2023 - Review Article - Swati - IJVP - Sept

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Indian J. Vet. Pathol., 47(3) : 189-200, 2023: DOI: 10.5958/0973-970X.2023.00018.

Mechanisms of cell death: Current perspectives


REVIEW ARTICLE

S.S. Patel1, B.J. Trangadia1, J.B. Kathiriya2 and D.T. Fefar1


1
Department of Veterinary Pathology, 2Department of Veterinary Public Health and Epidemiology, College of Veterinary
Science and Animal Husbandry, Kamdhenu University, Junagadh-362 001, Gujarat
Address for Correspondence
B.J. Trangadia, Associate Professor, Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry,
Kamdhenu University, Junagadh-362 001, Gujarat, E-mail: [email protected]

Received: 1.3.2023; Accepted: 20.8.2023

ABSTRACT
Cell death is vital in embryonic development, tissue molding during embryogenesis, immune system development and
removal of damaged cells. However, subsequent release of various inflammatory cytokines intracellularly leads to inflammatory
changes during necrosis, pyroptosis and necroptosis. Necroptosis is regulated cell death with mimicking feature of apoptosis and
necrosis. In NETosis and ferroptosis, reactive oxygen species leads to the oxidative cell death. In pyroptosis, caspase-1 enzyme
release pro-inflammatory molecules which lead to inflammatory changes.
Keywords: Apoptosis, autophagy, ferroptosis, necroptosis, necrosis, NETosis, pyroptosis

INTRODUCTION How to cite this article : Patel, S.S., Tran-


Cell death, proliferation, differentiation and survival are fundamental gadia, B.J., Kathiriya, J.B. and Fefar, D.T. 2023.
biological processes. It is vital during embryonic development as it maintains Mechanisms of cell death: Current perspe-
equilibrium of organisms and eliminates damaged cells1. It also important ctives. Indian J. Vet. Pathol., 47(3) : 189-200.
for tissue molding and immune system development2. Cells have restricted
responses to injury viz., adaptation, degeneration and death; depending on type
the surrounding tissue can cause
of the cell and severity of the injury. Cells may react positively to a sub-lethal
tissue damage6. Cell injury can
injury and increase their efficiency or productivity or they may degenerate
vary from internal abnormalities
and lose their functional potential. The injury reaction might be reversible and
to injury by external factors. The
can eventually restorecellular structure and function or irreversible with cell
most common causes of necrosis
degeneration leading to death3.
include hypoxia, physical agents,
Nomenclature Commiee on Cell Death (NCCD) recommended definition chemical agents, biological agents
and interpretation of cell death from morphological, biochemical and functional and immunologic reactions. Loss
viewpoints4. During last few years, a series of highly regulated molecular of cell membrane integrity due
pathways have been discovered, which determine whether a cell will survive to exposure of noxious stimulus,
or succumb to injury2. Cell death can be classified as programmed or non- allows extracellular ions and fluid
programmed depending on their signals (Fig. 1)1. to move inside the cell, resulting in
swelling of cell and its organelles.
Programmed cell death (PCD) can be triggered by tightly controlled
Further, disruption of lysosomal
intracellular signal transduction pathways. Based on morphological features and
membrane allows release of
molecular mechanisms, PCD can be classified into two types, viz., apoptotic cell
proteolytic enzymes such as
death and non-apoptotic cell death. Apoptosis maintains cell membrane integrity
proteases, RNAases, DNAases
and is caspase-dependent (apoptosis and anoikis), whereas non-apoptotic cell
and phosphatases 7 and cause
death is caspase independent and characterized primarily by membrane rupture
DNA, RNA and protein damage;
(necroptosis, NETosis, ferroptosis, pyroptosis etc.)1.
and also cause cell destruction by
During non-programmed cell death, cells undergo physicochemical or digesting cellular components
mechanical stress within organisms and a sudden and uncontrollable structural andrupture of plasma membrane,
breakdown resulting in accidental cell death (ACD) or non-programmed cell allowing intracellular contents to
death (necrosis)5. spill into the surrounding tissue6.
Mechanism of necrosis
NECROSIS As a result of hypoxia, oxygen
It is a local cell death characterized by swelling of the cell organelles, rupture tension decreases intracellular,
of plasma membrane and cell lysis. Spillage of contents within the cells into which diminishes the process
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
190 Patel et al.

Fig. 1. Classification of cell death.

of oxidative phosphorylation and ATP generation. The lose their structural integrity and turn into a viscous mass.
depletion of ATP leads to decreased activity of Na+- K+ This morphology also occurs in most bacterial infections
pump, which is essential for maintaining osmolarity of the and the accumulation of necrotic material is called pus.
cell. This allow accumulation of sodium ions and water
Caseous necrosis: Caseous means "cheese-like" and it
inside the cell and diffusion of potassium outside the cell,
refers to the white appearance of the necrotic area. This
producing acute cellular swelling. There is decrease in
type of necrosis occurs in tuberculosis and the necrotic
cellular ATP, increase in adenosine monophosphorylase
area is called as granuloma11.
and increase rate of anaerobic glycolysis to maintain the
energy supply to cell. Glycolysis leads to accumulation of Fat necrosis: Injury to pancreas during acute
lactic acidand inorganic phosphate from the hydrolysis of pancreatitis causes release of pancreatic enzymes and
phosphate esters results in decrease pHof the cell, which subsequent liquefaction of peritoneal fat. These liquified
causes early clumping of nuclear chromatin. fat cells canmixed with calcium toformchalky white areas
referred as saponification12.
Decrease in pH along with low ATP level cause
ribosomal detachment from the endoplasmic reticulum
and dissociation of polysomes into monosomes, with APOPTOSIS
results in reduced protein synthesis. If hypoxia continues, For the first time, the term “apoptosis” introduced by
diminishing mitochondrial function and increased Kerr, Wyllie, and Currie in the year 1972, to characterize a
membrane permeability, surface blabs and myelin figures morphologically distinct form of cell death13. It is a normal
produced.However, all these changes are reversible process that occurs during development and ageing as a
if oxygen is restored but if ischemia persists, necrosis homeostatic mechanism to maintain cell populations in
(irreversible injury) occur8. tissues; and also in immunological responses as a defence
mechanism or during cell injury by disease or toxic
Necrosis can be classified into four types:
chemicals14. It is a coordinated, energy-dependent process
Coagulative necrosis: Ischemia is the primary cause that involves the activation of a number of caspases and a
of coagulative necrosis in most of the organs except brain. complicated chain of events that connects the beginning
Cells appear anucleated, eosinophillic with preserved of stimuli to the eventual demise of the cell15.
architectural details. Eventually, the dead cells are
Inflammatory response does not associated with
removed by phagocytosis9.
apoptosis or the removal of apoptotic cells because 1.
Liquefactive/colliquative necrosis: It is most common apoptotic cells do not release their cellular components
type of necrosis observed in central nervous system10. The into the surrounding tissue; 2. they are rapidly
dying cells are digested by hydrolytic enzymes and hence phagocytosed by surrounding cells, preventing secondary
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
Mechanisms of cell death 191

