5 Pt 1):
226–233
Voss E et al (2006) NOD2/CARD15 mediates induction of the antimicrobial peptide human beta-
defensin-2. J Biol Chem 281(4):2005–2011
Wang TT et al (2004) Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial
peptide gene expression. J Immunol 173(5):2909–2912
Wang J et al (2008) Vitamin D3 induces autophagy of human myeloid leukemia cells. J Biol Chem
283(37):25596–25605
Wang TT et al (2010) Direct and indirect induction by 1,25-dihydroxyvitamin D3 of the NOD2/
CARD15-defensin beta2 innate immune pathway defective in Crohn disease. J Biol Chem
285(4):2227–2231
Weber G et al (2005) Vitamin D induces the antimicrobial protein hCAP18 in human skin. J Invest
Dermatol 124(5):1080–1082
Wejse C et al (2009) Vitamin D as supplementary treatment for tuberculosis: a double-blind, ran-
domized, placebo-controlled trial. Am J Respir Crit Care Med 179(9):843–850
White JH (2010) Vitamin D as an inducer of cathelicidin antimicrobial peptide expression: past,
present and future. J Steroid Biochem Mol Biol 121(1-2):234–238
Yamshchikov AV et al (2010) Vitamin D status and antimicrobial peptide cathelicidin (LL-37)
concentrations in patients with active pulmonary tuberculosis. Am J Clin Nutr 92(3):603–611
Yang D, Chertov O, Oppenheim JJ (2001) Participation of mammalian defensins and cathelicidins
in anti-microbial immunity: receptors and activities of human defensins and cathelicidin (LL-
37). J Leukoc Biol 69(5):691–697
Yuk JM et al (2009) Vitamin D3 induces autophagy in human monocytes/macrophages via cathe-
licidin. Cell Host Microbe 6(3):231–243
Zierold C, Darwish HM, DeLuca HF (1995) Two vitamin D response elements function in the rat
1,25-dihydroxyvitamin D 24-hydroxylase promoter. J Biol Chem 270(4):1675–1678
Chapter 8
Dichotomous Roles of Cationic Polypeptides
Targeting HIV
Worldwide, by the end of 2013, the HIV/AIDS pandemic had killed nearly 40 mil-
lion people, and an estimated 35 million people at the time were infected with
HIV [www.who.int]. The use of antiretroviral therapy (ART) to combat the virus
has been steadily increasing, and the number of people globally receiving ART
approaches 13 million. With such a large number of people receiving ART, viral
resistance to most of the available drugs is inevitable, and thus, to control the
pandemic, additional strategies must be employed in concert with ART. While the
field still remains hopeful, essentially all vaccines targeting HIV-1 have shown
little promise or outright failure in the clinic (Haynes et al. 2014). Newer strate-
gies to prevent transmission of HIV-1 have demonstrated potential, including pre-
exposure prophylaxis (PrEP) targeting the infected, and to a lesser extent
uninfected, partner in a serodiscordant couple to reduce viral load and subsequent
viral transmission (McMahon et al. 2014). Conversely, topical antiretroviral
microbicides that are applied by the receptive partner to their own vagina or rec-
tum have not been successful in the clinic due to myriad factors, including product
adherence and psychosocial issues (Van Damme and Szpir 2012). Ensuring bio-
compatibility of microbicides with the milieu of the vagina or rectum has been
more recently realized (Trifonova et al. 2006), as several older-generation micro-
bicides likely failed due to incompatibility with the mucosal environment (Morris
and Lacey 2010).
Developing a mucosal vaccine or a topical microbicide that would be applied
to the anogenital mucosa to prevent viral transmission requires a comprehensive
understanding of the local mucosal environment to increase the likelihood of suc-
cess. While there are many topics that could be covered in this realm, including
the architecture and cellular composition of the respective mucosal tissues, this
chapter is focused particularly on cationic effector peptides and proteins that are
produced by certain immune cells and epithelia and often liberated into secre-
tions that overlie the mucosa. Historically, these cationic peptides and proteins
have been referred to as “antimicrobial polypeptides” or “AMPs,” due to their
broad-spectrum activity against bacteria, fungi, and viruses such as HIV-1
(Zasloff 1992), and more recently termed “host defense polypeptides” or “HDPs,”
because of expanded activities that are related to innate immunity but not neces-
sarily due to direct antimicrobial action (Mansour et al. 2014). Either definition
implies that these polypeptides are involved in protection of the host, and indeed
this is the ascribed function for many peptides and proteins detailed in other
chapters of this book.
Our chapter instead examines the dichotomous roles of cationic polypeptides in
their abilities to modulate HIV-1 infection. The first half of the chapter begins in a
similar vein as most reviews on the topic, by outlining the capacity of human cat-
ionic polypeptides to inhibit HIV-1 infection and transmission. The second part
presents a growing body of evidence that suggests many human cationic polypep-
tides are in fact enhancers of HIV-1 infection. As presented below, the distinction
between inhibiting and promoting HIV-1 infection is not mutually exclusive for
certain classes of polypeptides. Note also that all cationic polypeptides are bio-
logically relevant in the context of HIV-1 infection and transmission – they have
either been identified in the genital or anorectal mucosal areas that are the main
target tissues for HIV-1 transmission or are contained in human semen that is the
source of most transmitted HIV-1. Due to space constraints, background on the life
cycle of HIV-1 and events in sexual transmission of HIV-1 are not covered in this
chapter, but the reader is instead referred to an excellent review on the subject
(Haase 2011).
8 Dichotomous Roles of Cationic Polypeptides Targeting HIV 117
Trappin-2/elafin and secretory leukocyte protease inhibitor (SLPI) are 10–12 kDa
members of the whey acidic protein (WAP) motif family (Bingle and Vyakarnam
118 A.M. Cole and A.L. Cole
2008; Moreau et al. 2008). Reducing inflammation in the female reproductive tract
has been attributed to their antiprotease activity targeting neutrophil elastase and pro-
teinase-3 (Horne et al. 2008). The ability of trappin-2/elafin to prevent HIV-1 infec-
tion is supported by several lines of evidence (Ghosh et al. 2010). Only uterine cells
of the upper female reproductive tract (FRT) can be induced to express trappin-2/
elafin, while epithelia of both the upper and lower FRT can express trappin-2/elafin
constitutively (Ghosh et al. 2010). Importantly, the antiretroviral activity of trappin-2/
elafin was measured against HIV-1 that utilizes either coreceptor required for entry
(CXCR4 or CCR5), revealing that trappin-2/elafin targeted HIV-1 virions directly in
dose-dependent manner. Trappin-2/elafin concentrations are highest during the secre-
tory phase of the menstrual cycle, and HIV-infected patients contain less of this pro-
tein in their cervicovaginal lavage fluids than uninfected patients (Ghosh et al. 2010),
suggesting hormonal and other regulatory factors likely modulate its expression.
SLPI’s capacity to prevent HIV infection is equivocal. One report suggested that
very high (milligram per mL) concentrations of SLPI could not prevent HIV-1 infec-
tion (Turpin et al. 1996). Other studies revealed that very low concentrations (high
nanomolar) could block HIV-1 or uncoat the viral capsid independent of SLPI’s
function as a protease (McNeely et al. 1995, 1997). Moreover, reduced SLPI levels
in the cervicovaginal fluids of women with bacterial vaginosis or sexually transmit-
ted infections (STIs) is a potential cofactor for increased incidence of HIV-1 acqui-
sition (Draper et al. 2000). Indirect evidence in another study suggested that elevated
SLPI levels were associated with decreased perinatal acquisition of HIV, which was
specific to SLPI and not other cationic polypeptides measured (Pillay et al. 2001).
Further study is warranted to unmask the true antiretroviral role for SLPI, especially
in combination with other antiviral factors that may potentiate its action.
8.2.3 Defensins
needed for HIV-1 entry into target cells (Munk et al. 2003; Gallo et al. 2006). In a
study that utilized aminoglycosides to suppress the premature termination codon in
retrocyclins, intact retrocyclin peptides could be produced by promyelocytes and
vaginal epithelia and were active against HIV-1 (Venkataraman et al. 2009). Given
that θ-defensins are thought to be produced from the end-to-end ligation and mac-
rocyclization of two nonapeptides (Tang et al. 1999), this study also suggested that
the complex molecular machinery required to produce retrocyclin peptides remains
comparatively intact in human cells. It is therefore tempting to speculate whether
these antiretroviral retrocyclin peptides can still be produced by modern-day
humans under natural conditions that at least partially suppress the premature termi-
nation codon.
Human β-defensins (HBDs) are produced primarily by epithelial cells (Lehrer
and Ganz 2002), and although there are at least six β-defensin peptides produced
throughout the body, only HBD2 and HBD3 have been reported to exert activity
against HIV-1 (Klotman and Chang 2006). HBD2 and HBD3 downmodulate
CXCR4, one of two chemokine coreceptors required for HIV-1 entry into target
CD4+ cells (Quinones-Mateu et al. 2003), while only HBD3 antagonizes the CXCR4
receptor (Feng et al. 2006). In vaginal fluid (Valore et al. 2002) and cervical mucus
plugs (Hein et al. 2002), the concentration of HBD2 is present at nanograms per mL
concentrations, which are far below the low microgram per mL levels required for
anti-HIV-1 activity in vitro (Quinones-Mateu et al. 2003). Nevertheless, the physi-
ological amounts of HBD2 are within the range required to chemoattract potential
targets of HIV-1. HBD2 is a ligand for CCR6-expressing cells, including CD45RO+/
CD4+ T cells and immature dendritic cells (Hoover et al. 2002), while HBD2 and
HBD3 are ligands for CCR2-expressing cells, including macrophages/monocytes
and neutrophils (Rohrl et al. 2010). While HBD2 and HBD3 may unlikely directly
affect HIV-1 infection in vivo, their upregulation by inflammatory processes and the
ability of HBDs to recruit additional target cells to the genital and other mucosae
together would likely increase the incidence of viral transmission.
Human α-defensins are further divided into two subcategories, human neutrophil
peptides 1–4 (HNP1-4) and human defensins 5 and 6 (HD5, HD6). With regard to
HIV-1 infection, perhaps this class of defensins is the most intriguing: several HNPs
are active against HIV-1, while HD5 and HD6, which will be discussed in the next
section, are robust enhancers of HIV-1 infection. HNPs are present at very high con-
centrations in neutrophil azurophil granules where they can reach concentrations in
the millimolar range, comprising approximately one-third of the granules’ total pro-
tein content (Harwig et al. 1992; Valore and Ganz 1992; Ganz and Lehrer 1997). The
first mention of an anti-HIV-1 peptide began with a short communication describing
how α-defensins isolated from rabbits, rats, and guinea pigs could inhibit HIV-1
replication and reduce HIV-mediated cytopathology in a T-cell line (Nakashima
et al. 1993). Similarities between α-defensins and a looped region of the fusogenic
HIV-1 gp41 glycoprotein suggested an antiviral mechanism that prevented viral
entry or fusion (Monell and Strand 1994). More recently, an elegant study revealed
that HNP1 had a dual mode of action against HIV-1 by interfering with protein
kinase C activity as well as acting directly on the virus (Chang et al. 2005). Although
120 A.M. Cole and A.L. Cole
HNPs are intrinsically antiretroviral, these peptides are only liberated from neutro-
phils, and thus, their presence also signifies subclinical or clinical inflammation.
Similar to the upregulation of HBD2 and HBD3 that typifies epithelial inflamma-
tion, neutrophil-mediated inflammation can in turn render the anogenital mucosa
more susceptible to HIV-1 infection through the recruitment of CD4+ target cells.
