Nitric Oxide Inhibits Apoptosis by Preventing Increases in Caspase-3-like Activity via Two Distinct Mechanisms*

Nitric oxide (NO) has emerged as an important endogenous inhibitor of apoptosis, and here we report that NO prevents hepatocyte apoptosis initiated by the removal of growth factors or exposure to TNFα or anti-Fas antibody. We postulated that the mechanism of the inhibition of apoptosis by NO would include an effect on caspase-3-like protease activity. Caspase-3-like activity increased coincident with apoptosis due to all three stimuli, and treatment with the caspase-3-like protease inhibitorN-acetyl-Asp-Glu-Val-Asp-aldehyde inhibited both proteolytic activity and apoptosis. Endogenous or exogenous sources of NO prevented the increase in caspase-3-like activity in hepatocytes. Exposure of purified recombinant caspase-3 to an NO or NO+donor inhibited proteolytic activity. Dithiothreitol (DTT), but not glutathione, reversed the inhibition of recombinant caspase-3 by NO. When lysates from cells stimulated to express inducible NO synthase or cells exposed to NO donors were incubated in DTT, caspase-3-like activity increased to about 55% of cells not exposed to a source of NO. Similarly, administration of an NO donor to rats treated with TNFα and d-galactosamine also prevented the increase in caspase-3-like activity as measured in liver homogenates. The effect of the NO donor was reversed by about 50% if the homogenate was incubated with DTT. TNFα-induced apoptosis and caspase-3-like activity were also reduced in cultured hepatocytes exposed to 8-bromo-cGMP, and both effects were inhibited by the cGMP-dependent kinase inhibitor KT5823. The suppression in caspase-3-like activity in hepatocytes exposed to an NO donor was partially blocked by an inhibitor of soluble guanylyl cyclase, 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one, (ODQ), while the incubation of these lysates in DTT almost completely restored caspase-3-like activity to the level of TNFα-treated controls. These data indicate that NO prevents apoptosis in hepatocytes by either directly or indirectly inhibiting caspase-3-like activation via a cGMP-dependent mechanism and by direct inhibition of caspase-3-like activity through proteinS-nitrosylation.

[1]  Simon C Watkins,et al.  Targeting nitric oxide (NO) delivery in vivo. Design of a liver-selective NO donor prodrug that blocks tumor necrosis factor-alpha-induced apoptosis and toxicity in the liver. , 1997, Journal of medicinal chemistry.

[2]  M. Nehls,et al.  Suppression of Apoptosis by Nitric Oxide via Inhibition of Interleukin-1β–converting Enzyme (ICE)-like and Cysteine Protease Protein (CPP)-32–like Proteases , 1997, The Journal of experimental medicine.

[3]  T. Tsuruo,et al.  c-Jun NH2-terminal Kinase-mediated Activation of Interleukin-1β Converting Enzyme/CED-3-like Protease during Anticancer Drug-induced Apoptosis* , 1997, The Journal of Biological Chemistry.

[4]  Simon C Watkins,et al.  Nitric Oxide Protects Cultured Rat Hepatocytes from Tumor Necrosis Factor-α-induced Apoptosis by Inducing Heat Shock Protein 70 Expression* , 1997, The Journal of Biological Chemistry.

[5]  David S. Park,et al.  Induction of CPP32-like Activity in PC12 Cells by Withdrawal of Trophic Support , 1996, The Journal of Biological Chemistry.

[6]  Keisuke Kuida,et al.  Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice , 1996, Nature.

[7]  J. Lotem,et al.  Differential suppression by protease inhibitors and cytokines of apoptosis induced by wild-type p53 and cytotoxic agents. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[8]  Junying Yuan,et al.  Human ICE/CED-3 Protease Nomenclature , 1996, Cell.

[9]  R. Virmani,et al.  Apoptosis in myocytes in end-stage heart failure. , 1996, The New England journal of medicine.

[10]  Alfred Hausladen,et al.  Nitrosative Stress: Activation of the Transcription Factor OxyR , 1996, Cell.

[11]  M. Raff,et al.  Size Control: The Regulation of Cell Numbers in Animal Development , 1996, Cell.

[12]  M. Díaz-Guerra,et al.  Bacterial lipopolysaccharide antagonizes transforming growth factor β1–induced apoptosis in primary cultures of hepatocytes , 1996, Hepatology.

[13]  N. Thornberry,et al.  Apopain/CPP32 cleaves proteins that are essential for cellular repair: a fundamental principle of apoptotic death , 1996, The Journal of experimental medicine.