necrosis and 3. engulfing cells do not produce anti- executioners. Following the cleavage of an initiator
inflammatory cytokines16. caspase 8 to generate its active form, the enzymatic
Apoptosis is a morphological process that begins death programme is initiated by the rapid and sequential
with the condensation of chromatin in the nucleus, activation of other caspases. Execution caspases have a
followed by the fragmentation of the nuclear envelope wide range of cellular effects. They cleave cytoskeletal and
and DNA hydrolysis by DNAses 17. Apoptotic cells nuclear matrix proteins causing cytoskeleton disruption
lose their attachment with nearby cells18, and blebs and nucleus breakdown. Caspase activation targets
(membrane-bound vesicles) develop in plasma proteins involved in transcription, DNA replication
membrane accompanied by cell shrinkage17. Finally, and DNA repair in the nucleus. Caspase-3 activation in
the cell components are gathered into membrane- particular converts a cytoplasmic DNAase into an active
bound structures known as apoptotic bodies, which are form by cleaving an inhibitor of the enzyme; this DNAase
endocytosed by macrophages and/or surrounding cells19. induces the typical internucleosomal cleavage of DNA3.
Cell become shrunken and apoptotic bodies are formed
Mechanism of apoptosis which are phagocytized by macrophages.
Caspases, which act as both initiators and executioners
are critical in this process20. Although there are three Perforin/granzyme apoptosis pathway
apoptosis pathways, the extrinsic (death receptor) and This pathway is the primary signaling pathway used
intrinsic (mitochondrial) pathways are interconnected by cytotoxic lymphocytes to eliminate virus-infected and/
and can affect each other21. or transformed cells. In the calcium-dependent granule
exocytosis pathway, lymphocytes secrete perforin,
Extrinsic (death receptor) pathway
a membrane permeability protein and granzymes
This pathway begins when a death ligand binds to
(proteolytic enzymes) from cytotoxic granules towards
a death receptor. There are numerous death receptors;
the target cell. Perforin in the presence of calcium,
however, tumor necrosis factor receptor-1 (TNFR1) and
polymerizes and initiates changes in the target-cell
first apoptosis signal (Fas also known as Apo-1 or CD95
membrane to allow entry of granzymes into the cell.
are the most well-known receptors. These receptors
Granzymes are neutral serine proteases that can activate
have a death domain that require adaptor proteins viz;
caspases in the target cell. In addition, granzyme B might
Fas associated death domain (FADD) and TNF receptor
directly cleave the BCL2 family member BID to activate
associated death domain (TRADD)22. FADD binds to
the mitochondrial death pathway. During calcium
death receptors which then bind an inactive form of
independentCD95L pathway, lymphocyte exhibits the
caspase-8 via a death domain. As a result, multiple
procaspase-8 molecules are brought into close proximity death ligand CD95L on the cell surface and triggers
and cleave one another to produce active caspase-8. apoptosis through the CD95 receptor on the target cell21.
The active enzymes mediate the execution phase of
apoptosis3. AUTOPHAGY
Intrinsic (mitochondrial) Pathway Autophagy means self-death. It specifically refers to
As name implies, this pathway is activated by the formation of autophagosomes by membrane wrapping
internal inducers such as hypoxia and extremely high on part of the cytoplasm organelles and proteins that need
cytosolic Ca+2 concentration23. This pathway is activated to be degraded in the cells. Autophagosomes fused with
due to increasing mitochondrial permeability and lysosomes to form autophagolysosome and the contents
cytochrome C release into the cytosol24. The pathway is degraded by lysosomal enzymes. Autophagy activated
regulated by two groups of Bcl-2 proteins. First is anti- under stress conditions viz., nutritional starvation or
apoptotic proteins such as Bcl-2, Bcl-XL, Bcl-W, Bfl-1 and energy exhaustion25.
Mcl-1. These proteins regulate apoptosis by blocking the Autophagy is stress adaptation process activated and
release of cytochrome C from mitochondria22. Another caused by autophagy-related gene (ATG)26. Autophagy
group is pro-apoptotic proteins viz; Bak, Bax, Bid, Bcl-Xs, belongs to catabolism which is a highly conserved process
Bad, Bim, Bik and Hrk regulate apoptosis by promoting of energy dependent pathophysiological adaptation27.
the release of cytochrome C. The balance between the
actions of anti and pro-apoptotic proteins determine Classification of autophagy
the direction of apoptosis. The release of cytochrome C Macroautophagy: Formed by endoplasmic reticulum-
from the mitochondria via the intrinsic pathway results derived membrane wrapping around the degraded
in the formation of an apoptosome and the activation material, which fuse with lysosomes to degrade its
of caspases 8 and 9 which initiate the execution phase23. contents28.

Apoptosis Execution Phase Microautophagy: Direct phagocytosis by lysosome


Caspases such as caspase-3 and caspase-6 act as to degrade long-lived proteins in cells29.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023


192 Patel et al.

Historical Perspectives of Autophagy25


Novikoff and Colleagues, 1956 Discovered the location of lysosomal enrichment in rat liver using electron microscope. The
term “dense body” was given to cytoplasmic granules.
Christian de Duve, 1963 Proposed that the process of intracellular inclusion of organelles and part of cytoplasm into
lysosomes is called autophagy at the International Conference on lysosomes.
Edelman, 1974 Large number of concentrated organelle yeast vacuoles (trash pool) on the top of the centrifuge
tube when separating the nucleus of yeast cells at Rockefeller University in the United States.
Per Oar Seglen, 1982 Inhibiting the phosphatidylinositol 3 phosphate (PIP3), kinase inhibitor 3-methyladenine (3-
MA) inhibits autophagy and 3-MA has established itself as the most widely utilized autophagy
inhibitor.
Osumi Yoshinori, 1997 Isolated the first autophagy gene in yeast, apg1 (later agreed to be called atg1).
Matsuura and Colleagues, 1997 First time shown that the autophagy in yeast degrades cytoplasmic components in the presence
of nutritional shortage and demonstrated that the autophagy is essential for the survival of
yeast.
Osumi Yoshinori, October 3, 2016 Won Nobel prize in Physiology and Medicine for “discovering the mechanisms for autophagy”.