The previous section of this chapter described various human cationic antimicrobial
polypeptides that reportedly inhibit HIV-1 infection. Yet as has been observed for
defensins, the presence of HNPs and HBDs in the anogenital mucosa could poten-
tially be a double-edged sword, more carefully honed on the side of inflammation-
mediated enhancement of HIV-1 infection. Two other α-defensins reportedly
augment HIV-1 infection. Originally isolated from Paneth cells located at the base of
the crypts of Lieberkühn within the small intestine (Jones and Bevins 1992, 1993;
Ouellette 2005), HD5 and HD6 were found to be stored as inactive propeptides and
proteolytically activated by trypsin upon release of secretory granules (Ghosh et al.
2002). Similar to other defensins, these α-defensins have classically been considered
broad-spectrum antimicrobial peptides. In the female reproduction tract (FRT), HD5
has been localized to vaginal and ectocervical epithelia, granules within endocervi-
cal epithelium, and the surface of the endocervix (Quayle et al. 1998). Expression of
HD5 is highest during the secretory phase of the menstrual cycle (Quayle et al.
1998), suggesting hormonal influence on expression and periods in which the FRT,
particularly the cervix, is potentially more susceptible to HIV-1 infection. While all
other defensins have been shown to inhibit HIV-1 infection in vitro, HD5 and HD6
were instead found to enhance HIV-1 infection by promoting viral attachment to
target cells (Rapista et al. 2011) and could also antagonize the anti-HIV-1 activity of
polyanion topical microbicides (Ding et al. 2011). Given that the cervix is likely the
primary site for initial HIV-1 infection in women (Miller et al. 2005; Pudney et al.
2005; Salazar-Gonzalez et al. 2009), HD5 and HD6 expressed by cervical epithelia
may be vital determinants for establishing HIV-1 infection in the FRT.
Human semen and seminal plasma contain numerous cationic polypeptides, many
of which are proteolytic fragments of larger proteins. Proteomic analyses of seminal
plasma identified at least 52 separate cationic polypeptides, twenty of which were
derived from the semenogelin I and II parent proteins during the process of semen
8 Dichotomous Roles of Cationic Polypeptides Targeting HIV 121
liquefaction that follows ejaculation (Martellini et al. 2009). From this fluid, an
individual semenogelin-derived peptide “SG-1” was purified, which exhibited
activity against HIV-1 in vitro at sub-physiological concentrations (Martellini et al.
2009). While seminal plasma clearly contains this and potentially other cationic
antiviral components, greater emphasis has been placed on studying cationic poly-
peptides within this fluid that enhance HIV-1 infection. An important study reported
that semen-mediated enhancement of HIV transmission is due to a cationic peptide
“PAP286” derived from prostatic acid phosphatase, which forms amyloid fibrils
called semen-derived enhancers of viral infection or “SEVI” (Munch et al. 2007).
Further in vitro studies confirmed the role of SEVI in enhancing HIV-1 infection
(Roan et al. 2009; Olsen et al. 2010; Easterhoff et al. 2011; Martellini et al. 2011),
implicating that the cationic properties of PAP286 and SEVI underlie this ability
(Roan et al. 2009). The HIV-enhancing effects of SEVI may be diminished in vivo
due to resident proteases, such as prostate-specific antigen (PSA), matriptase and
prostasin, that were shown to cleave PAP286 and prevent SEVI fibril formation
(Martellini et al. 2011). Other studies have suggested that endogenous Zn2+ within
seminal plasma might protect the fibrils from proteolysis (Olsen et al. 2012), which
may explain why SEVI fibrils were detected in fresh human semen, at least for cer-
tain donors (Usmani et al. 2014a, b). Seminal plasma has also been shown to
enhance SEVI fibril formation, in part due to anionic components such as inorganic
phosphate and sodium bicarbonate (Olsen et al. 2012). Cationic polypeptide frag-
ments from semenogelins have also been shown to form SEVI-like amyloid fibrils
that can enhance HIV-1 infection (Roan et al. 2014). Given that many peptides and
proteins can form amyloid fibrils, it is possible that other similar enhancers of HIV-1
infection will be uncovered in semen or perhaps also in cervicovaginal and anorec-
tal fluids. Collectively, while both antiviral and viral-enhancing cationic polypep-
tides have been described in semen and seminal plasma, factors such as SEVI that
promote viral infection might outpace those that act to inhibit infection.
Several other human cationic peptides and proteins have also been reported to
enhance HIV-1 infection. The 18 kDa cathelicidin protein hCAP18 can be proteo-
lytically processed to at least three mature forms, LL-37, ALL-38, and FALL-39,
each of which differs only by one or two amino-terminal residues (Agerberth et al.
1995; Cowland et al. 1995; Larrick et al. 1995; Sorensen et al. 2003). LL-37 is
found in neutrophils and is expressed by a variety of mucosal epithelia, including
the FRT where it has been immunolocalized to inflamed ectocervical epithelia.
While the direct antibacterial actions of LL-37 are well known, this peptide can also
bind the G protein-coupled receptor, N-formyl peptide receptor 2 (FPR2), to exert
chemotactic and immunomodulatory functions. Using this mechanism, LL-37 can
antagonistically ligate FPR2 to downregulate chemokine coreceptors necessary for
122 A.M. Cole and A.L. Cole
HIV-1 to bind and enter peripheral blood mononuclear cells, including primary
CD4+ cells (Bergman et al. 2007). The level of LL-37 in healthy vaginal fluid has
been reported in the mid-to-high nanogram per mL range (Valore et al. 2002), which
is within the concentration necessary to exert FPR2-mediated anti-HIV-1 activity in
vitro. Conversely, LL-37 produced by herpes simplex virus-2 (HSV-2)-infected
keratinocytes has also been reported to increase the susceptibility of Langerhans
cells to HIV-1, which was neutralized by blocking LL-37 production (Ogawa et al.
2013).
Two additional polypeptides from neutrophils can also serve to promote HIV-1
infection. Cathepsin G is a serine protease that has been identified in human cervi-
covaginal fluids (Venkataraman et al. 2005). This protease can enhance HIV-1
infection of macrophages (Moriuchi et al. 2000) likely through its binding of the
gp120 envelope glycoprotein of HIV-1 (Avril et al. 1994, 1995). The mechanism
likely involves Gi protein-mediated signal transduction as cells treated with pertus-
sis toxin did not increase viral infection (Moriuchi et al. 2000). Additionally, cathep-
sin G can cleave RANTES, a natural ligand of the HIV-1 coreceptor CCR5, thus
reducing RANTES’ natural antiviral capacity (Lim et al. 2006). Calprotectin is a
heterodimeric protein composed of an 8 kDa polypeptide (termed MRP-8, S100A8,
or calgranulin A) and a 14 kDa polypeptide (termed MRP-14, S100A9, or calgranu-
lin B) and is produced by neutrophils, monocytes, and various epithelia. Within the
neutrophil, calprotectin accounts for over one-third of the cytoplasmic protein con-
tent (Brandtzaeg et al. 1995). In normal vaginal fluid, its levels are in the low to
mid-microgram per mL range, but due to its presence in neutrophils, much higher
concentrations can be found in inflammatory conditions. A study that isolated the
8 kDa subunit of calprotectin from cervicovaginal secretions revealed that it could
increase expression of HIV-1 in latently infected monocytes (Hashemi et al. 2001).
Interestingly, another study showed that the 14 kDa subunit could stimulate the anti-
HIV-1 activity of NK cells by ligating CD85j (Arnold et al. 2013). Assessing the
activity of both subunits together would be important in determining if one activity
will supersede the other or if their combined effects on HIV-1 infection will be
neutralized. Nevertheless, local mucosal inflammation that induces epithelia-
derived LL-37 and calprotectin and promotes neutrophil influx and granular release
of LL-37, calprotectin, and cathepsin G could serve to increase susceptibility to
HIV-1 infection and transmission.
Since the discovery of cationic antimicrobial polypeptides, the field has expended
much time and energy in promoting the ability of these molecules to serve as broad-
spectrum antibiotics, antifungals, and antivirals – with good reason. Many of these
polypeptides are highly effective against microbes and viruses, and a good number
could serve as scaffolds for the design and development of next-generation therapeu-
tics and preventatives to combat pathogens that are resistant to current antibiotic
8 Dichotomous Roles of Cationic Polypeptides Targeting HIV 123
References
Draper DL, Landers DV, Krohn MA, Hillier SL, Wiesenfeld HC, Heine RP (2000) Levels of vagi-
nal secretory leukocyte protease inhibitor are decreased in women with lower reproductive
tract infections. Am J Obstet Gynecol 183(5):1243–1248
Easterhoff D, DiMaio JT, Doran TM, Dewhurst S, Nilsson BL (2011) Enhancement of HIV-1
infectivity by simple, self-assembling modular peptides. Biophys J 100(5):1325–1334
Feng Z, Dubyak GR, Lederman MM, Weinberg A (2006) Cutting edge: human beta defensin 3 – a
novel antagonist of the HIV-1 coreceptor CXCR4. J Immunol 177(2):782–786
Fleming A (1922) On a remarkable bacteriolytic element found in tissues and secretions. Proc R
Soc Lond [Biol] 93:306–317
Gallo RL (2013) The birth of innate immunity. Exp Dermatol 22(8):517
Gallo SA, Wang W, Rawat SS, Jung G, Waring AJ, Cole AM, Lu H, Yan X, Daly NL, Craik DJ,
Jiang S, Lehrer RI, Blumenthal R (2006) Theta-defensins prevent HIV-1 Env-mediated fusion
by binding gp41 and blocking 6-helix bundle formation. J Biol Chem 281(27):18787–18792
Ganz T (2003) Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol
3(9):710–720
Ganz T, Lehrer RI (1997) Antimicrobial peptides of leukocytes. Curr Opin Hematol 4(1):53–58
Ghosh D, Porter E, Shen B, Lee SK, Wilk D, Drazba J, Yadav SP, Crabb JW, Ganz T, Bevins CL
(2002) Paneth cell trypsin is the processing enzyme for human defensin-5. Nat Immunol
3(6):583–590
Ghosh M, Shen Z, Fahey JV, Cu-Uvin S, Mayer K, Wira CR (2010) Trappin-2/Elafin: a novel
innate anti-human immunodeficiency virus-1 molecule of the human female reproductive tract.
Immunology 129(2):207–219
Gupta P, Lackman-Smith C, Snyder B, Ratner D, Rohan LC, Patton D, Ramratnam B, Cole AM
(2013) Antiviral activity of retrocyclin RC-101, a candidate microbicide against cell-associated
HIV-1. AIDS Res Hum Retroviruses 29(2):391–396
Haase AT (2011) Early events in sexual transmission of HIV and SIV and opportunities for inter-
ventions. Annu Rev Med 62:127–139
Harwig SS, Park AS, Lehrer RI (1992) Characterization of defensin precursors in mature human
neutrophils. Blood 79(6):1532–1537
Hashemi FB, Mollenhauer J, Madsen LD, Sha BE, Nacken W, Moyer MB, Sorg C, Spear GT
(2001) Myeloid-related protein (MRP)-8 from cervico-vaginal secretions activates HIV repli-
cation. AIDS 15(4):441–449
Haynes BF, Moody MA, Alam M, Bonsignori M, Verkoczy L, Ferrari G, Gao F, Tomaras GD, Liao
HX, Kelsoe G (2014) Progress in HIV-1 vaccine development. J Allergy Clin Immunol 134(1):
3–10
Hein M, Valore EV, Helmig RB, Uldbjerg N, Ganz T (2002) Antimicrobial factors in the cervical
mucus plug. Am J Obstet Gynecol 187(1):137–144
Hoover DM, Boulegue C, Yang D, Oppenheim JJ, Tucker K, Lu W, Lubkowski J (2002) The struc-
ture of human macrophage inflammatory protein-3alpha /CCL20. Linking antimicrobial and
CC chemokine receptor-6-binding activities with human beta-defensins. J Biol Chem 277(40):
37647–37654
Horne AW, Stock SJ, King AE (2008) Innate immunity and disorders of the female reproductive
tract. Reproduction 135(6):739–749
Jones DE, Bevins CL (1992) Paneth cells of the human small intestine express an antimicrobial
peptide gene. J Biol Chem 267(32):23216–23225
Jones DE, Bevins CL (1993) Defensin-6 mRNA in human Paneth cells: implications for antimicro-
bial peptides in host defense of the human bowel. FEBS Lett 315(2):187–192
Klotman ME, Chang TL (2006) Defensins in innate antiviral immunity. Nat Rev Immunol 6(6):
447–456
Larrick JW, Hirata M, Balint RF, Lee J, Zhong J, Wright SC (1995) Human CAP18: a novel anti-
microbial lipopolysaccharide-binding protein. Infect Immun 63(4):1291–1297
Lee-Huang S, Huang PL, Sun Y, Huang PL, Kung HF, Blithe DL, Chen HC (1999) Lysozyme and
RNases as anti-HIV components in beta-core preparations of human chorionic gonadotropin.
