NEUROVASCULAR MEDICINE - Pursuing Cellular Longevity for Healthy Aging Part 9 ppsx

60 356 0
NEUROVASCULAR MEDICINE - Pursuing Cellular Longevity for Healthy Aging Part 9 ppsx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

NOVEL CELLULAR PATHWAYS 468 Bredesen DE, Rao RV, Mehlen P. 2006. Cell death in the nervous system. Nature. 443:796–802. Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu SS. 2004. Calcium, ATP, and ROS: a mitochondrial love- hate triangle. Am J Physiol Cell Physiol. 287:C817–C833. Brouillet E, Conde F, Beal MF, Hantraye P. 1999. Replicat- ing Huntington’s disease phenotype in experimental animals. Prog Neurobiol. 59:427–468. Browne SE, Bowling AC, MacGarvey U et al. 1997. Oxidative damage and metabolic dysfunction in Huntington’s disease: selective vulnerability of the basal ganglia. Ann Neurol. 41:646–653. Bubber P, Haroutunian V, Fisch G, Blass JP, Gibson GE. 2005. Mitochondrial abnormalities in Alzheimer brain: mechanistic implications. Ann Neurol. 57:695–703. Canevari L, Clark JB, Bates TE. 1999. Beta-amyloid frag- ment 25–35 selectively decreases complex IV activity in isolated mitochondria. FEBS Lett. 457:131–134. Carpenedo R, Pittaluga A, Cozzi A et al. 2001. Presynaptic kynurenate-sensitive receptors inhibit glutamate rele- ase. Eur J Neurosci. 13:2141–2147. Chagnon P, Gee M, Filion M, Robitaille Y, Belouchi M, Gauvreau D. 1999. Phylogenetic analysis of the mito- chondrial genome indicates signi cant differences between patients with Alzheimer’s disease and controls in a French–Canadian founder population. Am J Med Genet. 85:20–30. Chan DC. 2006. Mitochondria: dynamic organelles in dis- ease, aging, and development. Cell. 125:1241–1252. Chaudhuri TK, Paul S. 2006. Protein-misfolding diseases and chaperone-based therapeutic approaches. FEBS J. 273:1331–1349. Chavez E, Melendez E, Zazueta C, Reyes-Vivas H, Perales SG. 1997. Membrane permeability transition as induced by dysfunction of the electron transport chain. Biochem Mol Biol Int. 41:961–968. Chiarugi A, Calvani M, Meli E, Traggiai E, Moroni F. 2001. Synthesis and release of neurotoxic kynurenine metab- olites by human monocyte-derived macrophages. J Neuroimmunol. 120:190–198. Ciechanover A. 2005. Proteolysis: from the lysosome to ubi- quitin and the proteasome. Nat Rev Mol Cell Biol. 6:79–87. Ciechanover A, Brundin P. 2003. The ubiquitin proteasome system in neurodegenerative diseases: sometimes the chicken, sometimes the egg. Neuron. 40:427–446. Coyle JT, Schwarcz R. 1976. Lesion of striatal neurons with kainic acid provides a model of Huntington’s chorea. Nature. 263:244–246. Crompton M. 2000. Mitochondrial intermembrane junc- tional complexes and their role in cell death. J Physiol. 529:11–21. Darios F, Corti O, Lucking CB et al. 2003. Parkin prevents mitochondrial swelling and cytochrome c release in mitochondria-dependent cell death. Hum Mol Genet. 12:517–526. Davey GP, Canevari L, Clark JB. 1997. Threshold effects in synaptosomal and nonsynaptic mitochondria from hippocampal CA1 and paramedian neocortex brain regions. J Neurochem. 69:2564–2570. de Carvalho LP, Bochet P, Rossier J. 1996. The endoge- nous agonist quinolinic acid and the nonendogenous REFERENCES Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. 2004. Inclusion body formation reduces levels of mutant huntington and the risk of neuronal death. Nature. 431:805–810. Autere J, Moilanen JS, Finnila S, Soininen H et al. 2004. Mitochondrial DNA polymorphisms as risk factors for Parkinson’s disease and Parkinson’s disease dementia. Hum Genet. 115:29–35. Balakirev MY, Khramtsov VV, Zimmer G. 1997. Modulation of the mitochondrial permeability transition by nitric oxide. Eur J Biochem. 246:710–718. Baran H, Jellinger K, Deecke L. 1999. Kynurenine meta bol- ism in Alzheimer’s disease. J Neural Transm. 106:165–181. Beal MF. 1992. Does impairment of energy metabolism result in excitotoxic neuronal death in neurodegenera- tive illnesses? Ann Neurology. 31:119–130. Beal MF. 1997. Oxidative damage in neurodegenerative diseases. Neuroscientist. 3:21–27. Beal MF. 2000. Energetics in the pathogenesis of neurode- generative diseases. Trends Neurosci. 23:298–304. Beal MF, Kowall NW, Ellison DW, Mazurek MF, Swartz KJ, Martin JB. 1986. Replication of the neurochemical characteristics of Huntington’s disease by quinolinic acid. Nature. 321:168–171. Beal MF, Ferrante RJ, Swartz KJ, Kowall NW. 1991. Chronic quinolinic acid lesions in rats closely resemble Huntington’s disease. J Neurosci. 11:1649–1659. Beal MF, Matson WR, Storey E et al. 1992. Kynurenic acid concentrations are reduced in Huntington’s disease cerebral cortex. J Neurol Sci. 108:80–87. Beal MF, Brouillet E, Jenkins BG et al. 1993. Neuroche- mical and histologic characterization of striatal exci- totoxic lesions produced by the mitochondrial toxin 3-nitropropionic acid. J Neurosci. 13:4181–4192. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA. 1990. Apparent hydroxyl radical production by peroxyni- trite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA. 87:1620–1624. Bence NF, Sampat RM, Kopito RR. 2001. Impairment of the ubiquitin-proteasome system by protein aggregation. Science. 292:1552–1555. Bender A, Krishnan KJ, Morris CM et al. 2006. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet. 38:515–517. Bennett EJ, Bence NF, Jayakumar R, Kopito RR. 2005. Global impairment of the ubiquitin-proteasome sys - tem by nuclear or cytoplasmic protein aggregates pre- cedes inclusion body formation. Mol Cell. 17:351–365. Betarbet R, Sherer TB, Greenamyre JT. 2005. Ubiquitin- proteasome system and Parkinson’s diseases. Exp Neurol. 191(Suppl 1):S17–S27. Bolanos JP, Heales SJ, Land JM, Clark JB. 1995. Effect of peroxynitrite on the mitochondrial respiratory chain: differential susceptibility of neurones and astrocytes in primary culture. J Neurochem. 64:1965–1972. Bonifati V, Rizzu P, van Baren MJ et al. 2003. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science. 299:256–259. Chapter 18: Pathogenesis of Neurodegenerative Disorders 469 Goldberg AL. 2003. Protein degradation and protec- tion against misfolded or damaged proteins. Nature. 426:895–899. Gu M, Gash MT, Mann VM, Javoy-Agid F, Cooper JM, Schapira AH. 1996. Mitochondrial defect in Hunting- ton’s disease caudate nucleus. Ann Neurol. 39:385–389. Guillemin GJ, Smith DG, Kerr SJ et al. 2000. Characterisa- tion of kynurenine pathway metabolism in human astrocytes and implications in neuropathogenesis. Redox Rep. 5:108–111. Guillemin GJ, Kerr SJ, Smythe GA et al. 2001. Kynurenine pathway metabolism in human astrocytes: a paradox for neuronal protection. J Neurochem. 78:842–853. Guillemin GJ, Brew BJ. 2002. Implications of the kynure- nine pathway and quinolinic acid in Alzheimer’s disease. Redox Rep. 7:199–206. Guillemin GJ, Brew BJ, Noonan CE, Takikawa O, Cullen KM. 2005. Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hip- pocampus. Neuropathol Appl Neurobiol. 31:395–404. Harris ME, Wang Y, Pedigo NW Jr, Hensley K, Butter eld DA, Carney JM. 1996. Amyloid beta peptide (25–35) inhibits Na + -dependent glutamate uptake in rat hip- pocampal astrocyte cultures. J Neurochem. 67:277–286. Hartl FU, Hayer-Hartl M. 2002. Molecular chaperones in the cytosol: from nascent chain to folded protein. Science. 295:1852–1858. Heales SJ, Bolanos JP, Stewart VC, Brookes PS, Land JM, Clark JB. 1999. Nitric oxide, mitochondria and neuro- logical disease. Biochim Biophys Acta. 1410:215–228. Heyes MP, Saito K, Crowley JS et al. 1992. Quinolinic acid and kynurenine pathway metabolism in in amma- tory and non-in ammatory neurologic disease. Brain. 115:1249–1273. Hilmas C, Pereira EFR, Alkondon M, Rassoulpour A, Schwarcz R, Albuquerque EX. 2001. The brain metabo- lite kynurenic acid inhibits α7 nicotinic receptor activ- ity and increases non-α7 nicotinic receptor expression: physiological implications. J Neurosci. 21:7463–7473. Hirata F, Hayaishi O. 1971. Possible participation of super- oxide anion in the intestinal tryptophan 2,3-dioxyge- nase reaction. J Biol Chem. 246:7825–7826. Holmsen H, Robkin L. 1980. Effects of antimycin A and 2-deoxyglucose on energy metabolism in washed human platelets. Thromb Haemost. 42:1460–1472. Jauch DA, Sethy VH, Weick BG, Chase TN, Schwarcz R. 1993. Intravenous administration of -kynurenine to rhesus monkeys: effect on quinolinate and kynurenate levels in serum and cerebrospinal  uid. Neuropharmacology. 32:467–472. Joseph MH, Baker HF, Lawson AM. 1978. Positive identi - cation of kynurenine in rat and human brain. Biochem Soc Trans. 6:123–126. Kapoor R, Okuno E, Kido R, Kapoor V. 1997. Immuno- localization of kynurenine aminotransferase (KAT) in the rat medulla and spinal cord. Neuroreport. 8:3619–3623. Kessler M, Terramani T, Lynch G, Boundary M. 1989. A glycine site associated with N-methyl-D-aspartic acid receptors: characterization and identi cation of a new class of antagonists. J Neurochem. 52:1319–1328. homoquinolinic acid discriminate between NMDAR2 receptor subunits. Neurochem Int. 28:445–452. Delanty N, Dichter MA. 1998. Oxidative injury in the ner- vous system. Acta Neurol Scand. 98:145–153. de Vrij FM, Fischer DF, van Leeuwen FW, Hol EM. 2004. Protein quality control in Alzheimer’s disease by the ubiquitin-proteasome system. Prog Neurobiol. 74:249–270. DiFiglia M. 1990. Excitotoxic injury of the neostriatum: a model for Huntington’s disease. Trends Neurosci. 13:286–289. DiFiglia M, Sapp E, Chase KO et al. 1997. Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neuritis in brain. Science. 277:1990–1993. Du F, Schmidt W, Okuno E, Kido R, Köhler C, Schwarcz R. 1992. Localization of kynurenine aminotransferase immunoreactivity in the rat hippocampus. J Comp Neurol. 321:477–487. Dykens JA. 1997. Mitochondrial free radical production and oxidative pathophysiology: implications for neurode- generative disease. In Beal MF, Howell N, Bódis-Wollner I, eds. Mitochondria and Free Radicals in Neurodegenerative Diseases. New York: John Wiley-Liss, Inc, 29–56. Emerit J, Edeas M, Bricaire F. 2004. Neurodegenerative dis- eases and oxidative stress. Biomed Pharmacother. 58:39–46. Fadeel B, Orrenius S. 2005. Apoptosis: a basic biologi- cal phenomenon with wide-ranging implications in human disease. J Intern Med. 258:479–517. Fan TJ, Han LH, Cong RS, Liang J. 2005. Caspase fam- ily proteases and apoptosis. Acta Biochim Biophys Sin (Shanghai). 37:719–727. Farrer LA, Cupples LA, Haines JL et al. 1997. Effects of age, sex, and ethnicity on the association between apolipo- protein E genotype and Alzheimer disease. A meta- analysis APOE and Alzheimer disease meta Analysis Consortium. JAMA. 278:1349–1356. Foster AC, Miller LP, Oldendorf WH, Schwarcz R. 1984. Studies on the disposition of quinolinic acid after intracerebral or systemic administration in the rat. Exp Neurol. 84:428–440. Foster AC, Schwarcz R. 1989. Neurotoxic effects of quiono- linic acid in the central nervous system. In Stone TW, ed. Quinolinic Acid and Other Kynurenines. Boca Raton, FL: CRC Press, 173–192. Fukui S, Schwarcz R, Rapoport SI, Takada Y, Smith QR. 1991. Blood–brain barrier transport of kynurenines: implications for brain synthesis and metabolism. J Neurochem. 56:2007–2017. Gárdián G, Vécsei L. 2004. Huntington’s disease: path- omechanism and therapeutic perspectives. J Neural Transm. 111:1485–1494. Gárdián G, Browne SE, Choi DK et al. 2005. Neuroprotec- tive effects of phenylbutyrate in the N171–82Q trans- genic mouse model of Huntington’s disease. J Biol Chem. 280:556–563. Gilgun-Sherki Y, Melamed E, Offen D. 2001. Oxidative stress induced-neurodegenerative diseases: the need for antioxidants that penetrate the blood brain barrier. Neuropharmacology. 40:959–975. Goate A, Chartier-Harlin MC, Mullan M et al. 1991. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature. 349:704–706. NOVEL CELLULAR PATHWAYS 470 Kikuchi S, Kim SU. 1993. Glutamate neurotoxicity in mes- encephalic dopaminergic neurons in culture. J Neurosci Res. 36:558–569. Kish SJ, Bergeron C, Rajput A et al. 1992. Brain cyto- chrome oxidase in Alzheimer’s disease. J Neurochem. 59:776–779. Kiss C, Ceresoli-Borroni C, Guidetti P, Zielke CL, Zielke HR, Schwarcz R. 2003. Kynurenate produc- tion by cultured human astrocytes. J Neural Transm. 110:1–14. Kita T, Morrison PF, Heyes MP, Markey SP. 2002. Effects of systemic and central nervous system localized in am- mation on the contributions of metabolic precursors of the -kynurenine and quinolinic acid pools in brain. J Neurochem. 82:258–268. Kitada T, Asakawa S, Hattori N et al. 1998. Mutations in the parkin gene cause autosomal recessive juvenile parkin- sonism. Nature. 392:605–608. Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD. 1997. The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science. 275:1132–1136. Knyihar-Csillik E, Okuno E, Vécsei L. 1999. Effects of in vivo sodium azide administration on the immunohis- tochemical localization of kynurenine aminotrans- ferase in the rat brain. Neuroscience. 94:269–277. Knyihar-Csillik E, Csillik B, Pakaski M et al. 2004. Decreased expression of kynurenine aminotransferase-I (KAT-I) in the substantia nigra of mice after 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine (MPTP) treatment. Neuro- science. 126:899–914. Kopito RR. 2000. Aggresomes, inclusion bodies and pro- tein aggregation. Trends Cell Biol. 10:524–530. Kraytsberg Y, Kudryavtseva E, McKee AC et al. 2006. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet. 38:518–520. Krieger C, Duchen MR. 2002. Mitochondria, Ca 2+ and neu- rodegenerative disease. Eur J Pharmacol. 