[14]  S. Nagata,et al.  Sequential activation of ICE-like and CPP32-like proteases during Fas-mediated apoptosis , 1996, Nature.

[15]  T. Dawson,et al.  Resistance to neurotoxicity in cortical cultures from neuronal nitric oxide synthase-deficient mice , 1996, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[16]  K. Abe,et al.  Differential localization of allograft nitric oxide synthesis: comparison of liver and heart transplantation in the rat model , 1996, Immunology.

[17]  J. Stamler,et al.  S-nitrosohaemoglobin: a dynamic activity of blood involved in vascular control , 1996, Nature.

[18]  C. Harris,et al.  Nitric oxide-induced p53 accumulation and regulation of inducible nitric oxide synthase expression by wild-type p53. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[19]  B. Brüne,et al.  Nitric oxide (NO) in apoptotic versus necrotic RAW 264.7 macrophage cell death: the role of NO-donor exposure, NAD+ content, and p53 accumulation. , 1996, Archives of biochemistry and biophysics.

[20]  J. Stamler,et al.  Posttranslational Modification of Glyceraldehyde-3-phosphate Dehydrogenase by S-Nitrosylation and Subsequent NADH Attachment (*) , 1996, The Journal of Biological Chemistry.

[21]  J. Mankovich,et al.  Protease Activity of in Vitro Transcribed and Translated Caenorhabditis elegans Cell Death Gene (ced-3) Product (*) , 1996, The Journal of Biological Chemistry.

[22]  R. Ientile,et al.  Nitric oxide synthase in chick embryo retina during development , 1996, FEBS letters.

[23]  A. Chinnaiyan,et al.  ICE-LAP3, a Novel Mammalian Homologue of the Caenorhabditis elegans Cell Death Protein Ced-3 Is Activated during Fas- and Tumor Necrosis Factor-induced Apoptosis (*) , 1996, The Journal of Biological Chemistry.

[24]  M. Boese,et al.  S-Nitrosation of Serum Albumin by Dinitrosyl-Iron Complex (*) , 1995, The Journal of Biological Chemistry.

[25]  E. Sonnenblick,et al.  Stretch-induced programmed myocyte cell death. , 1995, The Journal of clinical investigation.

[26]  J. Mankovich,et al.  Inhibition of ICE family proteases by baculovirus antiapoptotic protein p35. , 1995, Science.

[27]  D. Webb,et al.  α2-Macroglobulin Functions as a Cytokine Carrier to Induce Nitric Oxide Synthesis and Cause Nitric Oxide-dependent Cytotoxicity in the RAW 264.7 Macrophage Cell Line (*) , 1995, The Journal of Biological Chemistry.

[28]  V. Wahn,et al.  Nitric oxide induces apoptosis in mouse thymocytes. , 1995, Journal of immunology.

[29]  R. Mage,et al.  Preferential expansion and survival of B lymphocytes based on VH framework 1 and framework 3 expression: "positive" selection in appendix of normal and VH-mutant rabbits. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[30]  B. Spur,et al.  Peroxynitrite-induced Apoptosis in HL-60 Cells (*) , 1995, The Journal of Biological Chemistry.

[31]  B. Hoffman,et al.  Molecular controls of growth arrest and apoptosis: p53-dependent and independent pathways. , 1995, Oncogene.

[32]  Patrick R. Griffin,et al.  Identification and inhibition of the ICE/CED-3 protease necessary for mammalian apoptosis , 1995, Nature.

[33]  Muneesh Tewari,et al.  Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase , 1995, Cell.

[34]  C. Nathan,et al.  Altered responses to bacterial infection and endotoxic shock in mice lacking inducible nitric oxide synthase , 1995, Cell.

[35]  M. Leist,et al.  Tumor necrosis factor-induced hepatocyte apoptosis precedes liver failure in experimental murine shock models. , 1995, The American journal of pathology.

[36]  T. Billiar Nitric Oxide Novel Biology with Clinical Relevance , 1995, Annals of surgery.

[37]  C. Martínez-A,et al.  Splenic B lymphocyte programmed cell death is prevented by nitric oxide release through mechanisms involving sustained Bcl-2 levels. , 1995, The Journal of clinical investigation.

[38]  L. Dubertret,et al.  The nitric oxide donors, azide and hydroxylamine, inhibit the programmed cell death of cytokine‐deprived human eosinophils , 1995, FEBS letters.

[39]  H. Steller Mechanisms and genes of cellular suicide , 1995, Science.