Proc Natl Acad Sci U S A 96(6):2678–2681
8 Dichotomous Roles of Cationic Polypeptides Targeting HIV 125
Lee-Huang S, Maiorov V, Huang PL, Ng A, Lee HC, Chang YT, Kallenbach N, Huang PL, Chen
HC (2005) Structural and functional modeling of human lysozyme reveals a unique nonapep-
tide, HL9, with anti-HIV activity. Biochemistry 44(12):4648–4655
Lehrer RI, Ganz T (2002) Defensins of vertebrate animals. Curr Opin Immunol 14(1):96–102
Lim JK, Lu W, Hartley O, DeVico AL (2006) N-terminal proteolytic processing by cathepsin G
converts RANTES/CCL5 and related analogs into a truncated 4–68 variant. J Leukoc Biol
80(6):1395–1404
Mansour SC, Pena OM, Hancock RE (2014) Host defense peptides: front-line immunomodulators.
Trends Immunol 35(9):443–450
Martellini JA, Cole AL, Venkataraman N, Quinn GA, Svoboda P, Gangrade BK, Pohl J, Sorensen
OE, Cole AM (2009) Cationic polypeptides contribute to the anti-HIV-1 activity of human
seminal plasma. FASEB J Off Publ Federation Am Soc Exp Biol 23(10):3609–3618
Martellini JA, Cole AL, Svoboda P, Stuchlik O, Chen LM, Chai KX, Gangrade BK, Sorensen OE,
Pohl J, Cole AM (2011) HIV-1 enhancing effect of prostatic acid phosphatase peptides is
reduced in human seminal plasma. PLoS One 6(1):e16285
McMahon JM, Myers JE, Kurth AE, Cohen SE, Mannheimer SB, Simmons J, Pouget ER, Trabold
N, Haberer JE (2014) Oral Pre-Exposure Prophylaxis (PrEP) for Prevention of HIV in
Serodiscordant Heterosexual Couples in the United States: Opportunities and Challenges.
AIDS Patient Care STDS 28(9):462–474
McNeely TB, Dealy M, Dripps DJ, Orenstein JM, Eisenberg SP, Wahl SM (1995) Secretory leu-
kocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency
virus 1 activity in vitro. J Clin Invest 96(1):456–464
McNeely TB, Shugars DC, Rosendahl M, Tucker C, Eisenberg SP, Wahl SM (1997) Inhibition of
human immunodeficiency virus type 1 infectivity by secretory leukocyte protease inhibitor
occurs prior to viral reverse transcription. Blood 90(3):1141–1149
Miller CJ, Li Q, Abel K, Kim EY, Ma ZM, Wietgrefe S, La Franco-Scheuch L, Compton L, Duan
L, Shore MD, Zupancic M, Busch M, Carlis J, Wolinsky S, Haase AT (2005) Propagation and
dissemination of infection after vaginal transmission of simian immunodeficiency virus. J Virol
79(14):9217–9227
Monell CR, Strand M (1994) Structural and functional similarities between synthetic HIV gp41
peptides and defensins. Clin Immunol Immunopathol 71(3):315–324
Moreau T, Baranger K, Dade S, Dallet-Choisy S, Guyot N, Zani ML (2008) Multifaceted roles of
human elafin and secretory leukocyte proteinase inhibitor (SLPI), two serine protease inhibi-
tors of the chelonianin family. Biochimie 90(2):284–295
Moriuchi H, Moriuchi M, Fauci AS (2000) Cathepsin G, a neutrophil-derived serine protease,
increases susceptibility of macrophages to acute human immunodeficiency virus type 1 infec-
tion. J Virol 74(15):6849–6855
Morris GC, Lacey CJ (2010) Microbicides and HIV prevention: lessons from the past, looking to
the future. Curr Opin Infect Dis 23(1):57–63
Munch J, Rucker E, Standker L, Adermann K, Goffinet C, Schindler M, Wildum S, Chinnadurai
R, Rajan D, Specht A, Gimenez-Gallego G, Sanchez PC, Fowler DM, Koulov A, Kelly JW,
Mothes W, Grivel JC, Margolis L, Keppler OT, Forssmann WG, Kirchhoff F (2007) Semen-
derived amyloid fibrils drastically enhance HIV infection. Cell 131(6):1059–1071
Munk C, Wei G, Yang OO, Waring AJ, Wang W, Hong T, Lehrer RI, Landau NR, Cole AM (2003)
The theta-defensin, retrocyclin, inhibits HIV-1 entry. AIDS Res Hum Retroviruses 19(10):
875–881
Nakashima H, Yamamoto N, Masuda M, Fujii N (1993) Defensins inhibit HIV replication in vitro.
AIDS 7(8):1129
Nguyen TX, Cole AM, Lehrer RI (2003) Evolution of primate theta-defensins: a serpentine path to
a sweet tooth. Peptides 24(11):1647–1654
Ogawa Y, Kawamura T, Matsuzawa T, Aoki R, Gee P, Yamashita A, Moriishi K, Yamasaki K,
Koyanagi Y, Blauvelt A, Shimada S (2013) Antimicrobial peptide LL-37 produced by HSV-2-
infected keratinocytes enhances HIV infection of Langerhans cells. Cell Host Microbe
13(1):77–86
126 A.M. Cole and A.L. Cole
Olsen JS, Brown C, Capule CC, Rubinshtein M, Doran TM, Srivastava RK, Feng C, Nilsson BL,
Yang J, Dewhurst S (2010) Amyloid-binding small molecules efficiently block SEVI (semen-
derived enhancer of virus infection)- and semen-mediated enhancement of HIV-1 infection.
J Biol Chem 285(46):35488–35496
Olsen JS, DiMaio JT, Doran TM, Brown C, Nilsson BL, Dewhurst S (2012) Seminal plasma accel-
erates semen-derived enhancer of viral infection (SEVI) fibril formation by the prostatic acid
phosphatase (PAP248-286) peptide. J Biol Chem 287(15):11842–11849
Ouellette AJ (2005) Paneth cell alpha-defensins: peptide mediators of innate immunity in the small
intestine. Springer Semin Immunopathol 27(2):133–146
Owen SM, Rudolph DL, Wang W, Cole AM, Waring AJ, Lal RB, Lehrer RI (2004) RC-101, a
retrocyclin-1 analogue with enhanced activity against primary HIV type 1 isolates. AIDS Res
Hum Retroviruses 20(11):1157–1165
Pillay K, Coutsoudis A, Agadzi-Naqvi AK, Kuhn L, Coovadia HM, Janoff EN (2001) Secretory
leukocyte protease inhibitor in vaginal fluids and perinatal human immunodeficiency virus type
1 transmission. J Infect Dis 183(4):653–656
Pudney J, Quayle AJ, Anderson DJ (2005) Immunological microenvironments in the human
vagina and cervix: mediators of cellular immunity are concentrated in the cervical transforma-
tion zone. Biol Reprod 73(6):1253–1263
Quayle AJ, Porter EM, Nussbaum AA, Wang YM, Brabec C, Yip KP, Mok SC (1998) Gene
expression, immunolocalization, and secretion of human defensin-5 in human female repro-
ductive tract. Am J Pathol 152(5):1247–1258
Quinones-Mateu ME, Lederman MM, Feng Z, Chakraborty B, Weber J, Rangel HR, Marotta ML,
Mirza M, Jiang B, Kiser P, Medvik K, Sieg SF, Weinberg A (2003) Human epithelial beta-
defensins 2 and 3 inhibit HIV-1 replication. AIDS 17(16):F39–F48
Rapista A, Ding J, Benito B, Lo YT, Neiditch MB, Lu W, Chang TL (2011) Human defensins 5
and 6 enhance HIV-1 infectivity through promoting HIV attachment. Retrovirology 8:45
Roan NR, Munch J, Arhel N, Mothes W, Neidleman J, Kobayashi A, Smith-McCune K, Kirchhoff
F, Greene WC (2009) The cationic properties of SEVI underlie its ability to enhance human
immunodeficiency virus infection. J Virol 83(1):73–80
Roan NR, Liu H, Usmani SM, Neidleman J, Muller JA, Avila-Herrera A, Gawanbacht A, Zirafi O,
Chu S, Dong M, Kumar ST, Smith JF, Pollard KS, Fandrich M, Kirchhoff F, Munch J,
Witkowska HE, Greene WC (2014) Liquefaction of semen generates and later degrades a con-
served semenogelin peptide that enhances HIV infection. J Virol 88(13):7221–7234
Rohrl J, Yang D, Oppenheim JJ, Hehlgans T (2010) Human beta-defensin 2 and 3 and their mouse
orthologs induce chemotaxis through interaction with CCR2. J Immunol 184(12):6688–6694
Salazar-Gonzalez JF, Salazar MG, Keele BF, Learn GH, Giorgi EE, Li H, Decker JM, Wang S,
Baalwa J, Kraus MH, Parrish NF, Shaw KS, Guffey MB, Bar KJ, Davis KL, Ochsenbauer-
Jambor C, Kappes JC, Saag MS, Cohen MS, Mulenga J, Derdeyn CA, Allen S, Hunter E,
Markowitz M, Hraber P, Perelson AS, Bhattacharya T, Haynes BF, Korber BT, Hahn BH, Shaw
GM (2009) Genetic identity, biological phenotype, and evolutionary pathways of transmitted/
founder viruses in acute and early HIV-1 infection. J Exp Med 206(6):1273–1289
Sorensen OE, Gram L, Johnsen AH, Andersson E, Bangsboll S, Tjabringa GS, Hiemstra PS, Malm
J, Egesten A, Borregaard N (2003) Processing of seminal plasma hCAP-18 to ALL-38 by
gastricsin: a novel mechanism of generating antimicrobial peptides in vagina. J Biol Chem
278(31):28540–28546
Steinrauf LK, Shiuan D, Yang WJ, Chiang MY (1999) Lysozyme association with nucleic acids.