447:177–188. Kuroda Y, Mitsui T, Kunishige M et al. 2006. Parkin enhances mitochondrial biogenesis in proliferating cells. Hum Mol Genet. 15:883–895. Lehrmann E, Molinari A, Speciale C, Schwarcz R. 2001. Immunohistochemical visualisation of newly formed quinolinate in the normal and excitotoxically lesioned rat striatum. Exp Brain Res. 141:389–397. Leist M, Jaattela M. 2001. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol. 2:589–598. Leroy E, Boyer R, Auburger G et al. 1998. The ubiquitin pathway in Parkinson’s disease. Nature. 395:451–452. Levy-Lahad E, Wasco W, Poorkaj P et al. 1995. Candidate gene for the chromosome 1 familial Alzheimer’s dis- ease locus. Science. 269:973–977. Lu F, Selak M, O’Connor J et al. 2000. Oxidative damage to mitochondrial DNA and activity of mitochondrial enzymes in chronic active lesions of multiple sclerosis. J Neurol Sci. 177:95 –103. Luchowski P, Luchowska E, Turski WA, Urbanska EM. 2002. 1-Methyl-4-phenylpyridinium and 3-nitropropionic acid diminish cortical synthesis of kynurenic acid via interference with kynurenine aminotransferases in rats. Neurosci Lett. 330:49–52. Luchowska E, Luchowski P, Wielosz M, Turski WA, Urbanska EM. 2003. FK506 attenuates 1-methyl- 4- phenylpyridinium- and 3-nitropropionic acid-evoked inhibition of kynurenic acid synthesis in rat cortical slices. Acta Neurobiol Exp (Wars). 63:101–108. Lyubchenko YL, Sherman S, Shlyakhtenko LS, Uversky VN. 2006. Nanoimaging for protein misfolding and related diseases. J Cell Biochem. 99:52–70. Maccioni RB, Munoz JP, Barbeito L. 2001. The molecular bases of Alzheimer’s disease and other neurodegenera- tive disorders. Arch Med Res. 32:367–381. Manfredi G, Beal F. 2000. The role of mitochondria in the pathogenesis of neurodegenerative diseases. Brain Pathol. 10:462–472. Martin LJ, Pan Y, Price AC et al. 2006. Parkinson’s disease alpha-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death. J Neurosci. 26:41–50. McGeer EG, McGeer PL. 1976. Duplication of biochemical changes of Huntington’s chorea by intrastriatal injec- tion of glutamic and kainic acids. Nature. 263:517–519. McNaught KS, Olanow CW. 2003. Proteolytic stress. A uni- fying concept in the etiopathogenesis of familial and sporadic Parkinson’s disease. Ann Neurol. 53(Suppl 3): S73–S84. McNaught KS, Olanow CW. 2006. Protein aggregation in the pathogenesis of familial and sporadic Parkinson’s disease. Neurobiol Aging. 27:530–545. Miranda AF, Boegman RJ, Beninger RJ, Jhamadas K. 1997. Protection against quinolinic acid-mediated excitotox- icity in nigrostriatal dopaminergic neurons by endog- enous kynurenic acid. Neuroscience. 78:967–975. Miranda AF, Sutton MA, Beninger RJ, Jhamadas K, Boegman RJ. 1999. Quinolinic acid lesion of the nigrostriatal pathway: effect on turning behaviour and protection by elevation of endogenous kynurenic acid in Rattus norvegicus. Neurosci Lett. 262:81–84. Moroni F. 1999. Tryptophan metabolism and brain func- tion: focus on kynurenine and other indol metabolites. Eur J Pharmacol. 375:87–100. Moroni F, Russi G, Lombardi G, Beni M, Carla V. 1988. Presence of kynurenic acid in the mammalian brain. J Neurochem. 51:177–180. Muchowski PJ, Wacker JI. 2005. Modulation of neurode- generation by molecular chaperones. Nat Rev Neurosci. 6:11–22. Mutisya EM, Bowling AC, Beal MF. 1994. Cortical cyto- chrome oxidase activity is reduced in Alzheimer’s dis- ease. J Neurochem. 63:2179–2184. Nagata S. 2005. DNA degradation in development and pro- grammed cell death. Annu Rev Immunol. 23:853–875. Nass S, Nass MM. 1963. Intramitochondrial  bers and DNA characteristics. II. Enzymatic and other hydrolytic treatments. J Cell Biol. 19:613–629. Nemeth H, Robotka H, Kis Z et al. 2004. Kynurenine admin- istered together with probenecid markedly inhibits pentylenetetrazol-induced seizures. An electrophysi- ological and behavioural study. Neuropharmacology. 47:916–925. Chapter 18: Pathogenesis of Neurodegenerative Disorders 471 Nemeth H, Toldi J, Vécsei L. 2005. Role of kynurenines in the central and peripheral nervous systems. Curr Neurovasc Res. 2:249–260. Nunomura A, Perry G, Aliev G et al. 2001. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 60:759–767. Ogawa T, Matson WR, Beal MF et al. 1992. Kynurenine path- way abnormalities in Parkinson’s disease. Neurology. 42:1702–1706. Okuno E, Nakamura M, Schwarcz R. 1991. Two kynure- nine aminotransferases in human brain. Brain Res. 542:307–312. Orth M, Schapira AH. 2001. Mitochondria and degenera- tive disorders. Am J Med Genet. 106:27–36. Papa S. 1996. Mitochondrial oxidative phosphoryla- tion changes in the life span. Molecular aspects and physiopathological implications. Biochim Biophys Acta. 1276:87–105. Paschen W, Mengesdorf T. 2005. Endoplasmic reticulum stress response and neurodegeneration. Cell Calcium. 38:409–415. Perkins MN, Stone TW. 1985. Actions of kynurenic acid and quinolinic acid in the rat hippocampus in vivo. Exp Neurol. 88:570–579. Perutz MF, Johnson T, Suzuki M, Finch JT. 1994. Glutamine repeats as polar zippers: their possible role in inherited neurodegenerative diseases. Proc Natl Acad Sci U S A. 91:5355–5358. Petit A, Kawarai T, Paitel E et al. 2005. Wild-type PINK1 prevents basal and induced neuronal apoptosis, a pro- tective effect abrogated by Parkinson’s disease-related mutations. J Biol Chem. 280:34025–34032. Petrucelli I, Dawson TM. 2004. Mechanism of neurode- generative disease: role of the ubiquitin proteasome system. Ann Med. 36:315–320. Pickart CM. 2001. Mechanisms underlying ubiquitination. Annu Rev Biochem. 70:503–533. Poderoso JJ, Carreras MC, Lisdero C, Riobo N, Schopfer F, Boveris A. 1996. Nitric oxide inhibits electron trans- fer and increases superoxide radical production in rat heart mitochondria and submitochondrial particles. Arch Biochem Biophys. 328:85–92. Polymeropoulos MH , Lavedan C, Leroy E, Ide SE et al. 1997. Mutation in the alpha-synuclein gene identi-  ed in families with Parkinson’s disease. Science. 276:2045–2047. Prescott C, Weeks AM, Staley KJ, Partin KM. 2006. Kynurenic acid has a dual action on AMPA receptor responses. Neurosci Lett. 402:108–112. Pyle A, Foltynie T, Tiangyou W et al. 2005. Mitochondrial DNA haplogroup cluster UKJT reduces the risk of PD. Ann Neurol. 57:564–567. Reynolds GP, Pearson SJ, Halket J, Sandler M. 1988. Brain quinolinic acid in Huntington’s disease. J Neurochem. 50:1959–1960. Rios C, Santamaria A. 1991. Quinolinic acid is a potent lipid peroxidant in rat brain homogenates. Neurochem Res. 16:1139–1143. Robotka H, Nemeth H, Somlai C, Vécsei L, Toldi J. 2005. Systemically administered glucosamine-kynurenic acid, but not pure kynurenic acid, is effective in decreasing the evoked activity in area CA1 of the rat hipocampus. Eur J Pharmacol. 