Open access

Ethanol-Induced Mitochondrial Induction of Cell Death-Pathways Explored

Written By

Harish Chinna Konda Chandramoorthy, Karthik Mallilankaraman and Muniswamy Madesh

Submitted: 15 March 2011 Published: 11 January 2012

DOI: 10.5772/30173

From the Edited Volume

Trends in Alcoholic Liver Disease Research - Clinical and Scientific Aspects

Edited by Ichiro Shimizu

Chapter metrics overview

2,916 Chapter Downloads

View Full Metrics

1. Introduction

Alcohol consumption is one of the major source for chronic liver diseases. It is striking that women are more susceptible to the toxic effects of alcohol although alcoholic liver disease (ALD) is common in men (1). In recent times, global burden on ALD has prompted researchers to investigate this disease based on age, gender, social status and race. However, in all these conditions and known variable severities of ALD, the basic pathophysiological condition is oxidative stress, which leads to liver damage (1, 2). In an overview, ALD leads to hepatocyte death, liver cirrhosis and organ dysfunction through production of reactive oxygen species (ROS), inflammatory cytokines and mitochondrial impairment. ROS are important mediators of apoptosis in liver diseases and are produced in response to paracrine factors such as ethanol (EtOH) (2). This chapter focuses on the role of EtOH induced ROS mediated cell death.

Over two decades, several pathways have been proposed in ALD. Recent studies have educated our understanding on these pathways, most of which work as cohort induced by direct/indirect effects of alcohol metabolism and clearance. Majority of cell death pathways (apoptosis, necrosis and the recently described necroptosis) converge at cellular damage associated with excessive production of ROS (superoxide (O2•–) and hydrogen peroxide (H2O2)) that results in oxidative stress (3, 4). Under pathophysiological conditions, NAD(P)H oxidase, xanthine oxidase (XO) and the mitochondrial respiratory chain are the major sources of ROS. Normally, 5% of the metabolized cellular oxygen is converted into ROS which are effectively detoxified by endogenous antioxidants such as superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase (Cat). ROS overproduction resulting from acute and chronic exposure to alcohol can exceed the capacity of endogenous antioxidants (5, 6). Excessive ROS triggers various cellular signaling pathways leading to cell death in both vascular and epithelial cells. Although ROS is known to elicit liver damage, the signaling pathways operative in alcohol induced ROS overproduction in liver cells remain elusive.

Mitochondrial respiratory chain is the second major source of cellular ROS. However, mitochondria itself is an important target for cellular ROS resulting in mitochondrial dysfunction and permeabilization of outer mitochondrial membrane (OMM) (7, 8). In addition, studies have demonstrated that inhibition of mitochondrial electron transport results in ROS production leading to alteration in mitochondrial morphology and bioenergetics (9). Furthermore, OMM permeabilization leads to cytochrome c release and mitochondrial dysfunction (10).

Multidomain proapoptotic Bcl-2 family proteins are suggested to play a role in O2•– induced mitochondrial dysfunction (11, 12). Studies have shown that chronic EtOH consumption increases the expression of anti-apoptotic Bcl-2 and Bcl-xL proteins by an interleukin-6-dependent mechanism (13, 14). Though, up regulation of proapoptotic Bax protein is observed in patients with ALD, the roles of Bax and Bak in initiating mitochondrial apoptotic events are poorly understood. Our previous studies have shown that O2•–-mediated mitochondrial phase of apoptosis is mainly dependent on Bid but not Bax (15, 16).

Enhanced circulation of TNF-α and other cytokines have been reported in both ALD patients and animal models (17). In ALD, alcohol-induced O2•– elicits production of proinflammatory cytokine such as TNF-α which subsequently sensitizes hepatocyte cell death through gangliosides (18, 23). Interestingly, in hepatocytes, TNF-α binds to either TNFR1 (type1 tumor necrosis factor receptor) or TNFR2 (type 2 tumor necrosis factor receptor) to initiate cell death. TNF-α mediated activation of apoptosis requires two adaptor molecules such as TNF receptor associated death domain protein (TRADD) and Fas –activated death domain protein (FADD). These in turn activate caspase 8 which further proteolytically cleaves downstream caspases and pro apoptotic bcl-2 family protein Bid. The active form of Bid (t-Bid) facilitates OMM permeabilization (15). On the other hand, ligation of TNF-α−TNFR1 recruits receptor-interacting protein 1 kinase (RIP1), TNFR death domain serine-theronine kinase 2 (TRAF2) which generates ceramide via activation of sphingomyelinases. Ceramide induces mitochondrial permeability transition pore (MPTP) opening, mitochondrial matrix swelling and membrane permeabilization, in concert with pro-apoptotic Bcl-2 family protein Bad (24). Recently our study has shown that TNF-α-induced necroptosis, the alternate form of cell death, requires TNFR adaptor protein FADD and NFκB downstream signaling molecule NEMO. FADD mediates the formation of necrosome consisting of RIP1-RIP3 kinases. The necrosome induced mitochondrial dysfunction in necroptosis requires Bax and Bak (25). TNFR1 mediated cell death is an extensively studied model and has been associated in many disease conditions including ALD.

