Acute pancreatitis is an inflammatory disorder of exocrine

Similar documents
shehab Moh Tarek ... ManarHajeer

Mechanistic Toxicology

Cell Injury MECHANISMS OF CELL INJURY

Chemical and Biochemical Mechanism Of Cell Injury.

Introduction to pathology lecture 5/ Cell injury apoptosis. Dr H Awad 2017/18

Is there a common mechanism of DILI, do we need to know?

Correction. (originally cited in print as reference #219; now cited as references 195 and 222)

Apoptosis Chapter 9. Neelu Yadav PhD

In 1993, the International Autoimmune Hepatitis Group

Mechanisms of Cell Injury

Calcium Dysregulation in Acute & Chronic Pancreatitis

#19 Apoptosis Chapter 9. Neelu Yadav PhD

Molecular and Cellular Changes in Acute Pancreatitis

Autoimmune Hepatitis. What Drug and for How Long? Hepatology Day May 30 th, 2015

Reperfusion Injury: How Can We Reduce It?

PUBLICATIONS. cells. J. Physiol. (London) 517P:91P (Manchester, England, UK).

Autoimmune Hepatitis in Clinical Practice

Neuronal Death After Hemorrhagic Stroke In Vitro and In Vivo Shares Features of Ferroptosis and Necroptosis

Comparing Efficacy Between Regimens in the Initial Treatment of Autoimmune Hepatitis

Inflammation plays a key role in tissue repair by eliminating

Lecture 14 - The cell cycle and cell death

hydrolyzes ATP to exchange 3 Na + in for 2 K + out generate the transcellular Na + and K + gradients provides the electrochemical gradient

Under the Radar Screen: How Bugs Trick Our Immune Defenses

Cellular Injury, Necrosis, Apoptosis

Subject Index. Bcl-2, apoptosis regulation Bone marrow, polymorphonuclear neutrophil release 24, 26

Long-term Outcomes of Patients With Autoimmune Hepatitis Managed at a Nontransplant Center

CASE 1 Plasma Cell Infiltrates: Significance in post liver transplantation and in chronic liver disease

CELL INJURY. Severity of Cell Injury

Supporting Information

Fat, ballooning, plasma cells and a +ANA. Yikes! USCAP 2016 Evening Specialty Conference Cynthia Guy

The liver in poisoning: what can we learn from animal models?

INTRODUCTION. Liver is the main organ responsible for the major metabolic and secretory functions in the

Anti-Apoptotic Effects of Cellular Therapy

Treatment options for autoimmune hepatitis: A systematic review of randomized controlled trials

Mechanisms of Cell Injury: Loss of Calcium Homeostasis

budesonide, 3mg, gastro-resistant capsules (Budenofalk ) SMC No. (1043/15) Dr Falk Pharma UK Ltd

Annals of RSCB Vol. XVI, Issue 1

NASH Bench to Bedside

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

REGULATION OF ENZYME ACTIVITY. Medical Biochemistry, Lecture 25

#19 Apoptosis Chapter 9. Neelu Yadav PhD

A Single Center Review of the Use of Mycophenolate Mofetil in the Treatment of Autoimmune Hepatitis

AperTO - Archivio Istituzionale Open Access dell'università di Torino

Alcoholic Hepatitis. Christian Doppler Research Laboratory for Gut Inflammation Medical University Innsbruck. Herbert Tilg

Impact factor: Reporter:4A1H0019 Chen Zi Hao 4A1H0023 Huang Wan ting 4A1H0039 Sue Yi Zhu 4A1H0070 Lin Guan cheng 4A1H0077 Chen Bo xuan

Akihiro Okabe, Masahiko Hirota, Yu Kimura, Keisuke Maeda, Michio Ogawa. Department of Surgery II, Kumamoto University Medical School.

Molecular biology :- Cancer genetics lecture 11

6. TNF-α regulates oxidative stress, mitochondrial function and autophagy in neuronal cells

Ultrastructural aspects of acute pancreatitis induced by 2, 2 -azobis (2-amidinopropane) dihydrochloride (AAPH) in rats

GMS 6644: Apoptosis. Introduction

ORIGINAL ARTICLES LIVER, PANCREAS, AND BILIARY TRACT

Autoimmune hepatitis

Pathophysiology and treatment of focal cerebral ischemia

Cell Quality Control. Peter Takizawa Department of Cell Biology

This student paper was written as an assignment in the graduate course

) one consumes in breathing is converted to:, which of the following would be found in the oxidized state?