[40]  C. Thompson,et al.  Apoptosis in the pathogenesis and treatment of disease , 1995, Science.

[41]  M. Tewari,et al.  Fas- and Tumor Necrosis Factor-induced Apoptosis Is Inhibited by the Poxvirus crmA Gene Product (*) , 1995, The Journal of Biological Chemistry.

[42]  K. Aldape,et al.  Expression of nitric oxide synthase in human central nervous system tumors. , 1995, Cancer research.

[43]  R. Eisenberg,et al.  Apoptosis abnormalities of splenic lymphocytes in autoimmune lpr and gld mice. , 1995, Journal of immunology.

[44]  J. Stamler,et al.  Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation , 1994, Cell.

[45]  D. Newmeyer,et al.  Cell-free apoptosis in Xenopus egg extracts: Inhibition by Bcl-2 and requirement for an organelle fraction enriched in mitochondria , 1994, Cell.

[46]  J. Stamler,et al.  Constitutive and inducible nitric oxide synthase gene expression, regulation, and activity in human lung epithelial cells. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[47]  J. Stamler,et al.  Redox signaling: Nitrosylation and related target interactions of nitric oxide , 1994, Cell.

[48]  S. Abramson,et al.  Nitric oxide reacts with intracellular glutathione and activates the hexose monophosphate shunt in human neutrophils: evidence for S-nitrosoglutathione as a bioactive intermediary. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[49]  W. A. Bradley,et al.  Superoxide and peroxynitrite in atherosclerosis. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[50]  Shai Shaham,et al.  The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1β-converting enzyme , 1993, Cell.

[51]  V. Laubach,et al.  Purification and cDNA sequence of an inducible nitric oxide synthase from a human tumor cell line. , 1993, Biochemistry.

[52]  Joseph Loscalzo,et al.  A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds , 1993, Nature.

[53]  L. J. McDonald,et al.  Stimulation by nitric oxide of an NAD linkage to glyceraldehyde-3-phosphate dehydrogenase. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[54]  J. Albina,et al.  Nitric oxide-mediated apoptosis in murine peritoneal macrophages. , 1993, Journal of immunology.

[55]  J. Lancaster,et al.  Nonheme iron-nitrosyl complex formation in rat hepatocytes: detection by electron paramagnetic resonance spectroscopy. , 1993, Archives of biochemistry and biophysics.

[56]  D. Vaux Toward an understanding of the molecular mechanisms of physiological cell death. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[57]  C. Lowenstein,et al.  Cytokines, endotoxin, and glucocorticoids regulate the expression of inducible nitric oxide synthase in hepatocytes. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[58]  D. Wink,et al.  Reactions of the bioregulatory agent nitric oxide in oxygenated aqueous media: determination of the kinetics for oxidation and nitrosation by intermediates generated in the NO/O2 reaction. , 1993, Chemical research in toxicology.

[59]  J. Stamler,et al.  S-nitrosylation of tissue-type plasminogen activator confers vasodilatory and antiplatelet properties on the enzyme. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[60]  S. Tannenbaum,et al.  DNA damage and mutation in human cells exposed to nitric oxide in vitro. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[61]  T. Billiar,et al.  Association between synthesis and release of cGMP and nitric oxide biosynthesis by hepatocytes. , 1992, The American journal of physiology.

[62]  J. Stamler,et al.  S-nitrosylation of proteins with nitric oxide: synthesis and characterization of biologically active compounds. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[63]  R. L. Simmons,et al.  Modulation of Nitrogen Oxide Synthesis In Vivo: NG‐Monomethyl‐L‐Arginine Inhibits Endotoxin‐Induced Nitrate/Nitrate Biosynthesis While Promoting Hepatic Damage , 1990, Journal of leukocyte biology.

[64]  P. Goering,et al.  Metallothionein and other cadmium-binding proteins: recent developments. , 1990, Chemical research in toxicology.

[65]  S. Tannenbaum,et al.  Nitrosation of amines by stimulated macrophages. , 1987, Carcinogenesis.

[66]  L. Ignarro,et al.  Hepatic cyclic GMP formation is regulated by similar factors that modulate activation of purified hepatic soluble guanylate cyclase. , 1987, The Journal of biological chemistry.

[67]  H. Horvitz,et al.  Genetic control of programmed cell death in the nematode C. elegans , 1986, Cell.

[68]  S. Tannenbaum,et al.  Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. , 1982, Analytical biochemistry.

[69]  B. Saville A scheme for the colorimetric determination of microgram amounts of thiols , 1958 .