Chaperone-mediated autophagy: The process by proteins are required for the elongation, expansion and
which intracellular proteins (soluble) bind to molecular closure of autophagosome membranes. Autophagosomes
chaperones and are transported to lysosomes and then fuse with late endosomes or lysosomes to gain lytic
digested by lysosomal enzymes30. capacity. Fusion in mammalian cells requires the
lysosomal integral membrane protein (LAMP-2).
Mechanism of autophagy
Following autophagosome fusion, materials contained
There are four stages of autophagic pathway as
in the inner membrane are degraded by the action of
shown in Fig. 233. The majority of autophagy-inducing
lysosomal hydrolases. Building blocks (amino acids, fay
signals merge on the mammalian target of rapamycin
acids, etc.) are then transported back to the cytosol for
(mTOR) protein complexes (mTORC1 and mTORC2),
reuse in the various metabolic processes of cells33.
which coordinate anabolic and catabolic processes31.
AMP-Activated protein kinase (AMPK), a cellular energy
sensor, directly regulates mTOR and thus contributes NECROPTOSIS
to the regulation of autophagic activity. Under normal Necroptosis is specific cell death with mixed
conditions, mTORC1 inhibits autophagy by inactivating characteristic of both apoptosis and necrosis. Necroptosis
the autophagy activating kinase (ULK1/2) autophagy is a type of cell death that is morphologically comparable
complex. ULK1 and ATG13 phosphorylation by mTORC1 to necrosis (early breakdown of membrane integrity, cell
result in the inactivation of ULK1/2 complex and down and intracellular organelle enlargement), but it can be
regulation of autophagy32. Stress inhibits mTORC1 and regulated in a caspase-independent manner4, regulated
dephosphorylates the ULK1/2 complex. ULK1/2 then by activation of receptor-interacting protein 1 (RIP1),
phosphorylates Atg13 and FIP200 activate autophagy. RIP3, and mixed lineage kinase domain-like (MLKL)34.
The membrane nucleation stage and initial phagophore It can be activated by ligands for death receptors, such
formation are controlled by a class III phosphatidylinositol as TNF-α35.
3-kinase (PI3K) complex that includes the lipid kinase Instead of cell shrinkage that occurs during apoptosis,
VPS34 and the regulatory protein Beclin-1. cells undergo rapid swelling with simultaneous swelling
Elongation of the isolation membrane depends of organelles during necroptosis. However, instead
on two ubiquitin-like conjugation systems. In the first of blebbing of the plasma membrane, necroptotic
system, autophagy-related gene 12 (ATG12) is covalently cells undergo early plasma membrane breakage,
conjugated to the ATG5 protein through the action of phosphatidylserine exposure and organelle breakdown,
ATG7 (E1-like) and ATG10 (E2-like) proteins. Then, leads to leakage of intracellular content, resulting in
recruitment of the ATG16L1 protein to ATG12-5 dimer inflammation34. It involved in a variety of pathological
results in the formation of a larger complex. Then forming processes such as neurodegeneration, inflammatory
ATG12-5-16L1 oligomers serve as E3 ligases that conjugate disorders, malignancies and renal injury. Necroptosis
lipid molecules (such as phosphatidylethanolamine) to induced by various stimuli, such as TNF-α, interferon
ATG8 orthologs microtubule associated protein 1, light γ, lipopolysaccharide (LPS), pathogen and damage-
chain 3 (MAP1LC3), golgi associated ATPase enhancer associated molecular paerns (DAMP)36.
of 16kDa (GATE16), gamma aminobutyric acid receptors Necroptosis ensures that cells are not fully inert
associated protein (GABARAP). Lipid-conjugated ATG8 in the face of external injury, reducing cell damage by
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
Mechanisms of cell death 193

Fig. 2. Stages of the autophagy pathway. A. Upstream signaling, B. Membrane nucleation stage, C. Elongation and closure stage, D.
Autophagosome-lysosome fusion stage.

controlling a series of signals38. Necroptosis induces Necroptosis can be regulated via genetics,
local inflammation as a result of the release of cell pharmacology and other factors, however, caspase
inclusions as well as the infiltration and activation of inactivation can convert apoptosis into complete necrosis
inflammatory cells39. Through the release of DAMPs, or a specific cell death with mixed characteristics of
necrosis promotes inflammation. DAMPs are a poorly apoptosis and necrosis35. A small specialized molecular
defined class of molecules that are normally restricted to compound called as necroptosis specific inhibitor-1 (Nec-
viable cells. DAMPs (alarmins) promote the activation of 1) can prevent necroptosis37.
macrophages, dendritic cells, and other immune system Mechanism of necroptosis
sentinel cells to initiate inflammation when released into Tumor necrosis factor (TNF) signaling or other
the extracellular space as a result of necrosis. The major damaging stimuli that acts on homologous cell death
DAMPs may be members of the extended IL-1 family40. receptors, can causes mitochondrial alterations, leads
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
194 Patel et al.

to different forms of cell death41. Permeability of the protein 2, IκB kinase (IKK) and nuclear factor-κB essential
mitochondrial outer membrane is altered, resulting modulator (NEMO) regulatory subunit45. When IKKα
in the release of proteins from the mitochondrial and IKKβ are activated, IκB can be phosphorylated,
inter-membranous region, which activate apoptosis resulting in nuclear factor-κB (NF-κB) release and
via mitochondrial pathways37. Caspase independent mitogen-activated protein kinase (MAPK) activation. Cell
apoptosis begins with the release of apoptosis-inducing survival is promoted as a result of this NF-κB signalling
factor and endonuclease G. Changes in mitochondrial pathway37.
inner membrane permeability cause a rapid drop in
RIP1 polyubiquitylation not only influences NF-κB
mitochondrial membrane potential, disrupting electron
activation, but also affects the transition from complex I
transport in the respiratory chain42. As a result, adenosine
to II. Deubiquitylated RIP1 cooperates with its cognate
triphosphate (ATP) production and cell membranes
kinase RIP3 for recruitment to complex II, which
are destroyed resulting in necrosis-like cell death43.
includes TRADD, Fas-associated protein with death
Apoptosis is caused by damage to the mitochondrial
domain (FADD) and caspase-8. In this complex, RIP1
outer membrane, while necroptosis is caused by damage
and RIP3 are inhibited by caspase-8 and proteolytic
to the mitochondrial inner membrane (Fig. 3)37.
cleavage is induced; therefore, the pro-apoptotic caspase
There are several mechanisms of necroptosis activation cascade starts37. By contrast, when caspase-8
including the tumor necrosis factor alpha-TNF receptor1 is inactivated, complex II does not initiate the apoptotic
(TNFα-TNFR1)-related signaling pathway, TNF- program and the TNFα-mediated necroptotic pathway is
related apoptosis- inducing ligand (TRAIL) and factors used46. Caspase-8 and the interaction between RIP1 and
associated with the apoptosis ligand signaling pathway, RIP3 expression could be recognized as the diverging
the RIP3-mitochondrial-reactive oxygen species (mtROS) point of apoptosis and necroptosis47. Nec-1 could halt
metabolic pathway and the benzyloxycarbonyl- the necroptosis signaling pathways by inhibiting RIPI
Val-Ala-Asp, (zVAD) mediated protein kinase C and RIP3 kinase activity39,35.
(PKC)-mitogen-activated protein kinase (MAPK)-AP-1-
related signaling pathway44. The formation of FADD RIP1 RIP3 NEMO complex
has an essential role on TNFα-mediated necroptotic
When ligands bind to these receptors their trimer pathways 48 . Mixed lineage kinase domain-like
conformation changes, allowing their cytosolic tails to pseudokinase (MLKL) is indeed required for TNFα-
recruit multiple proteins and a complex I forms near the induced necroptosis, as key residues of MLKL mutants
cell membrane, which can include TNFR1-associated cannot be phosphorylated, which prevents the activation
death domain (TRADD), receptor-interacting protein of the necrosome49.
(RIP1) kinase, TNFR associated factor 2 (TRAF2), TRAF5,
cellular inhibitor of apoptosis 1 (cIAP1) and cIAP237.
NETOSIS
Complex I was stabilised by ubiquitylation of RIP1 by
cIAP1 and cIAP2, which was subsequently coupled with NETosis is the formation of neutrophil extracellular
transforming growth factor-activated kinase 1-binding traps (NETs) which consist of modified chromatin