Biochem Biophys Res Commun 266(2):366–370
Tang YQ, Yuan J, Osapay G, Osapay K, Tran D, Miller CJ, Ouellette AJ, Selsted ME (1999) A
cyclic antimicrobial peptide produced in primate leukocytes by the ligation of two truncated
alpha-defensins. Science 286(5439):498–502
Trifonova RT, Pasicznyk JM, Fichorova RN (2006) Biocompatibility of solid-dosage forms of anti-
human immunodeficiency virus type 1 microbicides with the human cervicovaginal mucosa
modeled ex vivo. Antimicrob Agents Chemother 50(12):4005–4010
8 Dichotomous Roles of Cationic Polypeptides Targeting HIV 127
Turpin JA, Schaeffer CA, Bu M, Graham L, Buckheit RW Jr, Clanton D, Rice WG (1996) Human
immunodeficiency virus type-1 (HIV-1) replication is unaffected by human secretory leukocyte
protease inhibitor. Antiviral Res 29(2–3):269–277
Usmani S, Liu H, Pilcher CD, Witkowska HE, Kirchhoff F, Greene WC, Munch J, Roan NR
(2014a) HIV-enhancing amyloids are prevalent in fresh semen and are a determinant for
Semen’s ability to enhance HIV infection: relevance for HIV transmission. AIDS Res Hum
Retroviruses 30(Suppl 1):A183–A184
Usmani SM, Zirafi O, Muller JA, Sandi-Monroy NL, Yadav JK, Meier C, Weil T, Roan NR,
Greene WC, Walther P, Nilsson KP, Hammarstrom P, Wetzel R, Pilcher CD, Gagsteiger F,
Fandrich M, Kirchhoff F, Munch J (2014b) Direct visualization of HIV-enhancing endogenous
amyloid fibrils in human semen. Nat Commun 5:3508
Valore EV, Ganz T (1992) Posttranslational processing of defensins in immature human myeloid
cells. Blood 79(6):1538–1544
Valore EV, Park CH, Igreti SL, Ganz T (2002) Antimicrobial components of vaginal fluid. Am
J Obstet Gynecol 187(3):561–568
Van Damme L, Szpir M (2012) Current status of topical antiretroviral chemoprophylaxis. Curr
Opin HIV AIDS 7(6):520–525
Venkataraman N, Cole AL, Svoboda P, Pohl J, Cole AM (2005) Cationic polypeptides are required
for anti-HIV-1 activity of human vaginal fluid. J Immunol 175(11):7560–7567
Venkataraman N, Cole AL, Ruchala P, Waring AJ, Lehrer RI, Stuchlik O, Pohl J, Cole AM (2009)
Reawakening retrocyclins: ancestral human defensins active against HIV-1. PLoS Biol 7(4):e95
Zasloff M (1992) Antibiotic peptides as mediators of innate immunity. Curr Opin Immunol
4(1):3–7
Chapter 9
Antimicrobial Peptides in Host Defense:
Functions Beyond Antimicrobial Activity
Abstract Antimicrobial peptides are well known for their important roles in
host defense by enhancing the barrier function and limiting microbial popula-
tions of the skin and mucosa. However, many of these peptides are now known to
have additional roles assisting innate and adaptive immune functions. To facili-
tate innate immunity, antimicrobial peptides activate complement, chemoattract
cells (e.g., monocytes, macrophages, T cells, neutrophils, immature dendritic
cells, and mast cells), enhance phagocytosis, and modulate the production of
chemokines and proinflammatory cytokines in other cells. At local sites of initia-
tion, antimicrobial peptides can act as opsonins to enhance phagocytosis by
monocytes and phagocytes and can activate cells. In the latter, for example, treat-
ment of osteoblasts and osteoblast-like MG63 cells with human beta-defensin
(HBD)2 increases their proliferation rates. Treatment of osteoblast-like MG63
cells with HBD2 and HBD3 increases transcript levels of osteogenic markers for
differentiation, increases antileukoprotease (ALP) levels, and enhances mineral-
ized nodule formation. To facilitate adaptive immunity, antimicrobial peptides
assist the uptake of antigens by monocytes or other antigen-presenting cells and
later direct the process toward a Th1 or Th2 adaptive immune response. More
commonly though, antimicrobial peptides induce a mixed response characterized
by Th1-/Th2-specific antibodies and Th1/Th2 cytokines from antigen-exposed
splenocytes of immunized animals. Finally, antimicrobial peptides can be
detected in the margins around both oral and cutaneous wounds, and there is
growing evidence to suggest they also play a dynamic role in wound healing by
improving wound angiogenesis, vascularization, and reepithelialization.
9.1 Introduction
Antimicrobial factors in normal tissues and fluids were described as early as 1888
(Skarnes and Watson 1957). These factors, isolated from extracts of tissues, serum,
serous fluids, and leukocytes, later became the well-known members of innate
immunity: antibodies, complement, lysozyme, histones, and protamines (Skarnes
and Watson 1957). Small, linear, basic peptides (called tissue basic polypeptides)
with antimicrobial activity in normal tissues and fluids were also described as early
as 1947 (Bloom et al. 1947; Bloom and Prigmore 1952; Bloom and Blake 1948).
These peptides contained lysine (29–30 %) and arginine (3.5 %) amino acid resi-
dues with isoelectric points between pI 10 and 11.2. They were thought to be
attracted to the negatively charged surfaces of microbial cells via electrostatic bond-
ing and to alter microbial membrane integrity. These peptides are likely the group
we now know as the antimicrobial peptides (Skarnes and Watson 1957). The early
history of antimicrobial peptide discovery and research can be found in two excel-
lent reviews by Skarnes and Watson (1957) and Nakatsuji and Gallo (2012).
Almost immediately after the discovery of cationic peptides with antimicrobial
activity, investigators began to assess their secondary functions, and many of these
peptides did indeed have additional roles in innate and adaptive immunology
(Nakatsuji and Gallo 2012). This was not an unusual finding as the inverse was also
found to be true, and some other physiologically important peptides had antimicro-
bial activity. For example, some neuropeptides, peptide hormones, and chemokines
were found to have antimicrobial activities (Brogden et al. 2005; Cole et al. 2001;
Yang et al. 2003). These results clearly suggest that peptides with antimicrobial
activity are multifunctional in a variety of situations.
In this chapter, we present the alternate functions of peptides with antimicrobial
activity, a topic of a number of excellent comprehensive reviews (Yang et al. 2001,
2002, 2004; Yang and Oppenheim 2004; Bowdish et al. 2005; Pingel et al. 2007;
Rehaume and Hancock 2008; Semple et al. 2010; Semple and Dorin 2012; Greer
et al. 2013). We start by presenting the roles of antimicrobial peptides in innate
immunity and their ability to chemoattract and activate cells (Table 9.1). This
includes recent discoveries that antimicrobial peptides influence the properties of
human mesenchymal stem cells (hMSCs) and osteoblasts in addition to epithelial
cells, keratinocytes, and immune cells of myeloid or lymphoid origin. We then pres-
ent the roles of antimicrobial peptides in adaptive immunity and their ability to
influence Th1, Th2, and mixed Th1 and Th2 responses (Table 9.2). Third, we pres-
ent the ability and conditions of antimicrobial peptides to modulate chemokine and
proinflammatory cytokine responses, an exciting area of current research by a vari-
ety of investigators (Table 9.3). Finally, we present the roles of antimicrobial pep-
tides in wound healing, angiogenesis, and autoimmune diseases.
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 131
Table 9.2 Antimicrobial peptides influence Th1, Th2, and mixed Th1/Th2 adaptive immune
responses, often with adjuvant-like activities
Enhance Th1 responses
Murine β-defensin 2 (mDF2β) induces potent cell-mediated responses and antitumor
immunity when genetically fused with nonimmunogenic tumor antigens (Biragyn et al.
2001, 2002; Biragyn 2005)
Mice receiving L1210 cells expressing mDF2β have responses strong NK and CTL responses
with enhanced IL-12 and IFN-γ production, protecting them from lethal challenge with
L1210 cells (Ma et al. 2006)
Zebra fish immunized with zebra fish β-defensin 2 (zfBD2) develops a Th1 immune response
with an upregulated IFN-γ response (Garcia-Valtanen et al. 2014)
Enhance Th2 responses
Cationic peptide KLKL5KLK with ovalbumin induces a Th2 response. Immunized mice
produce ovalbumin-specific IgG1, but not IgG2, and splenocytes from immunized mice,
stimulated with ovalbumin, produce IL-4 and IL-5, but not IFN-γ (Fritz et al. 2004)
Enhance mixed Th1/Th2 responses
Melittin enhances a mixed Th1/Th2 response to tetanus toxoid in mice. Total IgG and IgG2a
responses are increased (Bramwell et al. 2003)
CRAMP enhances ovalbumin-specific IgG1, IgG2a, IgG2b, and IgG3 and Th1/Th2 cellular
ovalbumin-specific responses in mice (Kurosaka et al. 2005)
LL-37 enhances Th1 and Th2 humoral, cytotoxic, and protective responses in mice when
fused with M-CSF receptor cloned from J6-1 leukemia cells (M-CSFRJ6-1). Splenocytes
from immunized mice, stimulated with M-CSFRJ6-1, produce IFN-γ (An et al. 2005)
HNP-1, HNP-2, HNP-3 enhance keyhole limpet hemocyanin-specific and ovalbumin-
specific IgG1, IgG2a, and IgG2b responses, and splenocytes from immunized mice
stimulated with keyhole limpet hemocyanin produce KLH-specific IFN-γ (Th1 cytokine)
and IL-4 (Th2 cytokine), and splenocytes from immunized mice stimulated with
ovalbumin produce greater amounts of IL-5, IL-6, IL-10, and IFN-γ (Th1 and Th2
cytokines) (Lillard et al. 1999; Tani et al. 2000)
11, 14–17, 20, 23, and 25) and the location of claudins in cell membranes and ele-
vates transepithelial electrical resistance (Kiatsurayanon et al. 2014). This occurs
via HBD3-mediated pathways involving Rac1, atypical protein kinase C, glycogen
synthase kinase, and phosphatidylinositol 3-kinase (Kiatsurayanon et al. 2014).
Once produced, cathelicidins and defensins then attract a variety of cells to their
sites of induction (Table 9.1). LL-37, CRAMP, α-defensins, and β-defensins all are
reported to chemoattract monocytes, macrophages, T cells, neutrophils, immature
dendritic cells, and mast cells.
At the site of initiation, antimicrobial peptides then can act as opsonins
(Fleischmann et al. 1985), enhance phagocytosis by monocytes and phagocytes
(Ichinose et al. 1996), and activate cells. They can also degranulate mast cells. For
example, human, rabbit, and guinea pig α-defensins activate and degranulate mast
cells releasing histamine and prostaglandin D2 (Yamashita and Saito 1989; Befus
et al. 1999; Niyonsaba et al. 2001).
An additional and well-known function is the ability of antimicrobial peptides,
particularly defensins, to regulate complement activation (Prohaszka et al. 1997;
van den Berg et al. 1998). Here, C1q of C1 binds to HNP-1, HNP-2, and HNP-3,
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 133
Table 9.3 Peptides and proteins in oral secretions with antiinflammatory and proinflammatory
properties
Antiinflammatory activities
LL-37
Attenuates agonist-induced, chemokine, and proinflammatory cytokine responses in
macrophages (Scott et al. 2000), lung epithelial cells (Scott et al. 2000), peripheral blood
mononuclear cells (Molhoek et al. 2009), and whole blood leukocytes (Walters et al. 2010)
Attenuates MAPK pathway activation of p38 and ERK responses in gingival fibroblasts
(Inomata et al. 2010)
α-defensins
Inhibit the production of proinflammatory cytokines from macrophages (Miles et al. 2009)
Attenuate a chemokine and proinflammatory cytokine response in mice (Kohlgraf et al. 2010)
β-defensins
HBD3 attenuates agonist-induced, chemokine, and proinflammatory cytokine responses in
dendritic cells (Pingel et al. 2008; Harvey et al. 2013), in THP-1 human myelomonocytic
cells (Semple et al. 2010), peripheral blood monocyte-derived macrophages (Semple et al.
2010), and in RAW264.7 murine macrophages (Semple et al. 2010)
DEFB114 attenuates MAPK pathway p42/44 response and attenuates an agonist-induced
TNF-α response in RAW264.7 murine macrophages (Yu et al. 2013)
DEFB123 attenuates an agonist-induced MAPK pathway activation of p42/44 and p38 and
attenuates an agonist-induced TNF-α response in RAW264.7 murine macrophages
(Motzkus et al. 2006)
DEFB126 attenuates an agonist-induced proinflammatory response in RAW264.7 murine
macrophages (Liu et al. 2013a)
Attenuate a chemokine and proinflammatory cytokine response in mice (Kohlgraf et al. 2010)
θ-defensins
Retrocyclin RTD-1 attenuates agonist-induced, chemokine, and proinflammatory cytokine
responses in human peripheral blood leukocytes (Schaal et al. 2012)
Histatins
Histatin 5 attenuates agonist-induced, chemokine, and proinflammatory cytokine responses in
gingival fibroblasts (Imatani et al. 2000) and dendritic cells (Borgwardt et al. 2014)
CEMA (cecropin-melittin hybrid)
CEMA blocks the binding of LPS to LPS-binding protein, attenuates agonist-induced,
chemokine, and proinflammatory cytokine responses in murine macrophages (Scott et al.