Overview of PSC Making the Diagnosis

Apoptosis in chronic hepatitis C

Apoptotic Pathways in Mammals Dr. Douglas R. Green

Disclosures. Pathogenesis of Autoimmunity Normal immune response: 11/5/2011. Methotrexate and JUN Pathway Activation in Rheumatoid Arthritis

CLINICAL ADVANCES IN LIVER, PANCREAS, AND BILIARY TRACT

Focus Application. Compound-Induced Cytotoxicity

Role of trypsinogen activation in genesis of pancreatitis

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Prediction of invasiveness of hepatic tumor cells

Diagnostic Criteria for Autoimmune Hepatitis : Historical Review and Present Problems

150 mm HCO How Does the Pancreas Do It? Clues from Computer Modelling of the Duct Cell

Muse Assays for Cell Analysis

Dietary supplementation in treating non-alcoholic fatty liver disease Dr. Ahmad Saedi Associate Professor School of Nutritional Sciences and

Cellular responses to stress

Kinase Inhibitor p21 WAF1/CIP1 in Apoptosis and Autophagy

Pancreas Fox Chapter 18 part 2 (also Chapter 19.3 & 19.4)

Cell Adaptation, Cell Injury and Cell Death

Reactive oxygen species: Importance for ischemia/reperfusion (injury)

TITLE: Targeting trypsin-inflammation axis for pancreatitis therapy in a humanized pancreatitis model. Los Angeles, CA 90048

THE PATHOLOGICAL ROLE OF BCL-2 FAMILY PROTEINS AND THE PROTECTIVE ROLE OF CALCIUM REGULATOR CALMODULIN IN PANCREATIC ACINAR CELLS

Cell morphology. Cell organelles structure and function. Chapter 1: UNIT 1. Dr. Charushila Rukadikar

The most common nonmalignant gastrointestinal. Editorials, continued

News. Laboratory. CHANGES IN CARDIAC TROPONIN I REPORTING Gene Shaw, MD, Annu Khajuria, PhD. Inside This Issue

PREPARED BY P.DHARANI PRASAD II YEAR B.PHARM II SEM SUB:PATHOPHYSIOLOGY

SUPPLEMENTARY INFORMATION In format provided by JAATTELA (NOVEMBER 2005)

Role of metabolism in Drug-Induced Liver Injury (DILI) Drug Metab Rev. 2007;39(1):

2. List routes of exposure in the order of most rapid response.

Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bakindependent death of cancer cells

PBC features and management in the era of UDCA and Budesonide

Hangzhou, 15 March Ulrich Beuers Department of Gastroenterology and Hepatology Academic Medical Center University of Amsterdam

Hepatitis C wi w t i h Ju J dy y W y W a y t a t t

Nonalcoholic Fatty Liver Disease in Children: Typical and Atypical

Therapeutical potential of mitochondrial reactive oxygen species in carcinogenesis

renoprotection therapy goals 208, 209

Mitochondria in apoptosis. Jean-Claude Martinou, MD, Ph.D Department of cell biology University of Geneva Geneva, Switzerland

Ameliorating Reperfusion Injury During Resuscitation from Cardiac Arrest

Autoimmune hepatitis. Recent advances. Dominique-Charles VALLA

University of Milan spin-off founded in 2004 and now fully owned by Genextra S.p.A.

METABOLIC SYNDROME AND HCV: FROM HCV

Pro-Oxidant Environmental Exposures: Implications of Redox Imbalance in Autism S. Jill James, Ph.D.