Ca2+ has been known as an important intracellular second messenger that plays a dual role in cell survival and death. In liver, Ca2+ signaling is known to regulate a variety of cellular functions ranging from proliferation to apoptosis. Under pathological conditions, elevation in intracellular calcium ([Ca2+]i) facilitates cell death (26, 27) via inositol 1,4,5-triphosphate (InsP3) (28, 29) and oxidation of STIM1(30). Inositol 1,4,5-triphosphate receptor (InsP3R) mediated [Ca2+]i changes leads to rapid Ca2+ release from ER and the subsequent Ca2+ entry through slow-activating plasma membrane store operated channels (SOC) (31, 33). In hepatocytes, the Type II InsP3 R is known to trigger Ca2+ waves that can transmit through intercellular junctions throughout the liver (34). ER-mitochondria link and the mitochondrial Ca2+ ([Ca2+]m) uptake through uniporter is known to promote [Ca2+]m overload which subsequently leads to mitochondrial depolarization and increased mROS production (10, 28, 35, 36). The aberrant Ca2+ homeostasis has been linked with ALD (37, 38). Despite the vast knowledge, the actual intricacies on the mechanism of Ca2+ induced mitochondrial dysfunction remain largely unexplored. In addition to the functional damage, the structural damage to the mitochondrion is known to play a very important role in accelerating EtOH induced apoptosis in hepatocytes. In support, a recent study has evidenced the mitochondrial structural changes (fig.1) in an animal model for ALD (39).

Figure 1.

Mitochondria appearance under electron microscope (EM × 6000); A: Mitochondria in normal group; B: Mitochondria in model group. M: mitochondria, G: glycogen, N nucleus, ER: endoplasmic reticulum, LD: lipid droplet. The long arrow shows abnormally distributed chromatin in nuclei, the short one is megamitochondrion and the arrow head is U-type mitochondria (Electron micrograph reproduced with permission from 2007 Yan, M et al. Originally published in World J Gastroenterology 2007April 28;13(16): 2352-2356).

Advertisement

2. Role of ethanol in ROS production

Oxidative stress has been implicated to play a major role in ALD. The formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) represent an important cause of oxidative injury associated with free radical formation. ROS is known to damage and degrade lipids, proteins and DNA by which it affects the structure and function of the cell. Using animal models and samples from subjects with ALD, studies have shown the role of ROS in EtOH induced tissue damage (40, 41). Modification of mitochondrial proteins by ROS to disulphide, sulphenic, sulphinic and sulphonic residues and RNS to nitration products of tyrosine residues and nitrosation products of thiols have been well documented to occur in membrane and matrix proteins within mitochondria (42, 43). This section describes in detail the role of ROS in ALD. Oxygen is foremost common chemical frequently involved in the formation of free radical. Molecular oxygen is oxidized to generate two molecules of water by accepting four electrons and protons at one time. During this process several intermediary state of reactants exist like superoxide (O2•–); peroxide (O22-), which normally exists in cells as hydrogen peroxide (H2O2); and the hydroxyl radical (OH). Superoxide, peroxide, and the hydroxyl radical are considered the primary free radicals. It has been estimated that only about 3 to 5 percent of the O2 consumed by the mitochondrial respiratory chain is converted to ROS. Nevertheless, the toxic effects of oxygen in biological systems—such as oxidation of lipids, inactivation of enzymes, nucleic acid mutations and destruction of cell membranes are attributed to the reduction of O2 to free radicals. The first and foremost effect of alcohol metabolism in the cellular milieu is the loss of NAD+/NADH ratio that affects mitochondrial respiratory chain and subsequent generation of superoxide anion (44). In respect to EtOH induced ROS