2000)
Proinflammatory activities
LL-37
50–100 μg/ml enhances an agonist-induced IL-8 response in epithelial cells (Scott et al. 2002)
β-defensins
Pre-stimulation or post-stimulation of dendritic cells and mice with HBD3 enhances an
agonist-induced chemokine and proinflammatory cytokine response (Harvey et al. 2013)
Pre-stimulation of macrophages with MBD14 enhances an agonist-induced chemokine and
proinflammatory cytokine response (Barabas et al. 2013)
HD5 upregulates expression of genes involved in cell survival and inflammation in an
NF-kB-dependent fashion in epithelial cells (Lu and de Leeuw 2013)
134 K.A. Brogden et al.
Zebra fish immunized with a plasmid encoding zebra fish β-defensin 2 (zfBD2),
and the glycoprotein G of the spring viremia of carp virus (gpGsvcv) developed a Th1
immune response with an upregulated IFN-γ response (Garcia-Valtanen et al. 2014).
Expression of zfBD2 upregulated IS mx genes related to the activation of the type I
IFN system. It also induced the transcription of proinflammatory cytokine genes
tnfα and il1β, increased the presence of mhc2 transcripts related to MHC class II
presentation of antigens, enhanced granzyme and NK lysine transcripts related to
immune cytotoxic responses cytotoxic responses, and mediated recruitment of Th
cells at the injection site.
More commonly than the above two examples, antimicrobial peptides induce
immunized animals to produce a mixed response characterized by producing Th1-/
Th2-specific antibodies and Th1/Th2 cytokines from antigen-exposed splenocytes
of immunized animals. Melittin from bees enhances a mixed Th1/Th2 response to
tetanus toxoid in mice. Melittin increases tetanus toxoid total IgG and IgG2a anti-
body responses (Bramwell et al. 2003). Similarly CRAMP enhances mixed Th1/
Th2 antigen-specific immune responses to ovalbumin in mice. CRAMP increases
ovalbumin IgG1, IgG2a, IgG2b, and IgG3 antibody responses (Kurosaka et al.
2005). LL-37 enhances a mixed Th1/Th2 humoral, cytotoxic, and protective
response in mice when LL-37 was fused with M-CSFRJ6-1, an M-CSF receptor
cloned from J6-1 leukemia cells (M-CSFRJ6-1). Splenocytes from immunized mice,
stimulated with M-CSFRJ6-1, produced IFN-γ (An et al. 2005).
A mixed Th1/Th2 response is also induced by defensins. HNP-1, HNP-2, and
HNP-3 enhance keyhole limpet hemocyanin (KLH) IgG1, IgG2a, and IgG2b anti-
body responses (Tani et al. 2000), and HNP-1, HNP-2, and HNP-3 and human
β-defensins enhance ovalbumin-specific IgG1, IgG2a, and IgG2b antibody
responses (Lillard et al. 1999; Brogden et al. 2003). T cells from KLH-immunized
mice, stimulated with KLH, produce KLH-specific IFN-γ (Th1 cytokine) and IL-4
(Th2 cytokine) (Tani et al. 2000). T cells from ovalbumin immunized mice, stimu-
lated with ovalbumin, produce greater amounts of CD4+ Th1 and Th2 cytokines
(IFN-γ, IL-5, IL-6, and IL-10) (Lillard et al. 1999; Brogden et al. 2003).
136 K.A. Brogden et al.
Cells treated with antimicrobial peptides alone or with a microbial antigen have
both proinflammatory and antiinflammatory activities: a dichotomy that is not
entirely well understood (Harvey et al. 2013). These proinflammatory and antiin-
flammatory activities appear to be dependent upon a number of conditions that
include antimicrobial peptide concentration, antimicrobial peptide association with
proinflammatory agonists, or the temporal order of peptide exposure to cells, with
respect to agonist exposure to cells.
with 50.0–100.0 μg/ml LL-37 produce 200, 400, and >1,000 pg/ml MCP-1 (CCL2)
(Scott et al. 2002); and A549 human epithelial cells treated with 10–100 μg/ml
LL-37 produce 300–1,200 pg/ml IL-8 (Scott et al. 2002). Epithelial cells treated
with 100.0 μg/ml HNP-1, HNP-2, and HNP-3 induce ~17,000 pg/ml IL-8 (Van
Wetering et al. 1997).
This, too, is a recent and very exciting area of research. Antimicrobial peptides
given before or after a proinflammatory agonist induce cells to produce higher con-
centrations of proinflammatory mediators. When given together, LL-37 attenuates
LPS-induced TNF-α responses (Scott et al. 2011; Brown et al. 2011). However,
when THP-1 monocytes are treated with LL-37 for 1 h before the addition of LPS
(Scott et al. 2011) or 3 h after incubation with LPS (Brown et al. 2011), attenuation
is abolished. In yet another example, pre-mixing of leukocytes and E. coli for up to
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 139
Defensins and LL-37 bind to CpG, self-DNA, and RNA (Tewary et al. 2013; Frasca
and Lande 2012; Lande et al. 2007; Gilliet and Lande 2008). This interaction forms
complexes. HBD3 + human genomic DNA forms large complex DNA nets (Tewary
et al. 2013), and LL-37 + DNA forms aggregated and condensed structures (Lande
et al. 2007). These complexes are readily taken up by plasmacytoid dendritic cells in
a TLR9 (e.g., HBD3/DNA; LL-37/DNA)-dependent manner in the endocytotic path-
way and induce the production of IFN-α (Tewary et al. 2013; Lande et al. 2007). In
mice, CpG + HBD3 complexes administered intravenously alone induce proinflam-
matory cytokines in serum, administered subcutaneously alone induce the formation
of local inflammatory cell infiltrates, and administered intraperitoneally with an
immunogen like ovalbumin enhance both cellular and humoral responses to ovalbu-
min. Nakatsuji and Gallo suggest that this is an important normal physiological and
immunological function leading to the attraction of various immune cells (Nakatsuji
and Gallo 2012). Tewary and colleagues suggest that these complexes could improve
vaccine formulations and enhance immune responses (Tewary et al. 2013). However,
they also point out that these complexes are found to be a constituent of circulating
immune complexes isolated from sera in patients with autoimmune diseases (Tewary
et al. 2013). Frasca and Lande (2012) and Gilliet and Lande (2008) also suggest that
this mechanism may lead to autoimmune and autoinflammatory diseases.
There is a growing body of evidence that suggests defensins and LL-37 have
roles in autoimmune and autoinflammatory diseases like psoriasis, rosacea, ulcer-
ative colitis, rheumatic joint disease, and systemic lupus erythematosus (Frasca and
Lande 2012; Vordenbaumen et al. 2010). Subjects with autoimmune disease have
increased circulating concentrations of α- and β-defensins (Vordenbaumen et al.
2010). In the sera of subjects with systemic lupus erythematosus, concentrations of
HBD2 correlate with red blood cell count, dsDNA antibody titers, systemic lupus
erythematosus disease activity index, and clinical transverse myelitis and myositis
(Vordenbaumen et al. 2010). Similarly, serum HNP concentration correlates with
subject white blood cell counts and clinical transverse myelitis and rash
(Vordenbaumen et al. 2010). The relative amounts of HNP mRNA from neutrophils
correlate with C3c concentrations, systemic lupus erythematosus disease activity
index, and clinical renal involvement and rash (Vordenbaumen et al. 2010).
Clearly these are interesting findings, and further work is needed to clarify the
roles of defensins and LL-37 (and other antimicrobial peptides) in the pathophysiol-
ogy of autoimmune and autoinflammatory diseases (Frasca and Lande 2012; Gilliet
and Lande 2008).
References
An LL, Yang YH, Ma XT, Lin YM, Li G, Song YH, Wu KF (2005) LL-37 enhances adaptive
antitumor immune response in a murine model when genetically fused with M-CSFR (J6-1)
DNA vaccine. Leuk Res 29(5):535–543
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 141
Barabas N, Rohrl J, Holler E, Hehlgans T (2013) Beta-defensins activate macrophages and syner-
gize in pro-inflammatory cytokine expression induced by TLR ligands. Immunobiology
218(7):1005–1011
Befus AD, Mowat C, Gilchrist M, Hu J, Solomon S, Bateman A (1999) Neutrophil defensins
induce histamine secretion from mast cells: mechanisms of action. J Immunol 163(2):
947–953
Biragyn A (2005) Defensins – non-antibiotic use for vaccine development. Curr Protein Pept Sci
6(1):53–60
Biragyn A, Surenhu M, Yang D, Ruffini PA, Haines BA, Klyushnenkova E, Oppenheim JJ, Kwak
LW (2001) Mediators of innate immunity that target immature, but not mature, dendritic cells
induce antitumor immunity when genetically fused with nonimmunogenic tumor antigens.
J Immunol 167(11):6644–6653
Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A, Chertov O, Shirakawa AK,
Farber JM, Segal DM, Oppenheim JJ, Kwak LW (2002) Toll-like receptor 4-dependent activa-
tion of dendritic cells by beta-defensin 2. Science 298(5595):1025–1029
Bloom WL, Blake FG (1948) Studies on an antibacterial polypeptide extracted from normal tis-
sues. J Infect Dis 83(2):116–123
Bloom WL, Prigmore JR (1952) A method for preparation of antibacterial basic proteins of normal
tissues. J Bacteriol 64(6):855–858
Bloom WL, Watson DW et al (1947) Studies on infection with Bacillus anthracis; preparation and
characterization of an anthracidal substance from various animal tissues. J Infect Dis
80(1):41–52
Borgwardt, D.S. et al. (2014) Histatin 5 binds to Porphyromonas gingivalis hemagglutinin B (HagB)
and alters HagB-induced chemokine responses. Sci. Rep. 4, 3904; DOI:10.1038/srep03904
Bowdish DM, Hancock RE (2005) Anti-endotoxin properties of cationic host defence peptides and
proteins. J Endotoxin Res 11(4):230–236
Bowdish DM, Davidson DJ, Scott MG, Hancock RE (2005) Immunomodulatory activities of small
host defense peptides. Antimicrob Agents Chemother 49(5):1727–1732
Bramwell VW, Somavarapu S, Outschoorn I, Alpar HO (2003) Adjuvant action of melittin follow-
ing intranasal immunisation with tetanus and diphtheria toxoids. J Drug Target
11(8-10):525–530
Brogden KA, Heidari M, Sacco RE, Palmquist D, Guthmiller JM, Johnson GK, Jia HP, Tack BF,
McCray PB (2003) Defensin-induced adaptive immunity in mice and its potential in preventing
periodontal disease. Oral Microbiol Immunol 18(2):95–99
Brogden KA, Guthmiller JM, Salzet M, Zasloff M (2005) The nervous system and innate immu-
nity: the neuropeptide connection. Nat Immunol 6(6):558–564
Brown KL, Poon GF, Birkenhead D, Pena OM, Falsafi R, Dahlgren C, Karlsson A, Bylund J,
Hancock RE, Johnson P (2011) Host defense peptide LL-37 selectively reduces proinflamma-
tory macrophage responses. J Immunol 186(9):5497–5505
Caccavo D, Pellegrino NM, Altamura M, Rigon A, Amati L, Amoroso A, Jirillo E (2002)
Antimicrobial and immunoregulatory functions of lactoferrin and its potential therapeutic
application. J Endotoxin Res 8(6):403–417
Chaly YV, Paleolog EM, Kolesnikova TS, Tikhonov II, Petratchenko EV, Voitenok NN (2000)
Neutrophil alpha-defensin human neutrophil peptide modulates cytokine production in human
monocytes and adhesion molecule expression in endothelial cells. Eur Cytokine Netw
11(2):257–266
Chertov O, Michiel DF, Xu L, Wang JM, Tani K, Murphy WJ, Longo DL, Taub DD, Oppenheim
JJ (1996) Identification of defensin-1, defensin-2, and CAP37/azurocidin as T- cell chemoat-
tractant proteins released from interleukin-8-stimulated neutrophils. J Biol Chem 271(6):
2935–2940
Chertov O, Yang D, Howard OM, Oppenheim JJ (2000) Leukocyte granule proteins mobilize
innate host defenses and adaptive immune responses. Immunol Rev 177:68–78
Cole AM, Ganz T, Liese AM, Burdick MD, Liu L, Strieter RM (2001) Cutting edge: IFN-inducible
ELR- CXC chemokines display defensin-like antimicrobial activity. J Immunol 167(2):