Fig. 3. TNF-α TNFR1 related signaling pathway of cell survival, apoptosis and necroptosis.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023


Mechanisms of cell death 195

decorated with bactericidal proteins from granules and Reactive oxygen species produced by NADPH
cytoplasm50. Neutrophils produce granule proteins and oxidase and mitochondria plays a role in the induction
chromatin when they are activated which combine to of NETosis50. The main mechanism of NADPH oxidase
form extracellular fibers that bind Gram-positive and activation is associated with phosphorylation of its
Gram-negative bacteria. These NETs destroy bacteria by subunits by PKC which leads to the assembly of the active
degrading virulence factors51. Neutrophils have a lot of enzyme on the membrane56. PKC stimulates expression of
antibacterial "weaponry" in their granules, which allows myeloid cell leukemia-1 (Mcl-1), the main anti-apoptotic
them to phagocytose infections and destroy them50. protein of neutrophils57. The peptide chemoaractant
When antimicrobial granules of neutrophils fuse with N-formylmethionyl-leucyl-phenylalanine (fMLP)
the phagosome they ingest and kill germs. activated NADPH oxidase, resulting in oxidative
burst and degranulation50. Which binding to a specific
NET formation can be triggered by a variety
receptor activates phospholipase C (PLC) to stimulate
of pathogens including bacteria, fungi, protozoa,
the formation of diacylglycerol (DAG) and inositol
viruses and bacterial cell wall components such as
triphosphate (IP3) from the phosphatidylinositol 4, 5
lipopolysaccharides. NETosis can be induced by
bisphosphate. The activation of NADPH oxidase in this
antibodies and immune complexes, cytokines and
case depends on mitochondrial ROS (mtROS) and on the
chemokines (Interleukin8-IL8, TNF), microcrystals and
opening of mitochondrial permeability transition pore
other physiological stimuli50. NETosis is reactive oxygen
(mPTP). PKC can be activated with the phorbol ester
species (ROS) dependent and different from apoptosis
(PMA), which induces NADPH oxidase and NETosis
because it is caspase independentand does not induce
independently of mtROS and the mPTP. mtROS in the
DNA fragmentation. It has unique morphological
endothelium stimulated only one of the four isoforms of
features such as the loss of nuclear membrane 52 .
NADPH oxidase-NOX2, which is typical for neutrophils58.
The signaling mechanisms of NETosis may include
activation of phosphoinositide-3-kinase (PI3K)53, which The calcium ionophore A23187 causes NETosis by
also controls the autophagy inductionbut assembly of mobilising Ca2+ from the endoplasmic reticulum (ER),
autophagosomes is not required for NETosis54. which causes the activation of calcium release activated
channel (CRAC) in the cytoplasmic membrane and
There are two types of netosis viz., suicidal netosis
the passage of extracellular Ca2+ into the cytoplasm.
and vital netosis55. Suicidal NETosis induced by several
A23187 also catalyses the transport of Ca2+ over the
stimuli such as phorbol myristateacetate (PMA), (auto)
plasma membrane into the cytoplasm. Following Ca2+
antibodies or cholesterol crystals and occurs after hours
accumulation in the mitochondrial matrix, non-selective
of stimulation. ROS created as a result of nicotinamide
mPTP are activated, resulting in the production of
adenine dinucleotide phosphate (NADPH) oxidase
mtROS. Oxidative stress, in addition to Ca2+ excess, is
activation and peptidyl-argininedeiminase4 (PAD4)
an inducer of the mPTP. mtROS produced from the
is activated, resulting in chromatin decondensation.
mitochondria into the cytosol appear to activate NADPH
Followed by translocation of neutrophil elastase
oxidase with the help of PKC50.
(NE) and myeloperoxidase (MPO) into the nucleus to
promote additional chromatin unfolding resulting in Mechanism of NETosis
nuclear membrane breakdown. Chromatin is released The crucial event occurs into cell cytosol during
into the cytosol and enhanced with granular and NETosis is release of proteins from azurophil granules.
cytosolic proteins. Finally, NETs are released through These granules contain protein complex "azurosome",
plasma membrane breakdown and the neutrophil dies. having eight differentproteins, three of which are highly
Staphylococcus aureus induces vital NETosis within homologous serine proteases-NE, namely cathepsin
minutes via both complement receptors and toll like G, and azurocidin andmyeloperoxidase (MPO). MPO
receptor 2 (TLR2) ligands or E. coli directly via TLR4 or is an enzyme that generates hypochlorite anion from
indirectly via TLR4-activated platelets. PAD4 is activated chlorine and hydrogen peroxide as substrates. ROS cause
possibly without the need of oxidants which causes azurosome dissociationto release of serine proteases and
chromatin decondensation. NE is translocated into the MPO from the granules into the cytoplasm59.
nucleus as in suicidal NETosis to induce additional
Mainly NE-serine proteasesdegrade various
chromatin unfolding and nuclear-membrane rupture.
components of cytoskeleton to induce NETosis60. Then
Protein-decorated chromatinon the other hand released
they migrate to the nucleus where they decondensate
via vesicles and the neutrophil remains alive for other
chromatin, destroy the nuclear envelope and release
activities, such as phagocytosis. Mitochondrial ROS
chromatin into the cytoplasm by breaking down the
production involved in one or the other forms of NETosis
lamin and histones50. MPO plays an important role during
has not yet been completely elucidated55.
dissociation of the azurosome and the release of proteases
Mechanism of NET formation from the granules59.
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
196 Patel et al.