production, our laboratory has demonstrated that EtOH induced mROS production lead to mitochondrial morphology changes and functional alterations (Fig. 2). Briefly, (1) Acute delivery of EtOH (50mM) resulted in mitochondrial fragmentation (filamentous to globular morphology - fig.2A). (2) EtOH-fragmented mitochondria exhibit exaggerated O2•–production (fig.2B ). (3) EtOH treatment induced elevated mROS, altered mitochondrial Ca2+ handling and mitochondrial dysfunction (fig.2D&E). (4) O2•– induced mitochondrial membrane potential (ΔΨm) loss and cytochrome c release was abrogated by the antiapoptotic Bcl-2 protein Bcl-xL and (5) Bax/Bak double knockout cells are resistant to O2•– -mediated ΔΨm loss and cytochrome c release, however, Bak but not Bax is essential for O2•–-induced ΔΨm loss and cytochrome c release (fig 3A-D).

Figure 2.

EtOH augments alterations of mitochondrial morphology, O2•– production, and mitochondrial Ca2+ uptake in live cells. (A) Mito-eGFP (enhanced GFP)-expressing vascular endothelial cells (left panel) were exposed to 50 mM EtOH for 30 h (right panel). EtOH treatment resulted in short, globular mitochondrial tubules. (B) Mito-eGFP-expressing cells either left untreated (top) or exposed for 30 h to 50 mM EtOH (bottom) were loaded with the mitochondrion-derived O2.– indicator MitoSOX Red and imaged by confocal microscopy. EtOH-treated cells, but not control cells, displayed enhanced mitochondrial O2•– production. (C) Quantitation of mitochondrial ROS production in live cells. Following treatment, cells were loaded with the mitochondrial Ca2+ indicator rhod-2 for 45 min and stimulated with bradykinin (BK; 10 nM). Representative traces of mitochondrial Ca2+ uptake in response to bradykinin in (D) control and (E) EtOH-treated cells. EtOH-treated cells, but not control cells, displayed sustained mitochondrial Ca2+ elevation. f.a.u., fluorescence arbitrary units. (Reproduced with permission from © 2009 Madesh et al. Originally published in Mol Cell Biol. 2009 Jun;29(11):3099-112).

Figure 3.

A) Wild type, bax–/– bak–/– double knockout, bax–/– and bak–/– MEFs were probed for cytochrome c in O2•– -generating system. ΔΨm was measured after O2•– treatment in permeabilized, TMRE-loaded bax–/– bak–/– MEFs expressing (B) GFP alone or together with (C) Bak or (D) Bax. Cells were exposed to the O2•– -generating system or FCCP as indicated. (Reproduced with permission from © 2009 Madesh et al. Originally published in Mol Cell Biol. 2009 Jun;29(11):3099-112).

Taken together it is evident that O2•–evokes mitochondrial phase of apoptosis during chronic EtOH exposure. In addition, O2•– mediated tBid generation induces selective activation of mitochondrial Bak, triggering cytochrome c release and ΔΨm loss that lead to apoptosis (15). Though mitochondria is known to play a crucial role in EtOH induced cell death, the upstream signaling molecules other than O2•– that target mitochondria is a open area of research in ALD.