623–627
142 K.A. Brogden et al.
Dietrich DE, Xiao X, Dawson DV, Belanger M, Xie H, Progulske-Fox A, Brogden KA (2008)
Human alpha- and beta-defensins bind to immobilized adhesins from Porphyromonas gingiva-
lis. Infect Immun 76(12):5714–5720
Dunkelberger JR, Song WC (2010) Complement and its role in innate and adaptive immune
responses. Cell Res 20(1):34–50
Fleischmann J, Selsted ME, Lehrer RI (1985) Opsonic activity of MCP-1 and MCP-2, cationic
peptides from rabbit alveolar macrophages. Diagn Microbiol Infect Dis 3(3):233–242
Frasca L, Lande R (2012) Role of defensins and cathelicidin LL37 in auto-immune and auto-
inflammatory diseases. Curr Pharm Biotechnol 13(10):1882–97. Epub ahead of print
Fritz JH, Brunner S, Birnstiel ML, Buschle M, Gabain A, Mattner F, Zauner W (2004) The artifi-
cial antimicrobial peptide KLKLLLLLKLK induces predominantly a TH2-type immune
response to co-injected antigens. Vaccine 22(25-26):3274–3284
Gallo RL, Hooper LV (2012) Epithelial antimicrobial defence of the skin and intestine. Nat Rev
Immunol 12(7):503–516
Garcia-Valtanen P, Martinez-Lopez A, Ortega-Villaizan M, Perez L, Coll JM, Estepa A (2014) In
addition to its antiviral and immunomodulatory properties, the zebrafish beta-defensin 2
(zfBD2) is a potent viral DNA vaccine molecular adjuvant. Antiviral Res 101:136–147
Giesemann T, Guttenberg G, Aktories K (2008) Human alpha-defensins inhibit Clostridium diffi-
cile toxin B. Gastroenterology 134(7):2049–2058
Gilliet M, Lande R (2008) Antimicrobial peptides and self-DNA in autoimmune skin inflamma-
tion. Curr Opin Immunol 20(4):401–407
Gough M, Hancock REW, Kelly NM (1996) Antiendotoxin activity of cationic peptide antimicro-
bial agents. Infect Immun 64(12):4922–4927
Greer A, Zenobia C, Darveau RP (2013) Defensins and LL-37: a review of function in the gingival
epithelium. Periodontol 2000 63(1):67–79
Groot F, Geijtenbeek TB, Sanders RW, Baldwin CE, Sanchez-Hernandez M, Floris R, van Kooyk
Y, de Jong EC, Berkhout B (2005) Lactoferrin prevents dendritic cell-mediated human immu-
nodeficiency virus type 1 transmission by blocking the DC-SIGN – gp120 interaction. J Virol
79(5):3009–3015
Harvey LE, Kohlgraf KG, Mehalick LA, Raina M, Recker EN, Radhakrishnan S, Prasad SA,
Vidva R, Progulske-Fox A, Cavanaugh JE, Vali S, Brogden KA (2013) Defensin DEFB103
bidirectionally regulates chemokine and cytokine responses to a pro-inflammatory stimulus.
Sci Rep 3:1232
Heilborn JD, Nilsson MF, Kratz G, Weber G, Sorensen O, Borregaard N, Stahle-Backdahl M
(2003) The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of
human skin wounds and is lacking in chronic ulcer epithelium. J Invest Dermatol
120(3):379–389
Ichinose M, Asai M, Imai K, Sawada M (1996) Enhancement of phagocytosis by corticostatin I
(CSI) in cultured mouse peritoneal macrophages. Immunopharmacology 35(2):103–109
Imatani T, Kato T, Minaguchi K, Okuda K (2000) Histatin 5 inhibits inflammatory cytokine induc-
tion from human gingival fibroblasts by Porphyromonas gingivalis. Oral Microbiol Immunol
15(6):378–382
Inomata M, Into T, Murakami Y (2010) Suppressive effect of the antimicrobial peptide LL-37 on
expression of IL-6, IL-8 and CXCL10 induced by Porphyromonas gingivalis cells and extracts
in human gingival fibroblasts. Eur J Oral Sci 118(6):574–581
Jan MS, Huang YH, Shieh B, Teng RH, Yan YP, Lee YT, Liao KK, Li C (2006) CC chemokines
induce neutrophils to chemotaxis, degranulation, and alpha-defensin release. J Acquir Immune
Defic Syndr 41(1):6–16
Kiatsurayanon C, Niyonsaba F, Smithrithee R, Akiyama T, Ushio H, Hara M, Okumura K, Ikeda
S, Ogawa H (2014) Host defense (Antimicrobial) peptide, human beta-defensin-3, improves
the function of the epithelial tight-junction barrier in human keratinocytes. J Invest Dermatol
134(8):2163–2173
Kim C, Gajendran N, Mittrucker HW, Weiwad M, Song YH, Hurwitz R, Wilmanns M, Fischer G,
Kaufmann SH (2005) Human {alpha}-defensins neutralize anthrax lethal toxin and protect
against its fatal consequences. Proc Natl Acad Sci U S A 102(13):4830–4835
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 143
Kim C, Slavinskaya Z, Merrill AR, Kaufmann SH (2006) Human alpha-defensins neutralize toxins
of the mono-ADP-ribosyltransferase family. Biochem J 399(2):225–229
Kohlgraf KG, Ackermann A, Lu X, Burnell K, Belanger M, Cavanaugh JE, Xie H, Progulske-Fox
A, Brogden KA (2010) Defensins attenuate cytokine responses yet enhance antibody responses
to Porphyromonas gingivalis adhesins in mice. Future Microbiol 5(1):115–125
Kraus D, Deschner J, Jager A, Wenghoefer M, Bayer S, Jepsen S, Allam JP, Novak N, Meyer R,
Winter J (2012) Human beta-defensins differently affect proliferation, differentiation, and min-
eralization of osteoblast-like MG63 cells. J Cell Physiol 227(3):994–1003
Kurosaka K, Chen Q, Yarovinsky F, Oppenheim JJ, Yang D (2005) Mouse cathelin-related antimi-
crobial peptide chemoattracts leukocytes using formyl peptide receptor-like 1/mouse formyl
peptide receptor-like 2 as the receptor and acts as an immune adjuvant. J Immunol 174(10):
6257–6265
Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, Cao W, Wang YH, Su B,
Nestle FO, Zal T, Mellman I, Schroder JM, Liu YJ, Gilliet M (2007) Plasmacytoid dendritic
cells sense self-DNA coupled with antimicrobial peptide. Nature 449(7162):564–569
Lee SH, Jun HK, Lee HR, Chung CP, Choi BK (2010) Antibacterial and lipopolysaccharide (LPS)-
neutralising activity of human cationic antimicrobial peptides against periodontopathogens. Int
J Antimicrob Agents 35(2):138–145
Li D, Wang W, Shi HS, Fu YJ, Chen X, Chen XC, Liu YT, Kan B, Wang YS (2014) Gene therapy
with beta-defensin 2 induces antitumor immunity and enhances local antitumor effects. Hum
Gene Ther 25(1):63–72
Lillard JW Jr, Boyaka PN, Chertov O, Oppenheim JJ, McGhee JR (1999) Mechanisms for induc-
tion of acquired host immunity by neutrophil peptide defensins. Proc Natl Acad Sci U S A
96(2):651–656
Liu H, Yu H, Gu Y, Xin A, Zhang Y, Diao H, Lin D (2013a) Human beta-defensin DEFB126 is
capable of inhibiting LPS-mediated inflammation. Appl Microbiol Biotechnol 97(8):
3395–3408
Liu J, Du X, Chen J, Hu L, Chen L (2013b) The induction expression of human beta-defensins in
gingival epithelial cells and fibroblasts. Arch Oral Biol 58(10):1415–1421
Lu W, de Leeuw E (2013) Pro-inflammatory and pro-apoptotic properties of Human Defensin 5.
Biochem Biophys Res Commun 436(3):557–562
Ma XT, Xu B, An LL, Dong CY, Lin YM, Shi Y, Wu KF (2006) Vaccine with beta-defensin
2-transduced leukemic cells activates innate and adaptive immunity to elicit potent antileuke-
mia responses. Cancer Res 66(2):1169–1176
Martin SF (2014) Adaptation in the innate immune system and heterologous innate immunity. Cell
Mol Life Sci 71:4115–4130
Miles K, Clarke DJ, Lu W, Sibinska Z, Beaumont PE, Davidson DJ, Barr TA, Campopiano DJ,
Gray M (2009) Dying and necrotic neutrophils are anti-inflammatory secondary to the release
of alpha-defensins. J Immunol 183(3):2122–2132
Molhoek EM, den Hertog AL, de Vries AM, Nazmi K, Veerman EC, Hartgers FC, Yazdanbakhsh
M, Bikker FJ, van der Kleij D (2009) Structure-function relationship of the human antimicro-
bial peptide LL-37 and LL-37 fragments in the modulation of TLR responses. Biol Chem
390(4):295–303
Motzkus D, Schulz-Maronde S, Heitland A, Schulz A, Forssmann WG, Jubner M, Maronde E
(2006) The novel beta-defensin DEFB123 prevents lipopolysaccharide-mediated effects
in vitro and in vivo. FASEB J 20(10):1701–1702
Nakatsuji T, Gallo RL (2012) Antimicrobial peptides: old molecules with new ideas. J Invest
Dermatol 132(3 Pt 2):887–895
Niyonsaba F, Someya A, Hirata M, Ogawa H, Nagaoka I (2001) Evaluation of the effects of pep-
tide antibiotics human beta-defensins- 1/-2 and LL-37 on histamine release and prostaglandin
D(2) production from mast cells. Eur J Immunol 31(4):1066–1075
Niyonsaba F, Ushio H, Nakano N, Ng W, Sayama K, Hashimoto K, Nagaoka I, Okumura K,
Ogawa H (2007) Antimicrobial peptides human beta-defensins stimulate epidermal keratino-
cyte migration, proliferation and production of proinflammatory cytokines and chemokines.
J Invest Dermatol 127(3):594–604
144 K.A. Brogden et al.
Owen SM, Rudolph D, Wang W, Cole AM, Sherman MA, Waring AJ, Lehrer RI, Lal RB (2004a)
A theta-defensin composed exclusively of D-amino acids is active against HIV-1. J Pept Res
63(6):469–476
Owen SM, Rudolph DL, Wang W, Cole AM, Waring AJ, Lal RB, Lehrer RI (2004b) RC-101, a
retrocyclin-1 analogue with enhanced activity against primary HIV type 1 isolates. AIDS Res
Hum Retroviruses 20(11):1157–1165
Petrov V, Funderburg N, Weinberg A, Sieg S (2013) Human beta defensin-3 induces chemokines
from monocytes and macrophages: diminished activity in cells from HIV-infected persons.
Immunology 140(4):413–420
Pingel L, Lu X, Brogden KA (2007) Antimicrobial peptides as mucosal adjuvants. In: Brogden
KA, Stanton TB, Cornick N et al (eds) Virulence mechanisms of bacterial pathogens, 4th edn.