THE CLINICAL course of severe

Identifying Metabolic Phenotype Switches in Cancer Cells Using the Agilent Seahorse XF Analyzer in an Hypoxic Environment

Supplementary Figures

4. Lysosomes, Smooth Endoplasmic Reticulum, Mitochondria, and Inclusions

Transcription:

and management of autoimmune hepatitis. Hepatology 2010;51:2193 2213. 5. Soloway RD, Summerskill WH, Baggenstoss AH, et al. Clinical, biochemical, and histological remission of severe chronic active liver disease: a controlled study of treatments and early prognosis. Gastroenterology 1972;63:820 833. 6. Murray-Lyon IM, Stern RB, Williams R. Controlled trial of prednisone and azathioprine in active chronic hepatitis. Lancet 1973; 1:735 737. 7. Cook GC, Mulligan R, Sherlock S. Controlled prospective trial of corticosteroid therapy in active chronic hepatitis. Q J Med 1971; 40:159 185. 8. Kirk AP, Jain S, Pocock S, et al. Late results of the Royal Free Hospital prospective controlled trial of prednisolone therapy in hepatitis B surface antigen negative chronic active hepatitis. Gut 1980;21:78 83. 9. Alvarez F, Berg PA, Bianchi FB, et al. International Autoimmune Hepatitis Group Report: review of criteria for diagnosis of autoimmune hepatitis. J Hepatol 1999;31:929 938. 10. Hennes EM, Zeniya M, Czaja AJ, et al. Simplified criteria for the diagnosis of autoimmune hepatitis. Hepatology 2008;48:169 76. 11. Hoeroldt B, McFarlane E, Dube A, et al. Long-term outcomes of patients with autoimmune hepatitis managed at a non-transplant center. Gastroenterology 2011;140:1980 1989. 12. Feld JJ, Dinh H, Arenovich T, et al. Autoimmune hepatitis: effect of symptoms and cirrhosis on natural history and outcome. Hepatology 2005;42:53 62. 13. Czaja AJ, Freese DK. Diagnosis and treatment of autoimmune hepatitis. Hepatology 2002;36:479 497. 14. Muratori P, Granito A, Quarneti C, et al. Autoimmune hepatitis in Italy: the Bologna experience. J Hepatol 2009;50:1210 1218. 15. Manns MP, Woynarowski M, Kreisel W, et al. Budesonide induces remission more effectively than prednisone in a controlled trial of patients with autoimmune hepatitis. Gastroenterology 2010;139:1198 1206. 16. Kanzler S, Lohr H, Gerken G, et al. Long-term management and prognosis of autoimmune hepatitis (AIH): a single center experience. Z Gastroenterol 2001;39:339 341, 344 348. 17. Czaja AJ, Manns MP. The validity and importance of subtypes in autoimmune hepatitis: a point of view. Am J Gastroenterol 1995; 90:1206 1211. 18. Czaja AJ. Genetic factors affecting the occurrence, clinical phenotype, and outcome of autoimmune hepatitis. Clin Gastroenterol Hepatol 2008;6:379 388. 19. Muratori L, Muratori P, Lanzoni G, et al. Application of the 2010 American Association for the study of liver diseases criteria of remission to a cohort of Italian patients with autoimmune hepatitis. Hepatology 2010; 52: 1857 1858. Reprint requests Address requests for reprints to: Michael P. Manns, MD, Professor and Chairman, Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany. e-mail: manns.michael@mh-hannover.de. Conflicts of interest The author discloses the following: Michael P. Manns is a consultant for and received grant support and lecture fees from Falk Pharma GmbH, Germany. 2011 by the AGA Institute 0016-5085/$36.00 doi:10.1053/j.gastro.2011.04.026 Which Way to Die: the Regulation of Acinar Cell Death in Pancreatitis by Mitochondria, Calcium, and Reactive Oxygen Species See Reactive oxygen species induced by bile acid induce apoptosis and protect against necrosis in pancreatic acinar cells, by Booth DM, Murphy JA, Mukherjee R, et al, on page 2116. Acute pancreatitis is an inflammatory disorder of exocrine pancreas, which carries considerable morbidity and mortality; its pathophysiology remains elusive. 1 During the past decade, significant progress has been achieved in our understanding of the inflammatory response in pancreatitis. 1 Much less is known about the mechanisms mediating another key pathologic response of pancreatitis, namely acinar cell death. In human disease and experimental pancreatitis, acinar cells die through both apoptosis and necrosis. These 2 main types of cell death differ morphologically and biochemically. 2,3 Key molecular steps in the apoptotic pathway are the release of cytochrome c from mitochondria and activation of caspases, a specific class of cysteine proteases. Importantly, apoptosis preserves the plasma membrane integrity, whereas necrotic cell releases its constituents, damaging neighboring cells and promoting inflammation. Therefore, necrotic death is deadlier to the organism than apoptotic death. 2,3 Parenchymal necrosis is a major complication of pancreatitis, and greater amounts of necrosis are associated with a worse prognosis in the disease. 1 Intriguingly, the severity of pancreatitis in animal models correlates directly with the extent of necrosis and inversely with apoptosis. 4 8 Furthermore, stimulating acinar cell apoptosis in experimental models decreased necrosis and the severity of pancreatitis, whereas inhibition of acinar cell apoptosis (eg, with caspase inhibitors 7 ) poten- 1876