Advertisement

3. Calcium and its role in ROS mediated apoptosis

[Ca2+]m signals are known to control variety of responses in liver including apoptosis. Chronic EtOH exposure in rats leads to sustained Ca2+ elevation that triggers MPTP opening. MPTP opening leads to Ca2+ overload in the mitochondria and results in mitochondrial swelling a phenomenon observed in EtOH fed rats but not in control rats (45). Cells at basal metabolic rate tightly regulate free Ca2+ in the range of 100 to 200 nM in both cytosol and mitochondria through NCX (Na+/Ca2+ exchanger), PMCA (Plasma membrane Ca2+-ATPase) and SERCA (Sarcoendoplasmic reticulum Ca2+-ATPase) pumps. Mitochondria play an important role in rapid uptake of Ca2+ through a uniporter and is then released slowly back into the cytosol (46, 48). EtOH is known to induce elevated [Ca2+]i by altering the [Ca2+]m buffering capacity. Endothelial cells lining the capillaries and veins are first to encounter ethanol. Ethanol exposure activates the endothelial cells which are known to signal the immune cells. Our studies have previously shown ROS generation by activated macrophages evoked an [Ca2+]i transient in endothelial cells (28). However sustained increase in [Ca2+]i coupled with altered mitochondrial Ca2+ handling capacity leads to irreversible cell injury (16, 28, 49). Though, the exact source of increased cellular Ca2+ in ALD is poorly understood, several pathways have been proposed for the increased calcium flux. Receptor mediated pathways (G Protein-Coupled Receptor and tyrosine kinase receptor) that generate second messengers like InsP3 which binds to InsP3R on endoplasmic reticulum trigger Ca2+ release (50). Further the [Ca2+]m uptake was directly proportional to the magnitude of [Ca2+]c. Under pathophysiological conditions, the GPCR (G Protein-Coupled Receptor) Ca2+ linked mROS is essential for leukocyte/endothelial cell adhesion (50). EtOH exposure in HepG2 cells induces [Ca2+]m overload that triggers mROS (fig 2D & E). In the cellular milieu, Ca2+ is compartmentalized as gradients in different organelles in the range of μM to nM (Ca2+=ER>mitochondria>lysosomes>cytosol=nucleus). During ALD the alterations in Ca2+ homeostasis leads to [Ca2+]m overload. Under pathological or physiological conditions [Ca2+]m levels dictate the cells to program either towards cell death or survival signals in the liver. Accumulation of Ca2+ in mitochondria beyond the transition threshold opens the MPTP, resulting in Δψm loss, mitochondrial swelling, mROS overproduction and finally leading to cell death (51).

Advertisement

4. Mitochondrial permeability transition

Ca2+-linked cell death program in ALD may be either apoptotic or necrotic phenomenon determined by OMM permeabilization and MPTP opening respectively. Ca2+ overload leads to oxidative stress that permanently leads to MPTP opening exposing the mitochondrial inner membrane permeable to all solutes of molecular weight up to 1.5Kd (39). Furthermore, the persistent MPTP opening leads to irreversible mitochondrial depolarization. Mitochondrial depolarization, in conjunction with mROS overproduction and subsequent inner mitochondrial membrane (IMM) damage sets the stage for apoptosis (52). A major pathway that leads to mitochondrial damage in a broad spectrum of inflammatory or ischemia-related conditions results from the amplification of mitochondrial and cytosolic O2•– production (53). ROS mediated cell death, in particular O2•–-mediated apoptosis, begins with rupture of the outer mitochondrial membrane (OMM) and cytochrome c release that subsequently trigger MPTP opening resulting in mitochondrial swelling. MPTP opening is also known to be involved in initiation of the apoptotic machinery without damage to the OMM. ROS and [Ca2+]m overload acts synergistically to trigger MPTP opening, and evokes cytochrome c release and subsequent activation of caspases (10).

O2•– or H2O2 exposure amplifies the Ca2+-induced MPTP opening in a permeabilized cell system which in turn could be attenuated with either O2•– scavengers SOD or SOD mimetic, MnTBAP, or H2O2 scavenger catalase (fig 4A & B). However, O2•– -induced cytochrome c release was insensitive to inhibitors of MPTP (16). Thus, MPTP opening is not essential for O2•–-induced cytochrome c release. In addition, exogenous delivery of cytochrome c eliminated the O2•– -induced ΔΨm loss. These data suggest that integrity of the IMM and matrix space was preserved during O2•– -induced cytochrome c release (15, 16).

Figure 4.

Effect of ROS on Ca2+-induced PTP opening and Cytochrome c release in permeabilized HepG2 cells. (A) O2•– -generating system (xanthine [0.1mM] plus xanthine oxidase [20 mU/ml]) and (B) H2O2 (90 mM) augmented Ca2+-induced depolarization (three pulses, 30 M CaCl2 each) and decreased mitochondrial Ca2+ uptake. These effects were inhibited by an O2•–-scavenger, MnTBAP (20 μM; 68 ±4.5% decrease in depolarization and 78 ±13% decrease in [Ca2+]c rise at 900 s; n= 3), and catalase (Cat; 2500U/ml), respectively. At the end of the measurements, cells were exposed to FCCP (Unc; 1μM), a protonophore that caused rapid and complete dissipation of ΔΨm. (Reproduced with permission from © 2001 Madesh and Hajnóczky. Originally published in J. Cell Biol. 155:1003-1015).