513:75–80. Rodriguez-Martinez E, Camacho A, Maldonado PD et al. 2000. Effect of quinolinic acid on endogenous antioxi- dants in rat corpus striatum. Brain Res. 858:436–439. Rogaev EI, Sherrington R, Rogaeva EA et al. 1995. Familiar Alzheimer’s disease in kindreds with missense muta- tions in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature. 376:775–778. Rosenberg RN. 2000. The molecular and and genetic basis of Alzheimer’s disease: the end of the beginning. Neurology. 54:2045–2054. Santamaría A, Galván-Arzate S, Lisý V et al. 2001. Quinoli- nic acid induces oxidative stress in rat brain synapto- somes. Neuroreport. 12:871–874. Sas K, Robotka H, Toldi J, Vécsei L. 2007. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J Neur Sci. 257:221–239. Schapira AH, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD. 1989. Mitochondrial complex I de ciency in Parkinson’s disease. Lancet. 1:1269. Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD. 1990. Mitochondrial complex I de ciency in Parkinson’s disease. J Neurochem. 54:823–827. Schwarcz R. 2004. The kynurenine pathway of tryptophan degradation as a drug target. Curr Opin Pharmacol. 4:12–17. Schwarcz R, Whetsell WO Jr, Mangano RM. 1983. Quinoli- nic acid an endogenous metabolite that produces axon-sparing lesion in rat brain. Science. 219:316–318. Schwarcz R, Foster AC, French ED, Whetsell WO Jr, Kohler C. 1984. Excitotoxic models for neurodege- nerative disorders. Life Sci. 35:19–32. Schwarcz R, Tamminga CA, Kurlan R, Shoulson I. 1988. Cerebrospinal  uid levels of quinolinic acid in Huntington’s disease and schizophrenia. Ann Neurol. 24:580–582. Selkoe DJ. 2001. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 81:741–766. Shastry BS. 2003. Neurodegenerative disorders of protein aggregation. Neurochem Int. 43:1–7. Sherman MY, Goldberg AL. 2001. Cellular defenses against unfolded proteins a cell biologist thinks about neuro- degenerative diseases. Neuron. 29:15–32. Sherrington R, Rogaev EI, Liang Y et al. 1995. Cloning of the gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 375:754–760. Sies H. 1993. Strategies of antioxidant defense. Eur J Biochem. 215:213–219. Sitia R, Braakman I. 2003. Quality control in the endoplas- mic reticulum protein factory. Nature. 426:891–894. Small GW, Mazziotta JC, Collins MT et al. 1995. Apolipo- protein E type 4 allele and cerebral glucose metabo- lism in relatives at risk for familial Alzheimer disease. JAMA. 273:942–947. Soti C, Csermely P. 2003. Aging and molecular shaperones. Exp Gerontol. 38:1037–1040. Stewart VC, Heales SJ. 2003. Nitric oxide-induced mito- chondrial dysfunction: implications for neurodegen- eration. Free Radic Biol Med. 34:287–303. NOVEL CELLULAR PATHWAYS 472 Van Houten B, Woshner V, Santos JH. 2006. Role of mito- chondrial DNA in toxic responses to oxidative stress. DNA Repair (Amst). 5:145–152. Vécsei L, Beal MF. 1990. Intracerebroventricular injec- tion of kynurenic acid, but not kynurenine, induces ataxia and stereotyped behavior in rats. Brain Res Bull. 25:623–627. Vécsei L, Beal MF. 1991. Comparative behavioral and pharmacological studies with centrally administered kynurenine and kynurenic acid in rats. Eur J Pharmacol. 196:239–246. Vécsei L, Miller J, MacGarvey U, Beal MF. 1992. Kynurenine and probenecid inhibit pentylentet- razol- and NMDLA-induced seizures and increase kynurenic acid concentrations in the brain. Brain Res Bull. 28:233–238. Vécsei L, Beal MF. 1996. Huntington’s disease, behav- ioral disturbances, and kynurenines: preclinical  ndings and therapeutic perspectives. Biol Psychiatry. 39:1061–1063. Vécsei L, Dibo G, Kiss C. 1998. Neurotoxins and neurode- generative disorders. Neurotoxicology. 19:511–514. Virág L. 2005. Structure and function of poly(ADP-ribose) polymerase-1: role in oxidative stress-related patholo- gies. Curr Vasc Pharmacol. 3:209–214. Walton MR, Dragunow I. 2000. Is CREB a key to neuronal survival? Trends Neurosci. 23:48–53. Widner B, Leblhuber F, Walli J, Tilz GP, Demel U, Fuchs D. 1999. Degradation of tryptophan in neurodegenera- tive disorders. Adv Exp Med Biol. 467:133–138. Wyttenbach A, Swartz J, Kita H et al. 2001. Polyglutamine expansions cause decreased CREB-mediated tran- scription and early gene expression changes prior to cell death in an inducible cell model of Huntington’s disease. Hum Mol Genet. 10:1829–1845. Yakes FM, Van Houten B. 1997. Mitochondrial DNA dam- age is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A. 94:514–519. Yang Y, Gehrke S, Haque ME et al. 2005. Inactivation of Drosophila DJ-1 leads to impairments of oxidative stress response and phosphatidylinositol 3-kinase/Akt signaling. Proc Natl Acad Sci U S A. 102:13670–13675. Zhang L, Shimoji M, Thomas B et al. 2005. Mitochondrial localization of the Parkinson’s disease related protein DJ-1: implications for pathogenesis. Hum Mol Genet. 14:2063 –2073. Zimprich A, Biskup S, Leitner P et al. 2004. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 44:601–607. Stone TW. 2001. Kynurenic acid antagonists and kynure- nine pathway 45. Exp Opin Invest Drugs. 10:633. Stone TW. 2001. Kynurenines in the CNS: from endogenous obscurity to therapeutic importance. Progr Neurobiol. 64:185–218. Stone TW, Perkins MN. 1981. Quinolinic acid: A potent endogenous excitant at amino acid receptors in CNS. Eur J Pharmacol. 72:411–412. Stone TW, Connick JH. 1985. Quinolinic acid and other kynurenines in the central nervous system. Neuroscience. 15:597–617. Stoy N, Mackay GM, Forrest CM et al. 2005. Tryptophan metabolism and oxidative stress in patients with Huntington’s disease. J Neurochem. 93:611–623. Strauss KM, Martins LM, Plun-Favreau H et al. 2005. Loss of function mutations in the gene encoding Omi/HtrA2 in Parkinson’s disease. Hum Mol Genet. 14:2099–2111. Suzuki Y, Imai Y, Nakayama H, Takahashi K, Takio K, Takahashi R. 2001. A serine protease, HtrA2, is relea- sed from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell. 8:613–621. Swartz KJ, During MJ, Freese A, Beal MF. 1990. Cerebral synthesis and release of kynurenic acid: an endog- enous antagonist of excitatory amino acid receptors. J Neurosci. 10:2965–2973. Takuma K, Yao J, Huang J et al. 2005. ABAD enhances Abeta-induced cell stress via mitochondrial dysfunc- tion. FASEB J. 19:597–598. Tavares RG, Tasca CI, Santos CE et al. 2002. Quinolinic acid stimulates synaptosomal glutamate release and inhibits glutamate uptake into astrocytes. Neurochem Int. 40:621–627. Taylor JP, Hardy J, Fischbeck KH. 2002. Toxic proteins in neurodegenerative disease. Science. 296:1991–1995. Tipton KF, Singer TP. 1993. Advances in our understanding of the mechanisms of the neurotoxicity of MPTP and related compounds. J Neurochem. 61:1191–1206. Turski WA, Nakamura M, Todd WP, Carpenter BK, Whetsell WO Jr, Schwarcz R. 1988. Identi cation and quanti ca- tion of kynurenic acid in human brain tissue. Brain Res. 454:164–169. Valente EM, Abou-Sleiman PM, Caputo V et al. 2004. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 304:1158–1160. Valera AG, Diaz-Hernandez M, Hernandez F, Ortega Z, Lucas JJ. 2005. The ubiquitin-proteasome system in Huntington’s disease. Neuroscientist. 11:583–594. van der Walt JM, Nicodemus KK, Martin ER et al. 2003. Mitochondrial polymorphisms signi cantly reduce the risk of Parkinson’s disease. Am J Hum Genet. 72:804–811. 473 Chapter 19 REDOX SIGNALING AND VASCULAR FUNCTION J. Will Langston, Magdalena L. Circu, and Tak Yee Aw ABSTRACT Over the last two decades, redox signaling has emerged as an important regulator of cell function, and it is now well appreciated that reactive oxygen and nitrogen species act as second messengers that modulate vascular activity via direct interactions with specifi c enzymes, proteins, and transcription factors to regulate cell signaling and/or gene expression. The growing interest in the role of redox signaling in the vasculature stems primarily from evidence that oxida- tive stress-induced endothelial dysfunction underlies a number of cardiovascular pathologies includ ing hypertension, atherosclerosis, and diabetes, and that antioxidant intervention may be an important treatment modality in these vascular disorders. Of interest is the thiol antioxidant, reduced glutathione (GSH), a crucial regulator of cellular redox potential, and whose synthesis is transcriptionally upregulated under conditions of cellular oxidative stress. The tran- scriptional upregulation of the rate-limiting enzyme of GSH synthesis, glutamate cysteine ligase (GCL), under oxidative conditions by the transcription factor Nrf2 represents an important area of investigation in terms of its role in redox regulation of endothelial function, its role in vascular pathology, and its poten- tial as a therapeutic target for treatment of cardiovas- cular disorders that involve vascular oxidative stress. Keywords: GSH redox status and signaling, GSH and vascular function, redox regulation of GSH synthesis, mechanisms of redox signaling. R edox signaling and posttranslational redox modi cations of protein thiols are emerging to be fundamentally important signaling mechanisms in the regulation of mamma- lian cell function. In addition to redox regulation of cell signaling being a modulator of normal function, a disturbance of redox signaling has also been sug- gested to underpin a variety of pathologies, including vascular diseases. The current chapter will  rst focus on a general discussion of the concept of cellular redox status, the compartmentation of cellular redox systems, and the mechanisms of redox signaling and its targets. The rest of the chapter will be devoted to coverage of the speci c role of vascular-derived reac- tive oxygen and nitrogen species, the involvement of the glutathione redox system and Nrf2 in the path- ways of redox signaling in vascular function and dys- function, speci c oxidative stress–associated vascular diseases, and antioxidant therapy in treatment of vascular disorders. GENERAL CONSIDERATION OF THE REDOX STATE OF A CELL AND ITS SIGNIFICANCE The redox state of a cell is de ned by the ratio of the interconvertible reduced and oxidized forms of the dif- ferent cellular redox couples. More generally, the term redox environment has been used to describe the state of the cellular redox pairs (Schafer, Buettner 2001). NOVEL CELLULAR PATHWAYS 474 The intracellular thiol redox pairs are represented by the reduced glutathione/glutathione disul de (GSH/ GSSG), and the reduced and oxidized thioredoxin (Trx/TrxSS) systems, while the cysteine/cystine (Cys/ CySS) redox couple plays an important role in main- taining the redox state of the plasma. The pyridine nucleotide couples include nicotinamide adenine dinucleotide/reduced nicotinamide adenine dinucle- otide (NAD + /NADH) and NAD phosphate/reduced NAD phosphate (NADP + /NADPH). The oxidation–re- duction status of the redox components is responsible for creating an optimal redox environment within the cell, which directly affects the activity of different cel- lular proteins. Recently, Hansen et al. proposed that the cellular redox systems are differentially compart- mentalized among different organelles, where the dis- tribution of redox systems is independently controlled in the plasma membrane, cytosol, nucleus, mitochon- dria, and endoplasmic reticulum (ER) (Hansen, Go, Jones 2006). Thus, depending on the concentrations of the respective redox couples and their  uxes, the compartmentation of speci c redox systems may, in fact, represent a crucial and generalized mechanism for optimizing cell activity within mammalian cells. We have, in recent years established a paradigm that an oxidative shift in the cellular GSH/GSSG redox couple is an important determinant of cell fate; the phenotypic endpoint of proliferation, growth arrest or apoptosis is a function of the extent of GSH/GSSG imbalance (Aw 1999, 2003; Noda, Iwakiri, Fujimoto et al. 2001; Gotoh, Noda, Iwakiri et al. 2002). In vari- ous cell types, the loss of GSH/GSSG redox balance preceding cell apoptosis is an early event that occurred within a relatively narrow time window (30 minutes) post–oxidant challenge and is preventable by pre- treatment with the thiol antioxidant, N-acetylcysteine (NAC) (Wang, Gotoh, Jennings et al. 2000; Pias, Aw 2002a, 2002b; Pias, Ekshyyan, Rhoads et al. 2003; Ekshyyan, Aw 2005; Okouchi, Okayama, Aw 2005). These  ndings suggest that GSH/GSSG redox signal- ing may represent a generalized mechanism in oxida- tive cell killing in mammalian cells. The control of cellular apoptosis by mucosal GSH/GSSG redox status has been demonstrated in vivo (Tsunada, Iwakiri R, Noda et al. 2003). The current understanding of redox signaling is that it is a regulatory process in which the signal occurs through redox reactions induced by reactive oxygen species (ROS) or reactive nitrogen species (RNS) that results in posttranslational modi cation of proteins in various signal transduction pathways. Many proteins contain cysteine residues that provide a redox-sensitive switch for regulating protein function, and ROS-induced oxidation of cysteine-SH can result in the formation of intra- and/or interchain disul de bonds. Moreover, the direct addition of GSSG leads to S-glutathionylation of the thiol moiety. In addi- tion, nitric oxide (NO • ) can induce S-nitrosylation of speci c cysteine thiols in proteins such as soluble guanylate cyclase (sGC) and the newly discovered mitochondrial NO • /cy tochrome c oxidase signaling pathway (Shiva, Huang, Grubina et al. 2007; Landar, Darley-Usmar 2007). These redox signal transduction processes are important in various physiological and biological activities