Figure 1. Pathways mediating acinar cell death in models of pancreatitis induced by supramaximal CCK-8 or the bile acid TLC-S through Ca 2 (left panel) and ROS (right panel). Dashed lines indicate pathways that are likely but not yet experimentally proven in acinar cells. tiated necrosis and worsened pancreatitis. Thus, shifting the pattern of death responses of pancreatitis toward apoptosis and away from necrosis may have a therapeutic value. Mitochondria play a central role in regulating cell death because mitochondrial membrane permeabilization is a universal trigger of both necrosis and apoptosis. 2,3 Permeabilization results in loss of the mitochondrial membrane potential, m, through opening of the permeability transition pore (PTP), a nonselective channel permeating both outer and inner mitochondrial membranes. 3 Loss of m leads to adenosine triphosphate (ATP) depletion, inability to maintain ionic gradients across the plasma membrane, and ultimately necrosis. Mitochondrial permeabilization also triggers the apoptotic pathway through release of the mitochondria resident protein cytochrome c (likely, not via PTP 3 ). Once in the cytosol, cytochrome c interacts with and activates caspases, leading to the downstream apoptotic events. 2,3 Mitochondria are involved in cellular Ca 2 homeostasis and are a major source of reactive oxygen species (ROS). Further, Ca 2 and ROS critically regulate mitochondrial function (and dysfunction); in particular, elevations of cytosolic Ca 2 or ROS stimulate PTP opening. 3 Ca 2 is a major regulator of the acinar cell secretory function, but abnormal ( global and sustained) increase in cytosolic Ca 2 is a key pathologic signal associated with pancreatitis. 9 Oxidative stress is also implicated in the pathogenesis of pancreatitis 10 ; however, the sources of ROS in the acinar cell, and the exact roles and targets of ROS in pancreatitis, are poorly understood. Until recently, 7,8,11 16 little was known about the properties of pancreatic mitochondria, their regulation by Ca 2 and ROS, and the roles of mitochondria, Ca 2, and ROS in acinar cell death. The study by Booth et al in this issue of GASTROENTEROLOGY 17 advances further our knowledge of these regulations, especially the mechanisms involving ROS. This study applied an ex vivo model of acinar cell injury induced by bile acid (taurolithocholic acid salt [TLC-S]), which corresponds to animal (rodent) models of pancreatitis induced by administration of bile acids. 1,18 The bile acid models are clinically relevant, because gallstones are a major cause of acute pancreatitis. The previous data 7,8,11 14 were largely obtained in rodent and ex vivo models of pancreatitis induced by supramaximal CCK-8 or its analog cerulein, the most widely used and well-characterized experimental system to study acute pancreatitis 1,18 Both bile acid- and CCK/cerulein-induced pancreatitis is associated with acinar cell necrosis and apoptosis. The recent findings in these 2 dissimilar models, particularly the study by Booth et al, 17 reveal common mechanisms through which mitochondria, Ca 2, and ROS regulate acinar cell death (Figure 1). First, the results 7,8,11 13,16,17,19 provide convincing evidence that abnormal Ca 2 signal promotes acinar cell necrosis through mitochondrial depolarization and subsequent ATP drop. Both the supramaximal CCK/cerulein and TLC-S cause Ca 2 -dependent loss of m. Experiments on isolated pancreatic mitochondria showed that Ca 2 directly depolarizes the mitochondria through PTP opening. 13 Moreover, it seems that pancreatic mitochondria are highly sensitive to Ca 2 -induced depolarization 13 1877