PAD4 is transported from the cytoplasm to the inducing substances can directly or indirectly influence
nucleus to catalyse citrullination of histones, which glutathione peroxidase via many mechanisms, resulting
results in chromatin decondensation. Ca2+ stimulates in a loss in antioxidant capacity and an accumulation of
the activity of peptididylarginine deaminases which lipid ROS in cells, ultimately leading to oxidative cell
are Ca2+ binding proteins50. In contrast, when hydrogen death64.
peroxide is added, PAD is activated and produces histone
Ferroptosis is characterized by obvious shrinkage
citrullination61. It has recently been demonstrated that
of mitochondria with increased membrane density and
NETosis can occur as a result of the activation of cyclin
reduction in or disappearance of mitochondrial cristae65,
dependent kinases (CDKs) which facilitate cell cycle
while the cell membrane remains intact, the nucleus is
entrance62. Parts of the mitosis apparatus such as lamina
normal in size and there is no chromatin concentration,
phosphorylation and centrosomes separation are used to
a process distinct from other modes of cell death66.
destroy nuclear envelope during NETosis.
Mechanism of ferroptosis
FERROPTOSIS Inducing ferroptosis by suppressing system Xc
Cystine/glutamate antiporter (System Xc, SxC) is an
The Latin word ferrum (iron) and the Greek
amino acid antitransporter which is widely distributed
word ptosis (a fall). Ferroptosis characterized by
in phospholipid bilayers (Fig. 4)64. It's a heterodimer
accumulation of significant amount of iron and lipid
made up of two subunitsof solute carrier genes / proteins
peroxidation leads to cell death. Erastin had a selectively
(SLC7A11) and SLC3A264. System Xc- exchanges cysteine
deadly effect on rat sarcoma (RAS) expressing cancer
and glutamate in and out of the cell at a 1:1 ratio65. Under
cells, although the mode of cell death was distinct from
the action of glutathione peroxidases (GPXs), glutathione
what had previously been observed. There were no
(GSH) reduces ROS and reactive nitrogen species.
nuclear morphological alterations, deoxyribonucleic acid
Blocking system Xc-activity affects GSH production by
(DNA) fragmentation or caspase activation and caspase
inhibitors could not reverse the process63. During the year inhibiting cystine absorption, resulting in a drop in GPX
2012, Dixon and collogues formally called this cell death activity decrease in cell antioxidant capacity, lipid ROS
ferroptosis after researching the method by which erastin accumulation and eventually, the incidence of oxidative
killed cancer cells with RAS mutations. damage and ferroptosis64. In addition, P53 can also inhibit
system Xc- uptake of cystine by downregulating the
Ferroptosis is closely associated to the expression of SLC7A11 leads to ferroptosis67.
pathophysiological processes of many diseases including
cancers, nervous system diseases, ischemia-reperfusion Inducing ferroptosis by suppressing GPX4
injury, kidney injury and blood diseases. Ferroptosis- GPX4 is a crucial regulator of ferroptosis and plays

Fig. 4. Regulatory pathways of ferroptosis.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023


Mechanisms of cell death 197

a critical role in its occurrence mostly by limiting the lysis and release of pro-inflammatory molecules such as
synthesis of lipid peroxides. GPX4 transforms GSH to IL 1β and IL 1876. It plays an important role in clearing
oxidized glutathione (GSSG) and decreases harmful lipid numerous bacterial and viral infections by eliminating
peroxides (L-OOH) to alcohols (L-OH)64. Downregulated intracellular replication habitats and enhancing the host's
GPX4 expression were more sensitive to ferroptosis defensive responses77. Pore-forming Gasdermin D and
while the upregulation of GPX4 expression inhibits Gasdermin E proteins activate pyroptosis and apoptosis
ferroptosis66. by permeabilizing the plasma and mitochondrial
membranes78.
Ferroptosis mediated by mitochondrial voltage depen-
dent anion channels (VDACs) Mechanism of pyroptosis
VDACs are transmembrane channels that carry ions Caspase-1 is activated by various inflammasome com-
and metabolites and are crucial in ferroptosis regulation68. plexes in innate immunity
Erastin works on VDACs, causing mitochondrial Tschopp (2002) proposed the inflammasome complex
malfunction and a huge amount of released oxides as a molecular platform for caspase-1 activation 79.
ultimately culminating in iron-mediated cell death69. In response to infection or a specific immunological
challenge the oligomeric inflammasome complex is
Role of iron metabolism in ferroptosis
assembled by a paern recognition receptor(PRR) and
Ferrous ions formed by intestinal absorption or
a downstream adaptor81 such as apoptosis-associated
erythrocyte degradation can be oxidized by ceruloplasmin
speck-like protein containing a caspase recruit domain
to Fe 3+, which binds to transferrin (TF) on the cell
(ASC) or (Nucleotide-binding and oligomerization
membrane to form TF-Fe3+, which forms a complex domain-NOD) NOD like receptor (NLRC4). PRR detects
through membrane protein TF receptor 1 (TFR1) to the specific stimulus and signals to the adaptor, which
endocytose this complex70. Fe3+ is then reduced to Fe2+ binds to caspase-1 and oligomerization of the PRRs
by six-transmembrane epithelial antigen of the prostate brings multiple caspase-1 molecules into proximity for
3 (STEAP3) and Fe2+ is then stored in the unstable labile activation80. The AIM2/ASC inflammasome recognises
iron pool (LIP) and ferritin, which is mediated by divalent cytosolic double-stranded DNA with AIM2 as the DNA-
metal transporter 1 (DMT1) or Zinc-Iron regulatory binding receptor; theneuronal apoptosis inhibitory
protein family 8/14 (ZIP8/14)64. Excess Fe2+ is oxidized to protein(NAIP)/NLRC4 inflammasomes directly recognise
Fe3+ by ferroportin (FPN)71. This recycling of internal iron bacterial flagellin and type III secretion apparatus with
strictly controls iron homeostasis in cells64. Heat shock NAIP and NLRC4 as the receptor and signalling amplifier
protein beta-1 (HSPB1) can further reduce intracellular respectively. NLRP3/ASC inflammasome activated
iron concentrations by inhibiting TRF1 expression by various membrane damaging agents, the Pyrin/
and so overexpressed HSPB1 can significantly inhibit ASC inflammasome detects bacterial toxins indirectly
ferroptosis72. by inactivating host Rho guanosine triphosphatases
Regulation of the lipid metabolism pathway (GTPases); NLRP1 inflammasomedetects anthrax lethal
Iron-dependent lipid ROS accumulation is involved toxin activity or Toxoplasma gondii infection81. Activated
in ferroptosis in all pathways. Polyunsaturated fay acids caspase cleave the gasdermin D (GSDMD) and trigger
(PUFAs) are sensitive to lipid peroxidation and are one the pyroptosis.
of the essential elements for ferroptosis73. Free PUFAs Caspase-11/4/5 also triggers pyroptosis upon recogni-
must be esterified into membrane phospholipids and tion of cytosolic lipopolysaccharide
oxidized to transmit the ferroptosis signal. Studies have Consistent with LPS being the major component of
shown that phosphatidylethanolamine (PE) is the key the gram-negative bacterial cell wall, cytoplasmic LPS
phospholipid that induces ferroptosis in cells. Acyl-CoA has been identified as the bacterial signal responsible
synthetase long-chain family member 4 (ACSL4) and for caspase-11-mediated pyroptosis82. Moreover, the
lysophosphatidylcholine acyltransferase 3 (LPCAT3) role of toll-like receptor 4 in determining endotoxic
participate in the biosynthesis and remodeling of PE64. shock appears to be limited to transcriptional priming
PUFA-PE can play further oxidative roles under the of caspase-11 rather than the presumed cytokine storm.
catalysis of lipoxygenase (LOX) and eventually induce Caspase-4 and caspase-5 appear to have the same
ferroptosis in cells74. function in humans and are both activated by direct LPS
binding. Thus, caspase-11 and caspase-4/5 function in
PYROPTOSIS innate immunity as both PRRs and pyroptosis inducers81.
Cookson and Brennan gave the term pyroptosis Caspase-1 and Caspase-11/4/5 cleave Gasdermin D to
(Greek word) in 2001 in which, “pyro” means fever trigger pyroptosis
and“ptosis” means falling off75. It is an inflammatory form GSDMD is made up of approximately 480 amino
of PCD characterized by cellular swelling and rupture, acids that are divided into two domains the N-terminal
INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023
198 Patel et al.