Advertisement

5. Role of Bcl-2 family proteins in ROS-induced Δψm loss

Although ROS-induced Ca2+ dependent MPTP opening is associated with cytochrome c release, in particular, superoxide selectively triggers OMM permeabilization and cytochrome c release independent of Ca2+ dependant MPTP opening. O2•– produced by the mitochondrial respiratory chain has been reported to cause cardiolipid destruction in the IMM and dissipation of the ΔΨm (54, 55). However, O2•– produced under various pathophysiological conditions including ALD, causes OMM permeabilization in a Bax/Bak dependant manner. Antiapoptotic Bcl-2 family protein Bcl-xL prevents O2•–-induced ΔΨm loss and cytochrome c release, implying a role for proapoptotic Bcl-2 proteins Bax and Bak. Despite their high homology, Bax and Bak have distinct subcellular localization and functional regulation. Bax is largely a cytosolic protein that undergoes conformational change that is prerequisite for mitochondrial phase of apoptosis. In contrast, Bak is a mitochondrial integral membrane protein which undergoes oligomerization upon activation by proapoptotic BH3-only proteins (tBid). O2•– -induced mitochondrial functional changes require either Bax or Bak. BH3 which constitute a subset of pro-apoptotic members of the Bcl-2 protein family are necessary to induce apoptosis (10, 56). O2•– -mediated ΔΨm loss and cytochrome c release is absent in Bax/Bak (bax–/– bak–/–) doubly deficient cells. Interestingly, Bak is necessary and sufficient for O2•–-induced ΔΨm loss and cytochrome c release. Mitochondria isolated from heart of bak–/– mice are resistant to O2•–-induced mitochondrial depolarization. Further, bid–/– deficient MEFs are also insensitive to O2•– -induced mitochondrial phase of apoptosis. Conversely, mitochondria from Bax-deficient mice display O2•– -induced mitochondrial depolarization. Upon TNF, Fas ligand or O2•– challenge, the cytosolic BH3-only protein Bid undergoes proteolytic processing (caspase 8 and caspase 2) to generate active form of Bid-tBid. tBid elicited O2•–-induced mitochondrial depolarization and cytochrome c release requires Bak. Taken together, these findings

Figure 5.

Mitochondria are prime target for EtOH-induced cell death-Scheme.

implicate the requirement of Bak and Bid for O2•–-induced ΔΨm loss and cytochrome c release (15, 16, 24, 10, 57).

Advertisement

6. Conclusion

The aberrant rate of cell death is a hallmark of ALD. It is evident that ethanol induced ROS mediated oxidative stress is responsible for induction of apoptosis. The sequential events such as changes in redox status, increase in cytosolic ROS, sustained [Ca2+]m elevation and translocation of pro-apoptotic proteins from cytosol to mitochondria are intimately linked with ethanol metabolism (fig 5). Major cell death pathways such as apoptosis, necrosis and the recently described necroptosis are associated with oxidative stress. Though, ROS production is proposed as a major factor in ethanol induced cell death little is known about the downstream mechanisms of the multimode cell death. In conclusion, mitochondria are prime target where multiple stress signaling pathways converge to induce cell death in the context of ALD.

Advertisement

7. Acknowledgements

This work was supported by the National Institutes of Health grant (R01 HL086699, HL086699-01A2S1, 1S10RR027327-01) to MM. We thank Yanling Zheng, Temple University for her great help in literature search.