including vascular function. CONCEPT OF OXIDATIVE AND NITROSATIVE STRESS AND REDOX SIGNALING In redox signaling, modi cations of targeted proteins are initiated by ROS and RNS. The common ROS are superoxide anions (O 2 •– ), hydrogen peroxide (H 2 O 2 ), and hydroxyl radicals (HO•) while RNS comprise NO • and its derivatives, peroxynitrite (ONOO – ) or dinitrogen trioxide (N 2 O 3 ) (Fig. 19.1). Endogenous sources of O 2 •– include the mitochondrial respiratory chain, NADPH oxidase, xantine oxidase, and NADPH cytochrome P450 (Cross, Jones 1991). Intracellular derived O 2 •– is readily dismutated to H 2 O 2 by cytoso- lic or mitochondrial superoxide dismutases (SOD). In the presence of metal ions, H 2 O 2 and O 2 •– are con- verted to HO•, a highly potent oxidant that induces oxidative damage to cellular proteins, lipids, or DNA. Exogenous sources such as xenobiotics or UV/γ- radiations are known ROS generators that contrib- ute to the overall oxidant burden of a cell. O 2 •– can further react with NO • to form the reactive ONOO – that oxidizes cellular lipids or DNA, resulting in nitro- sative stress (for review see Pacher, Beckman, Liaudet 2007). NO • is generated by NO synthases (NOS), of which three isoforms exist in mammalian cells; these are endothelial NOS (eNOS), neuronal NOS (nNOS), and inducible NOS (iNOS). In the vasculature, eNOS is the predominant isoform and is responsible for maintaining vascular NO • homeostasis and vascu- lar tone. Oxidation of the essential NOS cofactor by ONOO – transforms eNOS into an ROS producer (Forstermann, Closs, Pollock et al. 1994). Another RNS derivative with a potential role in cellular sig- naling is N 2 O 3 , which participates in the nitrosation of thiol groups to form nitrosothiols, an important class of redox signaling molecules. Endothelial ROS and RNS generation and their speci c roles in vas- cular function are discussed in sections on cellular sources of endothelial ROS and ROS and vascular signaling. It is well recognized that different concentrations of ROS/RNS mediate distinct cellular responses. While high ROS/RNS concentration induces oxida- tive damage to macromolecules that lead to oxidative/ Chapter 19: Redox Signaling and Vascular Function 475 COMPARTMENTATION OF CELLULAR REDOX SYSTEMS AND REDOX PROTEINS IN CELL SIGNALING GSH/GSSG Redox System and its Subcellular Compartmentation Given its cellular abundance and its role in protein thiol modi cation, the status of the GSH/GSSG redox system re ects the redox buffering capacity of a cell. Indeed, an oxidative shift in the GSH-to-GSSG ratio is often used as an indicator of cellular oxidative stress. GSH/GSSG and Cellular Redox Balance GSH (γ-glutamylcysteinyl glycine) is the most abun- dant low-molecular-weight thiol in cells that exhibits important roles in the control of the thiol–disul de redox state of cellular proteins (Meister, Anderson 1983; Sies 1999). Additionally, GSH is involved in redox activation of transcription factors as part of an adaptive mechanism that participates in cell signaling and stress responses (Kamata, Hirata 1999). Within cells, GSH exists mainly in its biologically active, thiol-reduced form, and oxidation of GSH results in the formation of glutathione disul de, GSSG. The ratio of GSH and GSSG is maintained in favor of the nitrosative stress, low-to-moderate levels are impor- tant in cell signaling and the regulation of various biological processes. The balance of detrimental and bene cial actions caused by ROS/RNS is achieved through “redox regulation” mechanisms; this refers to enzymatic reactions with speci c roles in maintain- ing the redox homeostasis of targeted proteins that are essential for cell function and survival (Droge 2002). The cytotoxic effects of ROS/RNS are amelio- rated by intracellular antioxidant mechanisms that maintain a balance of the reduced and oxidized spe- cies (Fig. 19.1). For example, H 2 O 2 and hydroperox- ides are eliminated by GSH peroxidase at the expense of cellular GSH, and the resultant increase in GSSG is reduced by glutathione reductase with NADPH as the reductant. Peroxiredoxin (Prx) is another example of antioxidant redox proteins that are involved in the breakdown of cellular toxic hydroperoxides. Previous studies have demonstrated that these cysteine-speci c peroxidases can function as molecular switches sen- sitive to different levels of H 2 O 2 (Bozonet, Findlay, Day et al. 2005). The Trx/Trx reductase and glutare- doxin (Grx)/GSH system also contribute to cellular antioxidant defense against redox imbalance as do the sul redoxins (Srx) in the reduction of oxidized proteins (Fig. 19.1; section on redox proteins and cell signaling). Figure 19.1 Metabolic pathways of ROS/RNS formation and the interactions with antioxidant systems. ROS are formed from exogenous and endogenous sources. While highly reactive HO• induces oxidative stress, O2 •– and H 2 O 2 are either substrates for antioxidant enzymes or induce sequential oxidation of Cys residues of target proteins of various signaling pathways. The formation of GSSG can participate in the S-glutathionylation of proteins (P-SSG). NO radicals produced by NOS isoenzymes can form diverse RNS intermediates that either participate in S-nitrosylation of Cys residues of proteins to form GSNO/P-SNO derivatives or induce nitrosative stress and the formation of tyrosine nitrosative derivatives. The redox status of proteins is restored by the GSH, Trx, Grx, and the newly discovered Srx redox systems. CAT, catalase; GSH, glutathione; GPx, GSH peroxidase; GR, GSH reductase; Grx, glutaredoxin; GSSG, glutathione disulfi de; NO • nitric oxide radical; NOS, nitric oxide synthases; N 2 O 3 , dinitrogen trioxide; ONOO – , peroxynitrite; SOD, superoxide dismutase; Srx, sulfi redoxin; Trx, thioredoxin. Exo- and endogenous sources NOS ONOO - GSH P-SH GSNO GSH GSH P-SH P-SNO P-SSG P-SH Trx, Grx, GSH, Srx Cys-oxidation (P-SO n H) Nitrosative stress NO SOD CAT GSH H 2 O 2 HO• GSSG GPx GR NADP + Fe 3+ Fe 2+ NADPH Glucose O 2 + H 2 O N 2 O 3 Oxidative stress Cys-oxidation (P-SO n H) O 2 •– NOVEL CELLULAR PATHWAYS 476 in excess of 100-to-1 in liver cells, and this ratio signi - cantly decreases to less than 4 to 1 during oxidative stress. The mitochondria maintain a distinct GSH pool that is supported through GSH transport from the cytosolic compartment via the dicarboxylate and 2-oxoglutarate GSH carriers located in the mitochon- drial inner membrane (Chen, Lash 1998). This GSH redox compartment is metabolically separate from the cytosol with regard to synthetic rate, turnover, and sensitivity to chemical depletion. Matrix GSH concen- trations are between 5 and 10 mM and varies from 10% to 15% of the total GSH in the liver (Jocelyn, Kamminga 1974) to 15% to 30% of total GSH