gasdermin-N domain and the C-terminal gasdermin-C 11. Khalid N, Azimpouran N. 2022. Stat Pearls: Necrosis. StatePearls
domain, which are linked by a long loop. Caspase-1 and publishing.
12. Vasei N, Shishegar A, Ghalkhani F, Darvishi M. 2019. Fat
caspase-11 are activated and efficiently cleave GSDMD
necrosis in the Breast: A systematic review of clinical. Lipids
at an aspartate site within the linking loop. The same Health Dis 18: 139.
cleavage was seen with LPS-activated caspase-4/5, and 13. Kerr JF. 2002. History of the events leading to the formulation
inhibiting it, renders cells completely resistant to cytosolic of the apoptosis concept. Toxicology 181: 471-474.
LPS83. 14. Norbury CJ, Hickson ID. 2001. Cellular responses to DNA
damage. Annu Rev Pharmacol 41: 367-401.
The Gasdermin-N domain of GSDMD forms mem- 15. Elmore S. 2007. Apoptosis: a review of programmed cell
brane pores to trigger pyroptosis death. Toxicol Pathol 35: 495-516.
In mammalian cells, expression of the gasdermin-N 16. Kurosaka K, Takahashi M, Watanabe N, Kobayashi Y. 2003.
domain of GSDMD is sufficient to induce pyroptosis. Silent cleanup of very early apoptotic cells by macrophages.
The gasdermin-C domain of full-length GSDMD J Immunol 171: 4672-4679.
17. Jacobson MD, Burne JF, King MP, Miyashrta T, Reed JC, Raff
inhibits its activity by binding to gasdermin-N. The MC. 1993. Bcl-2 blocks apoptosis in cells lacking mitochondrial
gasdermin-N domain is also extremely toxic to bacteria, DNA. Nature 361: 365-369.
raising the possibility that it could directly target and 18. Ruoslahti E, Reed JC. 1994. Anchorage dependence, integrins,
lyse the membrane. Recombinant gasdermin-N of and apoptosis. Cell 77: 477-478.
GSDMD binds to phosphoinositides and cardiolipin 19. Hart SP, Hasle C, Dransfield I. 1996. Recognition of apoptotic
two lipids known to be present in the mammalian cells by phagocytes. Experientia 52: 950-956.
20. Wong RS. 2011. Apoptosis in cancer: from pathogenesis to
cell plasma membrane and bacterial inner membrane, treatment. J Exp Clin Cancer Res 30: 1-14.
respectively. The oligomerized gasdermin-N causes 21. Igney FH, Krammer PH. 2002. Death and anti-death: tumour
severe liposome leakage or biomembranes lysis. resistance to apoptosis. Nat Rev Cancer 2: 277-288.
Properties of gasdermin-N domain are similar to bacterial 22. Schneider P, Tschopp J. 2000. Apoptosis induced by death
pore-forming toxins. GSDMD's gasdermin-N domain receptors. Pharmacochemistry Library 31: 281-286.
can form large pores on phosphoinositide or cardiolipin- 23. Abdu-Alhameed A, Azab Elsayed A. 2019. Mechanisms of
programmed cell death. J Appl Biotechnol Bioeng 6: 156-158.
containing liposomes and on liposomes made of natural 24. Danial NN, Korsmeyer SJ. 2004. Cell death: critical control
polar lipid mixtures84. points. Cell 116: 205-219.
25. Cao W, Li J, Yang K, Cao D. 2021. An overview of autophagy:
Mechanism, regulation and research progress. Bull Cancer 108:
REFERENCES
304-322.
1. Yan GE, Elbadawi M, Efferth T. 2020. Multiple cell death mo-
26. Kang S, Shin KD, Kim JH, Chung T. 2018. Autophagy-related
dalities and their key features. World Academy Sci J 2: 39-48.
(ATG) 11, ATG9 and the phosphatidylinositol 3-kinase control
2. Tait SW, Ichim G, Green DR. 2014. Die another way-non-apop-
ATG2-mediated formation of autophagosomes in Arabidop-
totic mechanisms of cell death. J Cell Sci 127: 2135-2144.
sis. Plant Cell Rep 37: 653-664.
3. Zachary JF. 2017. Pathologic Basis of Veterinary Disease: Mech-
27. Galati S, Boni C, Gerra MC, Lazzarei M, Buschini A. 2019.
anisms and Morphology of Cellular adaptations, injury, and
Autophagy: a player in response to oxidative stress and DNA
death (6th ed.). Elsevier Health Sciences. pp. 243.
damage. Oxid Med Cell Longev 2019.
4. Galluzzi L, Kroemer G. 2008. Necroptosis: a specialized path-
28. Stavoe AK, Holzbaur EL. 2020. Neuronal autophagy declines
way of programmed necrosis. Cell 135: 1161-1163.
5. Santagostino SF, Assenmacher CA, Tarrant JC, Adedeji AO, substantially with age and is rescued by over expression of
Radaelli E. 2021. Mechanisms of regulated cell death: Current WIPI2. Autophagy 16: 371-372.
perspectives. Vet Pathol 58: 596-623. 29. Ghosh AK, Mau T, O'Brien M, Garg S, Yung R. 2016. Impaired
6. Ruffolo PR. 1964. The pathogenesis of necrosis. I. Correlated autophagy activity is linked to elevated ER-stress and inflam-
light and electron microscopic observations of the myocardial mation in aging adipose tissue. Aging 8: 2525.
necrosis induced by the intravenous injection of papain. Am 30. Oshima M, Seki T, Kurauchi Y, Hisatsune A, Katsuki H. 2019.
J Pathol 45: 741-756. Reciprocal regulation of chaperone-mediated autophagy/
7. Guicciardi ME, Gores GJ. 2013. Complete lysosomal disrup- microautophagy and exosome release. Biol Pharm Bull 42:
tion: a route to necrosis, not to the inflammasome. Cell Cycle 12: 1394-1401.
1995. 31. Saxton RA, Sabatini DM. 2017. mTOR signaling in growth,
8. Vegad JL. 2008. A Textbook of Veterinary General Pathology: metabolism and disease. Cell 168: 960-976.
Cell Injury and Cell Death (2nd ed.). International Book Dis- 32. Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura
tributing Co. pp. 19-104. Y, Iemura S, Natsume T, Takehana K, Yamada N, Guan JL,
9. Marunouchi T, Tanonaka K. 2015. Cell Death in the Cardiac Oshiro N, Mizushima N. 2009. Nutrient-dependent mTORC1
Myocyte. Biol Pharm Bull 38: 1094-1097. association with the ULK1-Atg13-FIP200 complex required for
10. Chung AG, Frye JB, Zbesko JC, Constantopoulos E, Hayes M, autophagy. Mol Biol Cell 20: 1981-1991.
Figueroa AG, Becktel DA, Antony Day W, Konhilas JP, McKay 33. Kocaturk NM, Gozuacik D. 2018. Crosstalk between mamma-
BS, Nguyen TV, Doyle KP. 2018. Liquefaction of the Brain fol- lian autophagy and the ubiquitin-proteasome system. Front
lowing Stroke Shares a Similar Molecular and Morphological Cell Dev Biol 6: 128.
Profile with Atherosclerosis and Mediates Secondary Neurode 34. Chen J, Kos R, Garssen J, Redegeld F. 2019. Molecular insights
generation in an Osteopontin-Dependent Mechanism. eNeuro into the mechanism of necroptosis: the necrosome as a poten-
5. tial therapeutic target. Cells 8: 1486.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023