(as compared, for example, with the classical liver mitochondria) and lose m even at submicromolar Ca 2 owing to a greater sensitivity of the pancreatic mitochondria PTP to Ca 2. CCK-induced mitochondrial depolarization significantly decreases ATP level in acinar cells and leads to necrosis. 8,14 Furthermore, Booth et al 17 now show that TLC-S induced acinar cell necrosis can be reversed by patched ATP supplementation. Evidence for a key role of mitochondrial depolarization in acinar cell necrosis also comes from a study 8 that showed that the prosurvival Bcl-xL and Bcl-2 proteins (known to stabilize mitochondria 2,3 ) protect acinar cells from necrosis and counteract the loss of m induced by the supramaximal CCK/cerulein. For example, Bcl-xL/Bcl-2 inhibitors or Bcl-xL small interfering RNA depolarized isolated pancreatic mitochondria and markedly decreased ATP level in acinar cells, resulting in necrosis. 8 Second, the findings illuminate the role of ROS in acinar cell death. Both the supramaximal CCK-8 and TLC-S increase the mitochondrial ([ROS]m) as well as intracellular (total) ROS ([ROS]i) levels in acinar cells. 13,17 However, the data from Booth et al 17 indicate that neither [ROS]m nor [ROS]i mediate TLC-S induced acinar cells necrosis. Moreover, the data suggest that ROS provide protection from TLC-S induced necrosis, as necrosis was potentiated by the antioxidant N-acetylcysteine and was decreased by elevating acinar cell [ROS]i. 17 This is different from many other cell types, for example, hepatocytes, in which ROS play the pro-necrotic role, so that antioxidants (eg, the same N-acetylcysteine) inhibit necrosis, whereas increasing ROS level (eg, by applying exogenous H 2 O 2 ) facilitates necrosis. 20,21 Why pancreatic acinar cells behave so differently is not clear; one reason could be that pancreatic mitochondria are less prone to ROS-induced depolarization, 13 a pathway leading to necrosis. Indeed, exogenous H 2 O 2 did not depolarize isolated pancreatic mitochondria, 13 differently from liver 22 and kidney 23 mitochondria. Thus, in contrast with abnormal Ca 2 signal, ROS do not mediate, and may even protect from, necrosis in pancreatitis (at least in the ex vivo models). At the same time, the results show that ROS, and in particular mitochondrial ROS, mediate acinar cell apoptosis in pancreatitis. Both CCK and TLC-S induced apoptosis was inhibited by blocking mitochondrial ROS production. 13,17 Conversely, increasing [ROS]m with rotenone, 13 or [ROS]i with the pro-oxidant menadione 15,17 stimulated acinar cell apoptosis. TLC-S induced caspase activation was inhibited by N-acetylcysteine and potentiated by elevating acinar cell [ROS]i. 17 A key effect whereby mitochondrial ROS promote apoptosis in acinar cells (similar to other cells 24 ) is through stimulating cytochrome c release. 13 The underlying mechanism is likely oxidation by ROS of cardiolipin, an anionic phospholipid that anchors cytochrome c to the inner mitochondrial membrane. Cardiolipin oxidation facilitates cytochrome c detachment from the inner membrane and, thus, its availability for release. 24 Finally, the findings in the bile acid and CCK models reveal complex regulation of acinar cell apoptosis by Ca 2, which involves Ca 2 effects on cytochrome c release, m, and ROS. Experiments on isolated pancreatic mitochondria 13 showed that Ca 2 has 2 opposing effects on cytochrome c release: Ca 2 per se stimulates cytochrome c release, whereas Ca 2 -induced mitochondrial depolarization inhibits cytochrome c release. In accord with the results on isolated mitochondria, increasing the cytosolic Ca 2 stimulated, whereas mitochondrial depolarization inhibited, cytochrome c release, caspase activation, and apoptosis in intact acinar cells. 12,13,16 One mechanism whereby mitochondrial depolarization in acinar cells inhibits cytochrome c release is by blocking mitochondrial ROS generation, which is driven by m. 13 Similarly, the data from Booth et al 17 indicate that decreasing the cytosolic Ca 2 does not inhibit TLC- S induced acinar cell apoptosis unless the mitochondrial ROS production is also blocked. Thus, the pattern of acinar cell death in pancreatitis is regulated at the mitochondrial level by interplay between Ca 2, m, and ROS. The schematic in Figure 1 illustrates the negative feedback regulations between mitochondrial signals mediating necrotic and apoptotic pathways in acinar cells. In particular, m loss caused by abnormal Ca 2 signal not only promotes necrosis but also inhibits apoptosis by limiting cytochrome c release (left panel). In addition, the ensuing decrease in cellular ATP limits caspase activation. Thus, mitochondrial depolarization plays a role of fulcrum in the balance between apoptosis and necrosis. In the opposite direction, caspase activation in acinar cells not only mediates apoptosis but also inhibits necrosis (right panel), as shown, for example, by applying caspase inhibitors in cerulein pancreatitis. 7 One plausible mechanism 7 by which caspases inhibit necrosis is through cleavage, and hence inactivation, of the receptor-interacting protein kinases, important mediators of necrosis. 25,26 Genetic ablation of receptor-interacting protein kinase-3 greatly decreased necrosis in cerulein pancreatitis. 26 The pro-necrotic effect of receptor-interacting protein kinases is mediated likely via mitochondrial permeabilization. 25 Caspases may also limit acinar cell necrosis by preventing accumulation of dysfunctional mitochondria with low m. 4 A novel aspect of these regulations, uncovered by Booth et al, 17 is that ROS may protect acinar cells from necrosis. Although the authors do not speculate on the nature of this effect, it could be explained by stimulation 1878