pool in the renal proximal tubule (Schnellmann 1991). Functionally, mitochondrial GSH preserves the integ- rity of mitochondrial proteins and lipids and con- trols mitochondrial generation of ROS. Early studies demonstrated that the status of mitochondrial GSH is a determining factor in oxidative vulnerability; in this regard, mitochondrial GSH loss has been linked to cytotoxicity induced by aromatic hydrocarbons (Hallberg, Rydstrom 1989), hypoxia (Lluis, Morales, Blasco et al. 2005), tert- butylhydroperoxide (tBH) (Olafsdottir, Reed 1988), and ethanol intoxication reduced state (>90% reduced), which is accomplished by three mechanisms: GSH synthesis, GSSG reduction, and GSH uptake. De novo synthesis of GSH from pre- cursor amino acids (glutamate, cysteine, and glycine) occurs in the cytosol and is catalyzed by two ATP- dependent enzymatic reactions, γ-glutamate cysteine ligase (GCL) and GSH synthetase (GS) (Fig. 19.2). GCL activity is rate limiting in GSH synthesis and is regulated by GSH and the availability of cysteine. GSSG reduction is catalyzed by glutathione reductase, and uptake of extracellular GSH occurs through spe- ci c carriers localized at the plasma membrane (Lash, Putt, Xu et al. 2007). Intracellular Compartmentation of GSH In mammalian cells, GSH is present in millimolar concentrations and is differentially distributed among various cellular compartments, such as the cytosol, mitochondria, ER, and nucleus where it forms sepa- rate and distinct redox pools (Fig. 19.2). Within the cytosol, GSH concentrations are maintained between 5 mM and 10 mM (Meister, Anderson 1983) and the redox pool is highly reduced; for example, the GSH- to-GSSG ratio under normal conditions is maintained Figure 19.2 GSH synthesis and compartmentation of thiol/disulfi de redox state and redox proteins. Synthesis of GSH from its constituent amino acids (glutamate, cysteine, glycine) takes place in the cytosol and is catalyzed by glutamate cysteine ligase (GCL) and glutathione synthase (GS) at the expense of two moles of ATP. Some cells can export GSH or Trx1 which, once outside the cells, can contribute to maintaining the redox environment of the plasma. Extracellular GSH is hydrolyzed to its component amino acids by γ-glutamyltransferase (γ-GT) and dipeptidase (DP). The main redox couples that participate in maintaining the cellular reduced-to-oxidized environment include GSH/GSSG and Trx/TrxSS. The cysteine residues in cellular proteins are maintained in the reduced state by GSH and thiol reductases, namely, Trx, Grx, Nrx, or PDI that have different cellular localization in the cytoplasm, mitochondria, endoplasmic reticulum, and nucleus. Cys, cysteine; Grx, glutaredoxin; GSH, glutathione; GSSG, glutathione disulfi de; Nrx, nucleoredoxin; PDI, protein disulfi de isomerase; PrSH, reduced protein; Pr-SSG, oxidized protein; Trx, thioredoxin; TrxSS, oxidized thioredoxin; Trx80, truncated form of Trx. Cytoplasm GSH (5–10 mM) Trx1 (2–14 µM) Grx1 Trx1 or Trx80 GSH/GSSG Trx1/TrxSS GSH/GSSG GSH/GSSG GSH/GSSG Prot-SH/ Prot-SSG GSH PDI GSH Trx1 Grx2 Nrx Trx1/TrxSS Trx2/TrxSS GSH/GSSG GSH Trx2 Grx2 Glu + Cys g-glu-cys gGT Cys, Gly DP GS GSH Nucleus Endoplasmic reticulum Mitochondria ATP ADP Extracellular GSH ATP ADP GCL Chapter 19: Redox Signaling and Vascular Function 477 current evidence suggests a passive diffusion of GSH from the cytosol into the nucleus via nuclear pores (Ho, Guenthner 1997). Redox Proteins and Cell Signaling Members of the Trx family of proteins are active in the redox regulation of cysteine residues of special- ized proteins that signi cantly impact cell signaling and function. Among the better-studied redox pro- teins are Trx, Grx, peroxiredoxin, and protein disul-  de isomerase (PDI). Thioredoxins Thioredoxins (Trx) are small ubiquitous redox pro- teins that contain two redox-active cysteine residues in the catalytic site (Cys-X-X-Cys). The intracellular concentrations of Trx are between 2 µM and 14 µM, which is three orders of magnitude lower than that of GSH (2 mM to 10 mM) (Nakamura, Nakamura, Yodoi 1997). Trx, together with NADPH and Trx reductase, functions to maintain the thiol/disul de redox state of proteins in mammalian cells. Trx catalyzes the reversible reduction of disul de bonds in oxidized proteins at the expense of cysteine residues in its active motif site; the active reduced Trx is regenerated by Trx reductase and NADPH. Mammalian cells contain two forms of Trx, Trx1, and Trx2, which are localized in different cellular compartments, cytosol, mitochondria, or nucleus. Trx 1 is expressed ubiquitously and is a cytosolic enzyme that can translocate to the nucleus during oxidative stress. Cytosolic Trx1 can function as a cofactor, bind- ing partner, or reductant. For example, as a cofactor for Prx, Trx1 functions in hydroperoxide elimina- tion. The binding of Trx1 with the apoptosis signal– regulated kinase 1 (ASK-1) plays an anti-apoptotic role (Saitoh, Nishitoh, Fujii et al. 1998). As a reduc- tant, Trx1 and Trx-like protein (TRP14) reactivate the protein tyrosine phosphatase (PTP), phosphatase- like tensin homolog (PTEN), which reverses phos- phoinositide 3-kinase (PI3K) signaling (Lee, Yang, Kwon et al. 2002). Through reduction of protein dis- ul des, Trx functions in redox-sensitive signaling and the activation of transcriptional factors (for review see Watson, Yang, Choi et al. 2004). During oxidative challenge, Trx1 translocates to the nucleus where it participates in the redox regulation of transcription factors such as activator protein 1 (AP-1), p53, and nuclear transcription factor kappa B (NF-κB). Trx1 involvement in redox control of transcriptional activ- ity is supported by the observation that the redox state of nuclear Trx1 is more reduced than that of the cyto- solic protein (Watson, Jones 2003). Several studies (Fernandez-Checa, Garcia-Ruiz, Ookhtens et al. 1991). Our recent studies validated that oxidant- induced apoptosis is triggered by a loss of mitochon- drial GSH/GSSG balance (Circu, Rodriguez, Maloney et al. 2008). Mechanistically, oxidative susceptibility is associated with an increase in mitochondrial ROS production secondary to matrix GSH decrease (Lluis, Buricchi, Chiarugi et al. 2007). The existence of a distinct GSH pool in the ER has been described and its concentration (6 mM to 10 mM) mirrors those in the cytosolic and mitochon- drial compartments (Fig. 19.2). Notably, the GSH redox environment in ER is highly oxidized (GSH-to- GSSG ratio of 3:1–1:1), a state that favors the oxida- tive folding of proteins (Bass, Ruddock, Klappa et al. 2004). The luminal GSSG concentration is, indeed, optimal for disul de bond formation (Lyles, Gilbert 1991), and appears to be generated through an oxida- tive pathway catalyzed by the oxidoreductase