References

  1. 1. World Health Organization. Global status report on alcohol and health, World Health Organization, Geneva.
  2. 2. WuD.CederbaumA. I. 2003 Alcohol, oxidative stress, and free radical damage, Alcohol Res Health 27 277284 .
  3. 3. ShawS.JayatillekeE.RossW. A.GordonE. R.LeiberC. S. 1981 Ethanol-induced lipid peroxidation: potentiation by long-term alcohol feeding and attenuation by methionine, J Lab Clin Med 98 417424 .
  4. 4. WheelerM. D.KonoH.YinM.RusynI.FrohM.ConnorH. D.MasonR. P.SamulskiR. J.ThurmanR. G. 2001 Delivery of the Cu/Zn-superoxide dismutase gene with adenovirus reduces early alcohol-induced liver injury in rats, Gastroenterology 120 12411250 .
  5. 5. FinkelT.HolbrookN. J. 2000 Oxidants, oxidative stress and the biology of ageing, Nature 408 239247 .
  6. 6. ThannickalV. J.FanburgB. L. 2000 Reactive oxygen species in cell signaling, Am J Physiol Lung Cell Mol Physiol 279, L10051028 .
  7. 7. AdrainC.CreaghE. M.MartinS. J. 2001 Apoptosis-associated release of Smac/DIABLO from mitochondria requires active caspases and is blocked by Bcl-2, EMBO J 20 66276636 .
  8. 8. VaughnA. E.DeshmukhM. 2008 Glucose metabolism inhibits apoptosis in neurons and cancer cells by redox inactivation of cytochrome c, Nat Cell Biol 10 14771483 .
  9. 9. Gonzalez-FlechaB.CutrinJ. C.BoverisA. 1993 Time course and mechanism of oxidative stress and tissue damage in rat liver subjected to in vivo ischemia-reperfusion, J Clin Invest 91 456464 .
  10. 10. MadeshM.ZongW. X.HawkinsB. J.RamasamyS.VenkatachalamT.MukhopadhyayP.DoonanP. J.IrrinkiK. M.RajeshM.PacherP.ThompsonC. B. 2009 Execution of superoxide-induced cell death by the proapoptotic Bcl-2-related proteins Bid and Bak, Mol Cell Biol 29 30993112 .
  11. 11. DuC.FangM.LiY.LiL.WangX. 2000 Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition, Cell 102 3342 .
  12. 12. MikhailovV.MikhailovaM.DegenhardtK.VenkatachalamM. A.WhiteE.SaikumarP. 2003 Association of Bax and Bak homo-oligomers in mitochondria. Bax requirement for Bak reorganization and cytochrome c release, J Biol Chem 278 53675376 .
  13. 13. Kendrick, S. F., O’Boyle, G., Mann, J., Zeybel, M., Palmer, J., Jones, D. E., and Day, C. P. Acetate, the key modulator of inflammatory responses in acute alcoholic hepatitis, Hepatology 51, 1988-1997.
  14. 14. HongF.KimW. H.TianZ.JarugaB.IshacE.ShenX.GaoB. 2002 Elevated interleukin-6 during ethanol consumption acts as a potential endogenous protective cytokine against ethanol-induced apoptosis in the liver: involvement of induction of Bcl-2 and Bcl-x(L) proteins, Oncogene 21 3243 .
  15. 15. MadeshM.AntonssonB.SrinivasulaS. M.AlnemriE. S.HajnoczkyG. 2002 Rapid kinetics of tBid-induced cytochrome c and Smac/DIABLO release and mitochondrial depolarization, J Biol Chem 277 56515659 .
  16. 16. MadeshM.HajnoczkyG. 2001 VDAC-dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release, J Cell Biol 155 10031015 .
  17. 17. HoekJ. B.PastorinoJ. G. 2002 Ethanol, oxidative stress, and cytokine-induced liver cell injury, Alcohol 27 6368 .
  18. 18. NiemelaO.ParkkilaS.PasanenM.IimuroY.BradfordB.ThurmanR. G. 1998 Early alcoholic liver injury: formation of protein adducts with acetaldehyde and lipid peroxidation products, and expression of CYP2E1 and CYP3A, Alcohol Clin Exp Res 22 21182124 .
  19. 19. ThurmanR. G. 1998 II. Alcoholic liver injury involves activation of Kupffer cells by endotoxin, Am J Physiol 275, G605611 .