of cytochrome c release by ROS, leading to caspase activation (Figure 1, right panel). The mutually negative regulations between necrosis and apoptosis in acinar cells are of general interest for understanding cell death responses. Specifically for pancreatitis, these negative feedbacks suggest a molecular mechanism underlying the puzzling inverse correlation between necrosis and apoptosis observed in experimental models of acute pancreatitis. 1,4 8 Some of these regulations are due to peculiar (or even unique) properties of pancreatic mitochondria. In pancreatic mitochondria, 13 Ca 2 overload causes a pronounced loss of m, but only limited amounts of cytochrome c are released, whereas ROS greatly stimulate cytochrome c release, but cause little depolarization. By contrast, in mitochondria from liver and other organs, Ca 2 and ROS stimulate both cytochrome c release and depolarization, resulting in parallel induction of both apoptosis and necrosis. 3 These differences could be due to non-classical properties of the pancreatic mitochondria PTP, that is, its high sensitivity to Ca 2 -induced depolarization and low sensitivity to ROS-induced depolarization. 13 The prediction from the above analysis is that a greater extent of mitochondrial depolarization, lower levels of acinar cell ROS, or lower Bcl-xL/Bcl-2 levels facilitate pancreatic necrosis and limit apoptosis, thus worsening the disease; in other words, these parameters could serve as a prognostic factor for a more severe, necrotizing pancreatitis. Thus, approaches aimed to inhibit PTP opening, maintain a higher level of mitochondrial ROS, or up-regulate Bcl-xL/Bcl-2 could prevent or attenuate necrosis in pancreatitis. The results from Booth et al 17 offer an explanation for the conflicting data on the effects of antioxidants in experimental pancreatitis and failure of antioxidant therapy in clinical trials. The authors emphasize the opposing roles of ROS produced by acinar and inflammatory cells in pancreatitis. Indeed, it was shown that neutrophils infiltrating the pancreas facilitate necrosis, 5,27,28 and, moreover, that ROS generated by neutrophils mediate pathologic responses of the disease. 27 Thus, rather than using broad-spectrum antioxidants, strategies to manipulate ROS in pancreatitis should be more targeted. Another interesting question addressed by Booth et al 17 is the role of autophagy in acinar cell death. The authors conclude that autophagy does not play a significant role, based in part on the absence of the effect of 3-methyladenine, a blocker of autophagosome formation, on TLC-S induced acinar cell death. However, an alternative explanation is that TLC-S may inhibit autophagy by impairing autolysosomal function downstream of autophagosomes, as was shown in other cells 29 (and similar to the action of CCK-8 in acinar cells 30 32 ); hence, little additional effect of 3-methyladenine on cell death. Investigation of autophagy in pancreatitis has only started, 30 34 and its role in acinar cell death requires more detailed studies. In general, the role of autophagy in cell death is a subject of intense research and much debate. 35 The predominant point of view is that efficient, physiologic autophagy is prosurvival, whereas defective, inefficient autophagy promotes cell death. Relevant to the study of Booth et al, 17 1 mechanism whereby defective autophagy stimulates cell death is through accumulation of uncoupled mitochondria overproducing ROS. 35 As with any good story, the study of Booth et al 17 provokes further questions. There is much more to be learned about the pathways of acinar cell death depicted in Figure 1. What are the mechanisms whereby TLC-S (as well as CCK-8) stimulates mitochondrial and cellular ROS? Is there a role for non-mitochondrial ROS sources in the acinar cell? How exactly does ROS protect acinar cells from necrosis? What properties of pancreatic mitochondria (ie, their PTP) make them more sensitive to Ca 2 -induced and less sensitive to ROS-induced depolarization? Do similar mechanisms operate in other models of pancreatitis, and importantly, in human disease (the data of Booth et al 17 on human acinar cells suggest so)? It should be also emphasized that Ca 2, m, and ROS are critical but not the sole regulators of cell death in pancreatitis. Other players include cathepsin B, PI 3-kinase, p53, and nuclear factor- B. 1 How these players act in the acinar cell death drama, within and outside pathways discussed here, remains to be determined. ANNA S. GUKOVSKAYA ILYA GUKOVSKY VA Greater Los Angeles Healthcare System and University of California Los Angeles Los Angeles, California References 1. Pandol SJ, Saluja AK, Imrie CW, et al. Acute pancreatitis: bench to the bedside. Gastroenterology 2007;132:1127 1151; erratum: ibid, 133:1056. 2. Galluzzi L, Maiuri MC, Vitale I, et al. Cell death modalities: classification and pathophysiological implications. Cell Death Differ 2007;14:1237 1243. 3. Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev 2007;87:99 163. 4. Gukovskaya AS, Perkins P, Zaninovic V, et al. Mechanisms of cell death after pancreatic duct obstruction in the opossum and the rat. Gastroenterology 1996;110:875 884. 5. Sandoval D, Gukovskaya AS, Reavey P, at al. The role of neutrophils and platelet-activating factor in mediating experimental pancreatitis. Gastroenterology 1996;111:1081 1091. 1879