ASM Press, Washington, DC, pp 281–295
Pingel LC, Kohlgraf KG, Hansen CJ, Eastman CG, Dietrich DE, Burnell KK, Srikantha RN, Xiao
X, Belanger M, Progulske-Fox A, Cavanaugh JE, Guthmiller JM, Johnson GK, Joly S, Kurago
ZB, Dawson DV, Brogden KA (2008) Human beta-defensin 3 binds to hemagglutinin B
(rHagB), a non-fimbrial adhesin from Porphyromonas gingivalis, and attenuates a pro-
inflammatory cytokine response. Immunol Cell Biol 86(8):643–649
Prohaszka Z, Nemet K, Csermely P, Hudecz F, Mezo G, Fust G (1997) Defensins purified from
human granulocytes bind C1q and activate the classical complement pathway like the trans-
membrane glycoprotein gp41 of HIV-1. Mol Immunol 34(11):809–816
Ramos R, Silva JP, Rodrigues AC, Costa R, Guardao L, Schmitt F, Soares R, Vilanova M,
Domingues L, Gama M (2011) Wound healing activity of the human antimicrobial peptide
LL37. Peptides 32(7):1469–1476
Rehaume LM, Hancock RE (2008) Neutrophil-derived defensins as modulators of innate immune
function. Crit Rev Immunol 28(3):185–200
Schaal JB, Tran D, Tran P, Osapay G, Trinh K, Roberts KD, Brasky KM, Tongaonkar P, Ouellette
AJ, Selsted ME (2012) Rhesus macaque theta defensins suppress inflammatory cytokines and
enhance survival in mouse models of bacteremic sepsis. PLoS One 7(12):e51337
Scott MG, Rosenberger CM, Gold MR, Finlay BB, Hancock RE (2000) An alpha-helical cationic
antimicrobial peptide selectively modulates macrophage responses to lipopolysaccharide and
directly alters macrophage gene expression. J Immunol 165(6):3358–3365
Scott MG, Davidson DJ, Gold MR, Bowdish D, Hancock RE (2002) The human antimicrobial
peptide LL-37 is a multifunctional modulator of innate immune responses. J Immunol
169(7):3883–3891
Scott A, Weldon S, Buchanan PJ, Schock B, Ernst RK, McAuley DF, Tunney MM, Irwin CR,
Elborn JS, Taggart CC (2011) Evaluation of the ability of LL-37 to neutralise LPS in vitro and
ex vivo. PLoS One 6(10):e26525
Semple F, Dorin JR (2012) beta-defensins: multifunctional modulators of infection, inflammation
and more? J Innate Immun 4(4):337–348
Semple F, Webb S, Li HN, Patel HB, Perretti M, Jackson IJ, Gray M, Davidson DJ, Dorin JR
(2010) Human beta-defensin 3 has immunosuppressive activity in vitro and in vivo. Eur
J Immunol 40(4):1073–1078
Shi J, Aono S, Lu W, Ouellette AJ, Hu X, Ji Y, Wang L, Lenz S, van Ginkel FW, Liles M, Dykstra
C, Morrison EE, Elson CO (2007) A novel role for defensins in intestinal homeostasis: regula-
tion of IL-1beta secretion. J Immunol 179(2):1245–1253
Sinno H, Prakash S (2013) Complements and the wound healing cascade: an updated review. Plast
Surg Int 2013:146764
Skarnes RC, Watson DW (1957) Antimicrobial factors of normal tissues and fluids. Bacteriol Rev
21(4):273–294
Suphasiriroj W, Mikami M, Shimomura H, Sato S (2013) Specificity of antimicrobial peptide
LL-37 to neutralize periodontopathogenic lipopolysaccharide activity in human oral fibro-
blasts. J Periodontol 84(2):256–264
9 Antimicrobial Peptides in Host Defense: Functions Beyond Antimicrobial Activity 145
Tani K, Murphy WJ, Chertov O, Salcedo R, Koh CY, Utsunomiya I, Funakoshi S, Asai O,
Herrmann SH, Wang JM, Kwak LW, Oppenheim JJ (2000) Defensins act as potent adjuvants
that promote cellular and humoral immune responses in mice to a lymphoma idiotype and car-
rier antigens. Int Immunol 12(5):691–700
Territo MC, Ganz T, Selsted ME, Lehrer R (1989) Monocyte-chemotactic activity of defensins
from human neutrophils. J Clin Invest 84(6):2017–2020
Tewary P, de la Rosa G, Sharma N, Rodriguez LG, Tarasov SG, Howard OM, Shirota H, Steinhagen
F, Klinman DM, Yang D, Oppenheim JJ (2013) beta-Defensin 2 and 3 promote the uptake of
self or CpG DNA, enhance IFN-alpha production by human plasmacytoid dendritic cells, and
promote inflammation. J Immunol 191(2):865–874
van den Berg RH, Faber-Krol MC, van Wetering S, Hiemstra PS, Daha MR (1998) Inhibition of
activation of the classical pathway of complement by human neutrophil defensins. Blood
92(10):3898–3903
Van Hemert JR, Recker EN, Dietrich D, Progulske-Fox A, Kurago ZB, Walters KS, Cavanaugh JE,
Brogden KA (2012) Human beta-defensin-3 alters, but does not inhibit, the binding of
Porphyromonas gingivalis haemagglutinin B to the surface of human dendritic cells. Int
J Antimicrob Agents 40(1):75–79
Van Wetering S, MannesseLazeroms SPG, Dijkman JH, Hiemstra PS (1997) Effect of neutrophil
serine proteinases and defensins on lung epithelial cells: modulation of cytotoxicity and IL-8
production. J Leukoc Biol 62(2):217–226
Vemula SV, Amen O, Katz JM, Donis R, Sambhara S, Mittal SK (2013) Beta-defensin 2 enhances
immunogenicity and protection of an adenovirus-based H5N1 influenza vaccine at an early
time. Virus Res 178(2):398–403
Vordenbaumen S, Fischer-Betz R, Timm D, Sander O, Chehab G, Richter J, Bleck E, Schneider M
(2010) Elevated levels of human beta-defensin 2 and human neutrophil peptides in systemic
lupus erythematosus. Lupus 19(14):1648–1653
Walters SM, Dubey VS, Jeffrey NR, Dixon DR (2010) Antibiotic-induced Porphyromonas gingi-
valis LPS release and inhibition of LPS-stimulated cytokines by antimicrobial peptides.
Peptides 31(9):1649–1653
Wang W, Cole AM, Hong T, Waring AJ, Lehrer RI (2003) Retrocyclin, an antiretroviral theta-
defensin, is a lectin. J Immunol 170(9):4708–4716
Wang W, Mulakala C, Ward SC, Jung G, Luong H, Pham D, Waring AJ, Kaznessis Y, Lu W,
Bradley KA, Lehrer RI (2006) Retrocyclins kill bacilli and germinating spores of Bacillus
anthracis and inactivate anthrax lethal toxin. J Biol Chem 281(43):32755–32764
Warnke PH, Voss E, Russo PA, Stephens S, Kleine M, Terheyden H, Liu Q (2013) Antimicrobial
Peptide coating of dental implants: biocompatibility assessment of recombinant human Beta
defensin-2 for human cells. Int J Oral Maxillofac Implants 28(4):982–988
Yamashita T, Saito K (1989) Purification, primary structure, and biological activity of guinea pig
neutrophil cationic peptides. Infect Immun 57(8):2405–2409
Yang D, Oppenheim JJ (2004) Multiple functions of antimicrobial peptides in host immunity. In:
Devine DA, Hancock REW (eds) Mammalian host defense peptides. Cambridge University
Press, Cambridge, pp 39–68
Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, Anderson M, Schroder JM,
Wang JM, Howard OM, Oppenheim JJ (1999) b-defensins: linking innate and adaptive immu-
nity through dendritic and T cell CCR6. Science 286(5439):525–528
Yang D, Chen Q, Chertov O, Oppenheim JJ (2000) Human neutrophil defensins selectively che-
moattract naive T and immature dendritic cells. J Leukoc Biol 68(1):9–14
Yang D, Chertov O, Oppenheim JJ (2001) The role of mammalian antimicrobial peptides and
proteins in awakening of innate host defenses and adaptive immunity. Cell Mol Life Sci
58(7):978–989
Yang D, Biragyn A, Kwak LW, Oppenheim JJ (2002) Mammalian defensins in immunity: more
than just microbicidal. Trends Immunol 23(6):291–296
146 K.A. Brogden et al.
Yang D, Chen Q, Hoover DM, Staley P, Tucker KD, Lubkowski J, Oppenheim JJ (2003) Many
chemokines including CCL20/MIP-3alpha display antimicrobial activity. J Leukoc Biol
74(3):448–455
Yang D, Biragyn A, Hoover DM, Lubkowski J, Oppenheim JJ (2004) Multiple roles of antimicro-
bial defensins, cathelicidins, and eosinophil-derived neurotoxin in host defense. Annu Rev
Immunol 22:181–215
Yeom M, Park J, Lee B, Choi SY, Kim KS, Lee H, Hahm DH (2011) Lactoferrin inhibits the
inflammatory and angiogenic activation of bovine aortic endothelial cells. Inflamm Res
60(5):475–482
Yu H, Dong J, Gu Y, Liu H, Xin A, Shi H, Sun F, Zhang Y, Lin D, Diao H (2013) The novel human
beta-defensin 114 regulates lipopolysaccharide (LPS)-mediated inflammation and protects
sperm from motility loss. J Biol Chem 288(17):12270–12282
Chapter 10
Antimicrobial Peptides: Do They Have
a Future as Therapeutics?
Michael Zasloff
Abstract Antimicrobial peptides of higher organisms have been studied for the
past 25 years and their importance as components of innate immunity is now well
established. The basic simplicity of the chemical structure of antimicrobial peptides
along with the lower likelihood of the emergence of resistance compared with con-
ventional antibiotics have made them attractive candidates for development as ther-
apeutics. In this chapter, I describe the stories behind three drug candidates currently
in clinical trials: Pexiganan, Plectasin, and Brilacidin. Each of these compounds has
faced specific challenges in development and has a high likelihood of reaching com-
mercialization. Antimicrobial peptides appear to be coming of age as therapeutics.
10.1 Introduction
I have been asked to address the question as to whether antimicrobial peptides have
a future as therapeutics. Antimicrobial peptides of multicellular organisms were
characterized in the 1980s from insects, mammals, and frogs (Zasloff 2002). As yet,
antimicrobial agents of this class have not yet been approved as drugs. It is reason-
able to ask what the problems have been that have interfered with their commercial-
ization. In this piece I will focus on three examples of antimicrobial peptides that
have advanced deeply into development but faced specific challenges that inter-
rupted their progress: Pexiganan developed by Magainin Pharmaceuticals; plecta-
sin, developed by Novozymes; and brilacidin, developed by Polymedix. Although
there are other compounds that could be included, I have selected these because I
am personally most familiar with these three, and I believe the lessons learned apply
broadly to all compounds of this class destined for therapeutic development.
Fig. 10.1 The amino acid sequences of pexiganan, magainin 1 and magainin 2, are compared.
Each peptide has an amide at the carboxyl terminus. Lysine residues are highlighted in gray
10.2 Pexiganan
intolerable, due to side effects such a diarrhea, and frequently stop taking them despite
being fully aware of the dire consequences of progressive infection.
Pexiganan was developed as a locally applied alternative to a systemically
administered broad-spectrum antibiotic. The antibiotic spectrum was broad enough
to cover the range of microbes known to be present on the diabetic ulcer. Furthermore,
as a topical agent, high local concentrations in the superficial soft tissues could be
achieved easily, surpassing the concentrations that could be reached systemically.
We chose as our intended diabetic patients those with the most superficial of infected
ulcers, excluding those cases where deep penetration of a topically applied ointment
could not occur, including those with soft tissue infections in close proximity to
bone. We argued that high concentrations of pexiganan would also reduce the likeli-
hood of selection of resistant organisms and thus could be used repeatedly over the
course of a patient’s lifetime.