enzyme, Er01 (Tu, Weissman 2004). Also notable is that less than 50% of the ER thiol pool (GSH + GSSG) is free; the majority of GSH is reversibly bound to proteins as protein-mixed disul des formed through thiol oxi- dation by GSSG. Functionally, it is believed that high concentrations of protein-mixed disul des serve as GSH reserve within the ER, in the maintenance of oxidoreductase catalytic function, or as a redox buf- fer against ER-generated ROS (Jessop, Bulleid 2004; Chakravarthi, Jessop, Bulleid 2006). Elevated levels of reduced GSH disrupt ER function and activate the unfolded protein response (UPR) that triggers cellu- lar apoptosis (Frand, Kaiser 2000). An independent nuclear GSH pool functions in DNA synthesis and protection against oxidative and ionizing radiation induced DNA damage (Cotgreave 2003). The size of the nuclear GSH pool is unknown, and recent evidence suggests that the cytosolic and nuclear redox pools are not in equilibrium. Bellomo and coworkers (Bellomo, Palladini, Vairetti 1997) demonstrated a GSH ratio of 3:1 between the nuclear and cytosolic compartments, while Thomas et al. (1995) and Soderdahl et al. (2003) reported lower ratios. Moreover, nuclear proteins are more prone to thiol oxidation (Soderdahl, Enoksson, Lundberg et al. 2003). Interestingly, nuclear GSH distribution is dynamic and directly correlates with cell cycle pro- gression where nuclear GSH was 4-fold higher than cytosolic GSH in the proliferative state, but was equally distributed between the two compartments when cells reached con uency (Chen, Delannoy, Odwin et al. 2003; Markovic, Borras, Ortega et al. 2007). These results suggest a speci c role for nuclear GSH in pre- serving nuclear proteins in a reducing environment that is essential for gene transcription during cell cycle progression (Chen, Delannoy, Odwin et al. 2003). The mechanism for nuclear GSH transport is unresolved; [...]... binding of AP-1 is attenuated by S-glutathionylation of the cysteine residue in the AP-1 catalytic site in response to decreased cellular GSH/GSSG ratio (Klatt, Molina, De Lacoba et al 199 9) Similarly, NO• modulates AP-1 DNAbinding by reversible S-nitrosylation (Cys272 of c-Jun), but this modification is cell-type specific (Morris 199 5; Klatt, Molina, Lamas 199 9) Indirectly, ROS stimulates AP-1 activity... activation/↑NF-κB and AP-1 Vascular complication of diabetes Higai et al 2006 ↑NADPH oxidase/↑ROS/JNK1 and p38 MAPK activation/↑ c-Fos, c-Jun and JunB expression, ↑AP-1 activity Mitogenesis associated with atherosclerosis, aging, or cancer Rao et al 199 9 ↑ROS-RNS/↑AP-1 activity/↑MMP-2/cardiac remodeling Response to I/R injury Alfonso-Jaume et al 2006 ↑I/R-ROS/NF-κB and AP-1 activation/ICAM-1 upregulation/acute... 199 5 (Mulcahy, Gipp 199 5), the human GCLc promoter notably contained consensus sites for AP-1, AP-2, SP-1, and Nrf2 Four AREs are uncovered (Mulcahy, Wartman, Bailey et al 199 7), of which the most distal ARE4 is responsible for constitutive and β-naphthflavone (β-NF)–induced GCLc promoter activity; specifically, a TRE within ARE4 controls constitutive GCLc promoter activity (Wild, Gipp, Mulcahy 199 8)... and NO signaling functions Cell 102(4): 499 –5 09 Fachinger G, Deutsch U, Risau W 199 9 Functional interaction of vascular endothelial-protein-tyrosine phosphatase with the angiopoietin receptor Tie-2 Oncogene 18(43): 594 8– 595 3 Fan H, Sun B, Gu Q, Lafond-Walker A, Cao S, Becker LC 2002 Oxygen radicals trigger activation of NF-kappaB and AP-1 and upregulation of ICAM-1 in reperfused canine heart Am J Physiol... a +2 state) and sulfonic acid (–SO3H, a + 4 state) are 480 NOVEL CELLULAR PATHWAYS P S P P-SH S P-SH P-SOH GSH GSSG ROS (Disulfide bond formation) (Cys P-SO3H P-SO2H oxidation) Irreversible modifications P-SSG GSH (S-glutathionylation) GSNO RNS P-SH P-SNO (S-nitrosylation) Figure 19. 3 Redox modification of protein thiols with role in cellular signaling Cysteine residues from target proteins can undergo... on activation, ASK-1 signaling leads to the activation of p38 and JNK pathways (Nishitoh, Saitoh, Mochida et al 199 8) Chapter 19: Redox Signaling and Vascular Function ROS Trx1 ASK-1 Trx-ASK-1 redox sensor Trx1 ASK-1 TrxSS TRAF2/6 Activated ASK-1 signalosome ROS P ASK-1 TRAF2/6 ASK-1 TRAF2/6 Mitochondria Trx2 Mito ASK1 P TRAF2/6 MKKs JNK Cyt c Apoptosis↑ Figure 19. 4 ROS-mediated ASK-1 signaling and... necrosis factor-α, ionizing radiation, and ROS leads to NF-κB activation (Baeuerle 199 8) It is well recognized that NF-κB activation and DNA binding are sensitive to the cellular redox status For example, NF-κB/DNA binding is decreased by thiol oxidants such as diamide and increased by thiol-reducing compounds such as dithiothreitol and β-mercaptoethanol (Hayashi, Ueno, Okamoto 199 3) Cytosolic NF-κkB activation... dissociation/↑Nrf2/ARE-responsive genes Vascular proliferation Villacorta et al 2007 EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; HO-1, heme oxygenase 1; JNK, c-Jun N-terminal kinase; Keap1, Kelch-like ECH-associated protein 1; LNO2, nitro-linoleic acids; moxLDL, moderately oxidized LDL; MAPK, mitogen-activated protein kinase; NO, nitric oxide Table 19. 2 Cellular Pathways... inhibition of NF-κB activation (Hirota, Murata, Sachi et al 199 9) whereas within the nucleus, Trx1 reduces Cys62 of the p50 subunit of NF-κB, resulting in increased binding to DNA (Matthews, Wakasugi, Virelizier et al 199 2) S-glutathionylation is another mode of redox control of NF-κB activity S-glutathionylation of Cys62 of the p50 subunit has been shown to inhibit NF-κB binding to DNA (Pineda-Molina, Klatt,... endothelial surface expression of adhesion molecules (E-selectin, Chapter 19: Redox Signaling and Vascular Function ICAM-1 and VCAM-1) serve to tether circulating leukocytes to the endothelium which increased O2•– and H2O2 formation For instance, endothelial VCAM-1 and ICAM-1 cross-linking respectively stimulates Nox-dependent (Matheny, Deem, Cook-Mills 2000) and XO-dependent O2•– production (section VII.1) Additional . al. 199 9). Similarly, NO • modulates AP-1 DNA- binding by reversible S-nitrosylation (Cys 272 of c-Jun), but this modi cation is cell-type speci c (Morris 199 5; Klatt, Molina, Lamas 199 9) S-glutathionylation. In addition, GSNO can induce both S-nitrosylation and S-glutathionylation of protein thiols. P-SH P-SH P-SOH GSH GSSG ROS RNS GSH GSNO P-SNO P-SSG P-SO 2 H P-SO 3 H P-SH PS PS Irreversible modifications (Disulfide. Srx, sulfi redoxin; Trx, thioredoxin. Exo- and endogenous sources NOS ONOO - GSH P-SH GSNO GSH GSH P-SH P-SNO P-SSG P-SH Trx, Grx, GSH, Srx Cys-oxidation (P-SO n H) Nitrosative stress NO SOD CAT GSH H 2 O 2 HO• GSSG GPx GR NADP + Fe 3+ Fe 2+ NADPH Glucose O 2 +

Ngày đăng: 08/08/2014, 13:21

Tài liệu cùng người dùng

Tài liệu liên quan