6. Kaiser AM, Saluja AK, Sengupta A, et al. Relationship between severity, necrosis, and apoptosis in five models of experimental acute pancreatitis. Am J Physiol Cell Physiol 1995;269:C1295 C1304. 7. Mareninova OA, Sung K-F, Hong P, et al. Cell death in pancreatitis: caspases protect from necrotizing pancreatitis. J Biol Chem 2006;281:3370 3381. 8. Sung K-F, Odinokova IV, Mareninova OA, et al. Prosurvival Bcl-2 proteins stabilize pancreatic mitochondria and protect against necrosis in experimental pancreatitis. Exp Cell Res 2009;315: 1975 1989. 9. Criddle DN, McLaughlin E, Murphy JA, et al. The pancreas misled: signals to pancreatitis. Pancreatology 2007;7:436 446. 10. Leung PS, Chan YC. Role of oxidative stress in pancreatic inflammation. Antioxid Redox Signal 2009;11:135 165. 11. Gukovskaya AS, Gukovsky I, Jung Y, et al. Cholecystokinin induces caspase activation and mitochondrial dysfunction in pancreatic acinar cells: roles in cell injury processes of pancreatitis. J Biol Chem 2002;277:22595 22604. 12. Voronina SG, Barrow SL, Gerasimenko OV, et al. Bile acids induce a cationic current, depolarizing pancreatic acinar cells and increasing the intracellular Na concentration. J Biol Chem 2004;279:27327 27338. 13. Odinokova IV, Sung K-F, Mareninova OA, et al. Mechanisms regulating cytochrome c release in pancreatic mitochondria. Gut 2009;58:431 442. 14. Voronina SG, Barrow SL, Simpson AW, et al. Dynamic changes in cytosolic and mitochondrial ATP levels in pancreatic acinar cells. Gastroenterology 2010;138:1976 1987. 15. Baumgartner HK, Gerasimenko JV, Thorne C, et al. Caspase-8- mediated apoptosis induced by oxidative stress is independent of the intrinsic pathway and dependent on cathepsins. Am J Physiol Gastrointest Liver Physiol 2007;293:G296 G307. 16. Baumgartner HK, Gerasimenko JV, Thorne C, et al. Calcium elevation in mitochondria is the main Ca2 requirement for mitochondrial permeability transition pore (mptp) opening. J Biol Chem 2009;284:20796 20803. 17. Booth DM, Murphy JA, Mukherjee R, et al. Reactive oxygen species induced by bile acid induce apoptosis and protect against necrosis in pancreatic acinar cells. Gastroenterology 2011;140: 2116 2125. 18. Lerch MM, Adler G. Experimental animal models of acute pancreatitis. Int J Pancreatol 1994;15:159 170. 19. Schild L, Matthias R, Stanarius A, et al. Induction of permeability transition in pancreatic mitochondria by cerulein in rats. Mol Cell Biochem 1999;195:191 197. 20. Bhogal RH, Curbishley SM, Weston CJ, et al. Reactive oxygen species mediate human hepatocyte injury during hypoxia/reoxygenation. Liver Transpl 2010;16:1303 1313. 