The choice of developing pexiganan as a topical agent was also influenced by sev-
eral practical and strategic considerations. The primary structure of the peptide makes
it susceptible to proteolytic cleavage, especially by trypsin-like enzymes. As a conse-
quence, the intact peptide does not pass from the soft tissue into the systemic circula-
tion. This property permitted us to drastically reduce very costly preclinical toxicology
prior to clinical evaluation, since chronic systemic exposure to an intact peptide would
not occur following long-term local application to an open wound. In addition, a
therapeutic course of local administration would require relatively small amounts of
peptide compared with systemic administration. Based on the minimal inhibitory
concentrations of pexiganan, one could estimate that intravenous dosing would
require about 1 g daily, not very different from the dosing of most antibiotics currently
in use. At the time we began the development process, the chemical synthesis of pexi-
ganan cost about $1000 per gram, using our proprietary solution-phase process. Only
if this peptide were dramatically more effective than the less expensive agents in use
would it make sense to develop it for systemic use, and we did not believe that this
was the case. As a topical agent, however, much lesser amounts of peptide could be
applied directly to a site of infection, and the cost of goods relative to the anticipated
price of the commercial product made sense. Eventually, we were able to bring the
cost of production of pharmaceutical grade pexiganan down to about $100 per gram.
We conducted two extensive pivotal phase III clinical trials in the late 1990s
involving about 1000 subjects (Lipsky et al. 2008). Topically applied pexiganan was
evaluated against orally administered ofloxacin in what is a called an “inferiority”
study. The hypothesis being tested was whether topically applied pexiganan exhib-
ited was less effective than orally administered ofloxacin, an antibiotic approved for
deep soft tissue infections (no specific agent had been approved for diabetic foot
infections, so the FDA permitted us to use this antibiotic as the comparator). In both
treatment arms, the wounds were debrided of dead tissue as part of the study. Ideally,
it would have been scientifically more exciting to have conducted a “superiority”
trial, where pexiganan treatment was compared to simple wound debridement (a
placebo-controlled trial), but all parties, including the FDA and participating physi-
cians and patients, felt this to be unethical.
Pexiganan proved itself to be as effective as oral ofloxacin in the treatment of
infected diabetic foot ulcers over about a month of treatment. Of particular interest
150 M. Zasloff
was that pexiganan-resistant bacteria did not appear on the treated wounds. In con-
trast, the microbes cultured from the wounds of those subjects treated with ofloxacin
shifted to a more resistant concentration range following the month of therapy. The
rates and extent of wound healing were comparable as were the impact on the clini-
cal appearance of the treated ulcers. As anticipated, those receiving topical treat-
ment reported fewer side effects than those taking the oral therapeutics.
Despite the positive outcome, the FDA Advisory Panel voted 7–5 against
approval of pexiganan. Their primary concern was the uncertainty over the “placebo”
outcome. Suppose these individuals had not been treated with any antibiotic, just
surgical curettage, what would the outcome have been? Despite being presented
with the historical data then available, the panel was not convinced. Indeed, Dr.
Julie Parsonnet of Stanford University suggested to the panel that all a doctor had to
do to treat an infected ulcer was “to cut out the infected tissue and drop it into a trash
can and forget the antibiotics!” (She herself had never treated such a patient, nor had
most members of that FDA panel). Several panelists could not believe that it was
possible for a topical agent to be as effective as we had claimed. In response to the
Advisory Panel, the FDA requested that we repeat a trial using a placebo arm. We
did not believe this to be practical, and the pexiganan program was halted.
Over the past few years, pexiganan’s development as a topical agent for the treat-
ment of infected diabetic foot ulcers has been taken up by Dipexium Pharmaceuticals,
Inc. Working closely with the FDA and many of the American experts in the treat-
ment of infected diabetic foot ulcers, the company has designed a pivotal phase III
placebo-controlled trial which should both fulfill the requirements of a superiority
study and also satisfy any concerns regarding the placing of untreated patients at
risk. Within the next few years, we will learn of this study’s outcome.
10.3 Plectasin
The discovery and characterization of plectasin was first reported in 2005 (Mygind
et al. 2005) (Fig. 10.2). A team of scientists at Novozymes, headed by Dr. Hans
Henrik Kristensen, had exploited a novel method for the discovery of antimicrobial
peptides from natural sources. The team had created a cDNA library from a wild
mushroom like fungus (Pseudoplectania nigrella). By reengineering the library, the
team searched for the expressed gene products that would be secreted (i.e., that con-
tained “signal” sequences). Of the cDNAs identified, one exhibited the primary
sequence of a defensin antimicrobial peptide. Using several fungal- and yeast-based
expression systems, the Novozymes team biosynthetically produced the peptide in
sufficient amounts to begin characterization. The defensin, named “plectasin,” was
found to have a primary, secondary, and tertiary structure very similar (surprisingly!)
to defensins from insects and mussels. Of particular interest was its antibacterial
spectrum. Of the bacteria studied, plectasin exhibited its greatest potency against
Streptococcus pneumonia (Mygind et al. 2005). In vivo, plectasin demonstrated
potency when administered systemically that equaled or exceeded conventional anti-
biotics such as penicillin. In addition, the antibiotic, following parenteral
10 Antimicrobial Peptides: Do They Have a Future as Therapeutics? 151
administration to the mouse, could be recovered intact from its urine. Plectasin was
essentially nontoxic in early preclinical experiments. Its mechanism of action was
established and shown to involve interaction with lipid II (Schneider et al. 2010).
Almost all of the clinical isolates of Pneumococcus studied exhibited a minimal
inhibitory concentration at sub-micromolar values, suggesting that most infections
would be effectively treated initially with this antibiotic. Furthermore, because plec-
tasin was of fungal origin, it could be expressed robustly in several recombinant
vectors used for the industrial production of commodity substances, such as enzymes
used in the food industry. Hence, it could potentially be produced inexpensively and
in large amounts.
From one perspective plectasin was an ideal antibiotic. Here was a narrow-
spectrum antibiotic that a physician could call upon to treat an infection caused by
Pneumococcus. In a setting where a specific bacterial organism is involved, a spe-
cific antibiotic should be used. Since many strains of Pneumococcus have devel-
oped resistance to beta-lactams, and plectasin retained activity against
penicillin-resistant Pneumococcus, plectasin could become the antibiotic of choice
for the treatment of pneumococcal infections.
Unfortunately, the economics of drug development intervened. The market for a
pneumococcal-specific antibiotic was too small to justify the expensive investment
required to bring the antibiotic through development into clinical use. The
Novozymes team began a long and highly creative series of structure-activity stud-
ies to extend the spectrum of plectasin so as to include human pathogens such as
Staphylococcus aureus. The Kristensen team succeeded creating the plectasin
derivative NZ2114 which was effective against systemic S. aureus infections in sev-
eral animal models (Andes et al. 2009; Xiong et al. 2011). Similar to plectasin,
NZ2114 could be produced at commercial viable expression levels through recom-
binant expression in a yeast system (Zhang et al. 2014).
In late 2009 Sanofi-Aventis entered a partnership with Novozymes to lead the
clinical development of NZ2114 and in early 2010 announced that planning for a
phase 1 clinical program had begun. Very little public information has been dis-
closed since then.
152 M. Zasloff
10.4 Brilacidin
Linear amphipathic alpha helical cationic antimicrobial peptides target the external
membranes of microbes. In some cases they damage the permeability of the mem-
brane; in others they translocate into to the cytoplasm and cause functional distur-
bance. Molecules that adopt comparable secondary structures can exhibit antimicrobial
activity, and many synthetic mimetics of the naturally occurring linear peptides have
been synthesized. In 2002 DeGrado reported the synthesis and characterization of a
novel family of arylamide amphipathic anionic polymers that exhibited many of the
physical and biological properties of antimicrobial peptides, such as magainin (Tew
et al. 2002). These mimetics could be synthesized from inexpensive monomers via
classical polymerization methods and were resistant to proteolytic hydrolysis. Through
structure-activity refinements, potency against Gram-positive bacteria was optimized
while maintaining minimal lytic activity against human erythrocytes, resulting in the
creation of brilacidin (Fig. 10.3) (Choi et al. 2009). This molecule adopts a planar
secondary structure, with guanidinyl and pyridinyl groups positioned on one edge of
the scaffold and trifluoromethane groups on another. As reported recently, brilacidin
acts by selectively damaging the microbial membrane, similar to linear antimicrobial
peptides and the lipopeptide in commercial use, daptomycin (Mensa et al. 2014).
Brilacidin was initially licensed to Polymedix, Inc., and subsequently to
Cellceutix, Inc. It has been advanced in development as a parenteral drug for the
treatment of acute skin and skin structure infections caused by Gram-positive bac-
teria, positioned to compete with drugs such as daptomycin.
Brilacidin advanced successfully through phase 1 and phase 2 clinical trials. On
October 23, 2014, Cellceutix reported unpublished results of a phase 2b randomized
double-blind study comparing brilacidin to daptomycin for the treatment of Gram-
positive acute skin and skin structure infections (http://cellceutix.com/cellceutix-
releases-confidence-interval-statistics-showing-clinical-success-rates-for-brilacidin-
in-treatment-of-absssi/#sthash.UpDWgko4.dpbs). Two-hundred and fifteen subjects
were enrolled. The primary end point was clinical success defined as reduction of at
least 20 % in area of the infected site when evaluated 48–72 h after the first dose of the
NH HN
NH H O H N N H O H NH
( ) N N N N ( )
H2N N 4 4 N NH2
H O O O O H
F F F F
F F
Brilacidin
study drug. As reported by Cellceutix: “In treated patients assessed at 48–72 h, 47/51
(92.2 %), 46/48 (95.8 %), 51/52 (98.1 %), and 45/48 (93.8 %) achieved clinical suc-
cess in the brilacidin 0.6 mg/kg single-dose group, brilacidin 0.8 mg/kg single-dose
group, brilacidin 1.2 mg/kg 3-day group, and daptomycin 7-day group, respectively.
All three brilacidin treatment arms (two single-dose regimens and one 3-day dose
regimen) reached the primary endpoint, with the clinical success rate for each dosing
regimen statistically comparable to the clinical success rate of the FDA-approved
7-day dosing regimen of daptomycin. All brilacidin treatment regimens were well
tolerated. There were six severe adverse events (SAE) reported across the study, none
of which were considered related to brilacidin by the principal investigator.” Brilacidin
will be advanced into a phase 3 clinical trial.
10.5 Conclusions
The three compounds that I have discussed are advancing through the therapeutic
development process. Unless they face clinical failure, due either to a lack of effi-
cacy or an unacceptable toxicity, they will represent the first “graduating class” of
antimicrobial peptides.
Each of the three highlights several attractive characteristics of antimicrobial
peptides:
1. Antimicrobial resistance does not readily occur.
2. The mechanism of action can be reproduced with molecules as structurally dis-
tant from a linear peptide as brilacidin.
3. Certain naturally occurring peptides from fungal sources with very narrow anti-
biotic spectra, such as plectasin, could be developed for the treatment of specific
bacterial infections. Many uncharacterized fungal defensins have already been
identified (Zhu 2008).
4. Naturally occurring antimicrobial peptides, such as plectasin, exhibit acceptable
therapeutic indices and can serve as the models upon which the structural prin-
ciples that govern bio-distribution of therapeutically effective antimicrobial pep-
tides can be deciphered.
5. Antimicrobial peptides and mimetics can be commercially synthesized
cost-effectively.
6. As yet, no class specific toxicity has become apparent.
Although it seems like an eternity that we have been awaiting the introduction of
antimicrobial peptides into the clinic, we are now on the threshold of seeing this
happen. We are now facing a medical crisis some have called the “End of the
Antibiotic Era.” Resistance of both Gram-positive and Gram-negative human patho-
gens to our conventional antibiotics has spread throughout the world. Infections that
could be treated routinely several years ago now present as life-threatening medical
challenges. The introduction of antimicrobial peptides as therapeutic drugs could
not be happening at a more critical moment in time than now.
154 M. Zasloff
References