  20. 20. KishoreR.HillJ. R.Mc MullenM. R.FrenkelJ.NagyL. E. 2002 ERK1/2 and Egr-1 contribute to increased TNF-alpha production in rat Kupffer cells after chronic ethanol feeding, Am J Physiol Gastrointest Liver Physiol 282, G615 .
  21. 21. LemastersJ. J.NieminenA. L.QianT.TrostL. C.ElmoreS. P.NishimuraY.CroweR. A.CascioW. E.BradhamC. A.BrennerD. A.HermanB. 1998 The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophagy, Biochim Biophys Acta 1366 177196 .
  22. 22. HatanoE.BradhamC. A.StarkA.IimuroY.LemastersJ. J.BrennerD. A. 2000 The mitochondrial permeability transition augments Fas-induced apoptosis in mouse hepatocytes, J Biol Chem 275 1181411823 .
  23. 23. HoekJ. B.CahillA.PastorinoJ. G. 2002 Alcohol and mitochondria: a dysfunctional relationship, Gastroenterology 122 20492063 .
  24. 24. RoyS. S.MadeshM.DaviesE.AntonssonB.DanialN.HajnoczkyG. 2009 Bad targets the permeability transition pore independent of Bax or Bak to switch between Ca2+-dependent cell survival and death, Mol Cell 33 377388 .
  25. 25. IrrinkiK. M.MallilankaramanK.ThapaR. J.ChandramoorthyH. C.SmithF. J.JogN. R.GandhirajanR. K.KelsenS. G.HouserS. R.MayM. J.BalachandranS.MadeshM. 2011 Requirement of FADD, NEMO and BAX/BAK for Aberrant Mitochondrial Function in TNF{alpha}-Induced Necrosis, Mol Cell Biol.
  26. 26. HajnoczkyG.DaviesE.MadeshM. 2003 Calcium signaling and apoptosis, Biochem Biophys Res Commun 304 445454 .
  27. 27. OrreniusS.ZhivotovskyB.NicoteraP. 2003 Regulation of cell death: the calcium-apoptosis link, Nat Rev Mol Cell Biol 4 552565 .
  28. 28. MadeshM.HawkinsB. J.MilovanovaT.BhanumathyC. D.JosephS. K.RamachandraraoS. P.SharmaK.KurosakiT.FisherA. B. 2005 Selective role for superoxide in InsP3 receptor-mediated mitochondrial dysfunction and endothelial apoptosis, J Cell Biol 170 10791090 .
  29. 29. SzalaiG.KrishnamurthyR.HajnoczkyG. 1999 Apoptosis driven by IP(3)-linked mitochondrial calcium signals, EMBO J 18 63496361 .
  30. 30. HawkinsB. J.IrrinkiK. M.MallilankaramanK.LienY. C.WangY.BhanumathyC. D.SubbiahR.RitchieM. F.SoboloffJ.BabaY.KurosakiT.JosephS. K.GillD. L.MadeshM.S-glutathionylationactivates. S. T. I. 1 and alters mitochondrial homeostasis, J Cell Biol 190 391405 .
  31. 31. PutneyJ. W.Jr BirdG. S. 1993 The signal for capacitative calcium entry, Cell 75 199201 .
  32. 32. ParekhA. B.PennerR. 1997 Store depletion and calcium influx, Physiol Rev 77 901930 .
  33. 33. BerridgeM. J.BootmanM. D.LippP. 1998 Calcium--a life and death signal, Nature 395 645648 .
  34. 34. HirataK.PuslT.O’NeillA. F.DranoffJ. A.NathansonM. H. 2002 The type II inositol 1 4 5 -trisphosphate receptor can trigger Ca2+ waves in rat hepatocytes, Gastroenterology 122, 1088-1100.
  35. 35. PintonP.GiorgiC.SivieroR.ZecchiniE.RizzutoR. 2008 Calcium and apoptosis: ER-mitochondria Ca2+ transfer in the control of apoptosis, Oncogene 27 64076418 .
  36. 36. AlbanoE. 2006 Alcohol, oxidative stress and free radical damage, Proc Nutr Soc 65 278290 .
  37. 37. PacherP.HajnoczkyG. 2001 Propagation of the apoptotic signal by mitochondrial waves, EMBO J 20 41074121 .
  38. 38. KingA. L.SwainT. M.DickinsonD. A.LesortM. J.BaileyS. M. Chronic ethanol consumption enhances sensitivity to Ca(2+)-mediated opening of the mitochondrial permeability transition pore and increases cyclophilin D in liver, Am J Physiol Gastrointest Liver Physiol 299 G954966 .
  39. 39. YanM.ZhuP.LiuH. M.ZhangH. T.LiuL. 2007 Ethanol induced mitochondria injury and permeability transition pore opening: role of mitochondria in alcoholic liver disease, World J Gastroenterol 13 23522356 .
  40. 40. ArteelG. E. 2003 Oxidants and antioxidants in alcohol-induced liver disease, Gastroenterology 124 778790 .