21. Conde de la Rosa L, Schoemaker MH, Vrenken TE, et al. Superoxide anions and hydrogen peroxide induce hepatocyte death by different mechanisms: involvement of JNK and ERK MAP kinases. J Hepatol 2006;44:918 929. 22. Nakagawa T, Shimizu S, Watanabe T, et al. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 2005;434:652 658. 23. Devalaraja-Narashimha K, Diener AM, Padanilam BJ. Cyclophilin D gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 2009;297:F749 F759. 24. Ott M, Gogvadze V, Orrenius S, et al. Mitochondria, oxidative stress and cell death. Apoptosis 2007;12:913 922. 25. Galluzzi L, Kepp O, Kroemer G. RIP kinases initiate programmed necrosis. J Mol Cell Biol 2009;1:8 10. 26. He S, Wang L, Miao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009; 137:1100 1111. 27. Gukovskaya AS, Vaquero E, Zaninovic V, et al. Neutrophils and NADPH oxidase mediate intrapancreatic trypsin activation in murine experimental acute pancreatitis. Gastroenterology 2002; 122:974 984. 28. Gukovskaya AS, Gukovsky I, Zaninovic V, et al. Pancreatic acinar cell produce, release, and respond to tumor necrosis factor- : role in regulating cell death and pancreatitis. J Clin Invest 1997; 100:1853 1862. 29. Larocca MC, Pellegrino JM, Rodriguez Garay EA, et al. Taurocholate-induced inhibition of hepatic lysosomal degradation of horseradish peroxidase. Biochim Biophys Acta 1999;1428:341 347. 30. Mareninova OA, Hermann K, French SW, et al. Impaired autophagic flux mediates acinar cell vacuole formation and trypsinogen activation in rodent models of acute pancreatitis. J Clin Invest 2009;119:3340 3355. 31. Gukovsky I, Gukovskaya AS. Impaired autophagy underlies key pathological responses of acute pancreatitis. Autophagy 2010; 6:428 429. 32. Gukovsky I, Pandol SJ, Gukovskaya AS. Organellar dysfunction in the pathogenesis of pancreatitis. Antioxid Redox Signal 2011, in press. 33. Hashimoto D, Ohmuraya M, Hirota M, et al. Involvement of autophagy in trypsinogen activation within the pancreatic acinar cells. J Cell Biol 2008;181:1065 1072. 34. Grasso D, Ropolo A, Lo Ré A, et al. Zymophagy, a novel selective autophagy pathway mediated by vmp1-usp9x-p62, prevents pancreatic cell death. J Biol Chem 2011;286:8308 8324. 35. Galluzzi L, Morselli E, Kepp O, et al. Mitochondrial gateways to cancer. Mol Aspects Med 2010;31:1 20. Reprint requests Address requests for reprints to: Anna S. Gukovskaya, PhD, West Los Angeles Healthcare Center, 11301 Wilshire Blvd, Blg.258, Rm.340, Los Angeles, California 90073. e-mail: agukovsk@ucla. edu. Conflicts of interest The authors disclose no conflicts. Funding Department of Veterans Affairs; NIH DK059936 and AA019730; Southern California Research Center for Alcoholic Liver and Pancreatic Diseases (NIH P50AA11999). 2011 by the AGA Institute 0016-5085/$36.00 doi:10.1053/j.gastro.2011.04.025 1880