Mechanisms of cell death 199

35. Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima Vandenabeele P. 2011. Neutrophil extracellular trap cell death
N, Cuny GD, Mitchison TJ, Moskowi MA, Yuan J. 2005. requires both autophagy and superoxide generation. Cell Res
Chemical inhibitor of nonapoptotic cell death with therapeutic 21: 290-304.
potential for ischemic brain injury. Nat Chem Biol 1: 112-119. 54. Germic N, Stojkov D, Oberson K, Yousefi S, Simon HU. 2017.
36. Chan FKM, Luz NF, Moriwaki K. 2015. Programmed necrosis Neither eosinophils nor neutrophils require ATG 5-dependent
in the cross talk of cell death and inflammation. Annual Review autophagy for extracellular DNA trap formation. Immunol 152:
of Immunol 33: 79. 517-525.
37. Liu C, Zhang K, Shen H, Yao X, Sun Q, Chen G. 2018. Necro- 55. Jorch SK, Kubes P. 2017. An emerging role for neutrophil ex-
ptosis: a novel manner of cell death associated with stroke. Int tracellular traps in non infectious disease. Nat Med 23: 279-287.
J Mol Med 41: 624-630. 56. Lu DJ, Furuya W, Grinstein S. 1993. Involvement of multiple
38. Su Z, Yang Z, Xu Y, Chen Y, Yu Q. 2015. Apoptosis, autophagy, kinases in neutrophil activation. Blood Cells 19: 343-9.
necroptosis and cancer metastasis. Mol Cancer 14: 1-14. 57. Hakkim A, Fuchs TA, Martinez NE, Hess S, Prinz H, Zych-
39. You Z, Savi SI, Yang J, Degterev A, Yuan J, Cuny GD, Mos- linsky A, Waldmann H. 2011. Activation of the Raf MEK ERK
kowi MA, Whalen MJ. 2008. Necrostatin-1 reduces histo- pathway is required for neutrophil extracellular trap forma-
pathology and improves functional outcome after controlled tion. Nat Chem Biol 7: 75-77.
cortical impact in mice. J Cereb Blood Flow Metab 28: 1564-1573. 58. Nazarewicz RR, Dikalova AE, Bikineyeva A, Dikalov SI. 2013.
40. Davidovich P, Kearney CJ, Martin SJ. 2014. Inflammatory Nox2 as a potential target of mitochondrial superoxide and
outcomes of apoptosis, necrosis and necroptosis. Biol Chem its role in endothelial oxidative stress. Am J Physiol Heart Circ
395: 1163-1171. Physiol 305: H1131-H1140.
41. Aggarwal BB. 2003. Signalling pathways of the TNF super- 59. Meler KD, Goosmann C, Lubojemska A, Zychlinsky A,
family: a double-edged sword. Nat Rev Immunol 3: 745-756. Papayannopoulos V. 2014. A myeloperoxidase-containing
42. Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N. complex regulates neutrophil elastase release and actin dy-
2013. Crosstalk between apoptosis, necrosis and autopha- namics during NETosis. Cell Rep 8: 883-896.
gy. Biochimi Biophys Acta-Mol Cell Res 1833: 3448-3459. 60. Papayannopoulos V, Meler KD, Hakkim A, Zychlinsky A.
43. Orrenius S. 2007. Reactive oxygen species in mitochondria-me- 2010. Neutrophil elastase and myeloperoxidase regulate the
diated cell death. Drug Metab Rev 39: 443-455. formation of neutrophil extracellular traps. J Cell Biol 191:
44. Wu YT, Tan HL, Huang Q, Sun XJ, Zhu X, Shen HM. 2011. 677-691.
zVAD-induced necroptosis in L929 cells depends on autocrine 61. Li P, Li M, Lindberg MR, Kenne MJ, Xiong N, Wang Y. 2010.
production of TNFα mediated by the PKC-MAPKs-AP-1 PAD4 is essential for antibacterial innate immunity mediated
pathway. Cell Death Differ 18: 26-37. by neutrophil extracellular traps. J Exp Med 207: 1853-1862.
45. Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ. 2006. Activation 62. Amulic B, Knackstedt SL, Abu Abed U, Deigendesch N, Har-
of IKK by TNFα requires site-specific ubiquitination of RIP1 bort CJ, Caffrey BE, Brinkmann V, Heppner FL, Hinds PW,
and polyubiquitin binding by NEMO. Mol Cell 22: 245-257. Zychlinsky A. 2017. Cell-Cycle Proteins Control Production
46. He S, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X. 2009. of Neutrophil Extracellular Traps. Dev Cell 43: 449-462.
Receptor interacting protein kinase-3 determines cellular 63. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. 2003. Iden-
necrotic response to TNF-α. Cell 137: 1100-1111. tification of genotype-selective antitumor agents using syn-
47. Deutsch M, Graffeo CS, Rokosh R, Pansari M, Ochi A, Levie thetic lethal chemical screening in engineered human tumor
EM, Van Heerden E, Tippens DM, Greco S, Barilla R, Tom- cells. Cancer Cell 3: 285-296.
koer L, Zambirinis CP, Avanzi N, Gulati R, Pachter HL, 64. Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B, Wang G.
Torres-Hernandez A, Eisenthal A, Daley D, Miller G. 2015. 2020. Ferroptosis: past, present and future. Cell Death Dis 11:
Divergent effects of RIP1 or RIP3 blockade in murine models 88.
of acute liver injury. Cell Death Dis 6: e1759. 65. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev
48. Irrinki KM, Mallilankaraman K, Thapa RJ, Chandramoorthy EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS,
HC, Smith FJ, Jog NR, Gandhirajan RK, Kelsen SG, Houser Morrison B, Stockwell BR. 2012. Ferroptosis: an iron-depen-
SR, May MJ, Balachandran S, Madesh M. 2011. Requirement dent form of nonapoptotic cell death. Cell 149: 1060-1072.
of FADD, NEMO and BAX/BAK for aberrant mitochondrial 66. Yang WS, Stockwell BR. 2008. Synthetic lethal screening iden-
function in tumor necrosis factor alpha-induced necrosis. Mol tifies compounds activating iron-dependent, nonapoptotic cell
Cell Biol 31: 3745-3758. death in oncogenic RAS harboring cancer cells. Chem Biol 15:
49. Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, 234-245.
Liu W, Lei X, Wang X. 2012. Mixed lineage kinase domain-like 67. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, Gu
protein mediates necrosis signaling downstream of RIP3 ki- W. 2015. Ferroptosis as a p53-mediated activity during tumour
nase. Cell 148: 213-227. suppression. Nature 520: 57-62.
50. Vorobjeva NV, Chernyak BV. 2020. NETosis: molecular mech- 68. Skonieczna M, Cieslar Pobuda A, Saenko Y, Foksinski M,
anisms, role in physiology and pathology. Biochemistry 85: Olinski R, Rzeszowska Wolny J, Wiechec E. 2017. The impact
1178-1190. of DIDS-induced inhibition of voltage-dependent anion chan-
51. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann nels (VDAC) on cellular response of lymphoblastoid cells to
Y, Weiss DS, Weinrauch Y, Zychlinsky A. 2004. Neutrophil ionizing radiation. Med Chem 13: 477-483.
extracellular traps kill bacteria. Science 303: 1532-1535. 69. Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang
52. Fuchs TA, Abed U, Goosmann C, Hurwi R, Schulze I, Wahn WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface
V, Weinrauch Y, Brinkmann V, Zychlinsky A. 2007. Novel cell JJ, Smith R, Lessnick SL, Sahasrabudhe S, Stockwell BR. 2007.
death program leads to neutrophil extracellular traps. J Cell RAS RAF MEK dependent oxidative cell death involving
Biol 176: 231-241. voltage dependent anion channels. Nature 447: 864-868.
53. Remijsen Q, Vanden Berghe T, Wirawan E, Asselbergh B, 70. Frazer DM, Anderson GJ. 2014. The regulation of iron trans-
Parthoens E, De Rycke R, Noppen S, Delforge M, Willems J, port. Biofactors 40: 206-214.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023