  41. 41. ChenY. L.ChenL. J.BairM. J.YaoM. L.PengH. C.YangS. S.YangS. C. Antioxidative status of patients with alcoholic liver disease in southeastern Taiwan, World J Gastroenterol 17 10631070 .
  42. 42. BaileyS. M.LandarA.Darley-UsmarV. 2005 Mitochondrial proteomics in free radical research, Free Radic Biol Med 38 175188 .
  43. 43. D’AutreauxB.ToledanoM. B. 2007 ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis, Nat Rev Mol Cell Biol 8 813824 .
  44. 44. WuD.CederbaumA. I. 2009 Oxidative stress and alcoholic liver disease, Semin Liver Dis 29 141154 .
  45. 45. KingA. L.SwainT. M.DickinsonD. A.LesortM. J.BaileyS. M. 2010 Chronic ethanol consumption enhances sensitivity to Ca(2+)-mediated opening of the mitochondrial permeability transition pore and increases cyclophilin D in liver, Am J Physiol Gastrointest Liver Physiol 299, G954966 .
  46. 46. CsordasG.VarnaiP.GolenarT.RoyS.PurkinsG.SchneiderT. G.BallaT.HajnoczkyG. Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface, Mol Cell 39 121132 121132
  47. 47. BerridgeM. J.BootmanM. D.RoderickH. L. 2003 Calcium signalling: dynamics, homeostasis and remodelling, Nat Rev Mol Cell Biol 4 517529 .
  48. 48. HajnoczkyG.CsordasG.MadeshM.PacherP. 2000 The machinery of local Ca2+ signalling between sarco-endoplasmic reticulum and mitochondria, J Physiol 529 Pt 1 6981 .
  49. 49. ShiY.InoueS.ShinozakiR.FukueK.KougoT. 1998 Release of cytokines from human umbilical vein endothelial cells treated with platinum compounds in vitro, Jpn J Cancer Res 89 757767 .
  50. 50. HawkinsB. J.SoltL. A.ChowdhuryI.KaziA. S.AbidM. R.AirdW. C.MayM. J.FoskettJ. K.MadeshM. 2007 G protein-coupled receptor Ca2+-linked mitochondrial reactive oxygen species are essential for endothelial/leukocyte adherence, Mol Cell Biol 27 75827593 .
  51. 51. HawkinsB. J.SoltL. A.ChowdhuryI.KaziA. S.AbidM. R.AirdW. C.MayM. J.FoskettJ. K.MadeshM. 2007 G protein-coupled receptor Ca2+-linked mitochondrial reactive oxygen species are essential for endothelial/leukocyte adherence, Mol Cell Biol 27 75827593 .
  52. 52. SmailiS. S.HsuY. T.CarvalhoA. C.RosenstockT. R.SharpeJ. C.YouleR. J. 2003 Mitochondria, calcium and pro-apoptotic proteins as mediators in cell death signaling, Braz J Med Biol Res 36 183190 .
  53. 53. GreenD. R.KroemerG. 2004 The pathophysiology of mitochondrial cell death, Science 305 626629 .
  54. 54. CohenJ. I.ChenX.NagyL. E. Redox signaling and the innate immune system in alcoholic liver disease, Antioxid Redox Signal 15 523534 523534
  55. 55. ZamzamiN.MarchettiP.CastedoM.DecaudinD.MachoA.HirschT.SusinS. A.PetitP. X.MignotteB.KroemerG. 1995 Sequential reduction of mitochondrial transmembrane potential and generation of reactive oxygen species in early programmed cell death, J Exp Med 182 367377 .
  56. 56. ZamzamiN.MarchettiP.CastedoM.ZaninC.VayssiereJ. L.PetitP. X.KroemerG. 1995 Reduction in mitochondrial potential constitutes an early irreversible step of programmed lymphocyte death in vivo, J Exp Med 181 16611672 .
  57. 57. KimH.Rafiuddin-ShahM.TuH. C.JeffersJ. R.ZambettiG. P.HsiehJ. J.ChengE. H. 2006 Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies, Nat Cell Biol 8 13481358 .
  58. 58. WeiM. C.LindstenT.MoothaV. K.WeilerS.GrossA.AshiyaM.ThompsonC. B.KorsmeyerS. J. 2000 tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c, Genes Dev 14 20602071 .

Written By

Harish Chinna Konda Chandramoorthy, Karthik Mallilankaraman and Muniswamy Madesh

Submitted: 15 March 2011 Published: 11 January 2012