200 Patel et al.

71. Bogdan AR, Miyazawa M, Hashimoto K, Tsuji Y. 2016. Reg- 78. Zheng Z, Li G. 2020. Mechanisms and therapeutic regulation
ulators of iron homeostasis: new players in metabolism, cell of pyroptosis in inflammatory diseases and cancer. Int J Mol
death and disease. Trends Biochem Sci 41: 274-286. Sci 21: 1456.
72. Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, Wang H, Cao L, 79. Martinon F, Burns K, Tschopp J. 2002. The inflammasome:
Tang D. 2015. HSPB1 as a novel regulator of ferroptotic cancer a molecular platform triggering activation of inflammatory
cell death. Oncogene 34: 5617-5625. caspases and processing of proIL-β. Mol Cell 10: 417-426.
73. Yang WS, Stockwell BR. 2016. Ferroptosis: death by lipid 80. Broz P, Dixit VM. 2016. Inflammasomes: mechanism of assem-
peroxidation. Trends Cell Biol 26: 165-176. bly, regulation and signalling. Nat Rev Immunol 16: 407-420.
74. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar 81. Shi J, Gao W, Shao F. 2017. Pyroptosis: gasdermin mediated
HH, Liu B, Tyurin VA, Ritov VB, Kapralov AA, Amoscato programmed necrotic cell death. Trends Biochem Sci 42: 245-254.
AA, Jiang J, Anthonymuthu T, Mohammadyani D, Yang Q, 82. Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. 2013.
Proneth B, Klein-Seetharaman J, Watkins S, Bahar I, Bayir H. Cytoplasmic LPS activates caspase-11: implications in TLR4
2017. Oxidized arachidonic and adrenic PEs navigate cells to independent endotoxic shock. Science 341: 1250-1253.
ferroptosis. Nat Chem Biol 13: 81-90. 83. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y. Cai
75. Cookson BT, Brennan MA. 2001. Pro inflammatory progra- T, Wang F, Shao F. 2015. Cleavage of GSDMD by inflammatory
mmed cell death. Trends Microbiol 9: 113-114. caspases determines pyroptotic cell death. Nature 526: 660-665.
76. Wallach D, Kang TB, Kovalenko A. 2014. Concepts of tissue 84. Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC,
injury and cell death in inflammation: a historical perspec- Shao F. 2016. Poreforming activity and structural autoinhibi-
tive. Nat Rev Immunol 14: 51-59. tion of the gasdermin family. Nature 535: 111-116.
77. Jorgensen I, Miao EA. 2015. Pyroptotic cell death defends
against intracellular pathogens. Immunol Rev 265: 130-142.

INDIAN JOURNAL OF VETERINARY PATHOLOGY | Volume 47 | Issue 3 | JULY - SEPTEMBER, 2023

You might also like