Skip to main content

Advertisement

Log in

Role of SIRT1 in autoimmune demyelination and neurodegeneration

  • Published:
Immunologic Research Aims and scope Submit manuscript

Abstract

Multiple sclerosis (MS) is a demyelinating disease characterized by chronic inflammation of the central nervous system, in which many factors can act together to influence disease susceptibility and progression. SIRT1 is a member of the histone deacetylase class III family of proteins and is an NAD+-dependent histone and protein deacetylase. SIRT1 can induce chromatin silencing through the deacetylation of histones and plays an important role as a key regulator of a wide variety of cellular and physiological processes including DNA damage, cell survival, metabolism, aging, and neurodegeneration. It has gained a lot of attention recently because many studies in animal models of demyelinating and neurodegenerative diseases have shown that SIRT1 induction can ameliorate the course of the disease. SIRT1 expression was found to be decreased in the peripheral blood mononuclear cells of MS patients during relapses. SIRT1 represents a possible biomarker of relapses and a potential new target for therapeutic intervention in MS. Modulation of SIRT1 may be a valuable strategy for treating or preventing MS and neurodegenerative central nervous system disorders.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Kuo MH, Allis CD. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays. 1998;20:615–26.

    CAS  PubMed  Google Scholar 

  2. Brandl A, Heinzel T, Kramer OH. Histone deacetylases: salesmen and customers in the post-translational modification market. Biol Cell. 2009;101:193–205.

    CAS  PubMed  Google Scholar 

  3. Kruszewski M, Szumiel I. Sirtuins (histone deacetylases III) in the cellular response to DNA damage—facts and hypotheses. DNA Repair (Amst). 2005;4:1306–13.

    CAS  Google Scholar 

  4. Khan O, La Thangue NB. HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol Cell Biol. 2012;90:85–94.

    CAS  PubMed  Google Scholar 

  5. Deng CX. SIRT1, is it a tumor promoter or tumor suppressor? Int J Biol Sci. 2009;5:147–52.

    PubMed Central  CAS  PubMed  Google Scholar 

  6. Lazo-Gomez R, Ramirez-Jarquin UN, Tovar YRLB, Tapia R. Histone deacetylases and their role in motor neuron degeneration. Front Cell Neurosci. 2013;7:243.

    PubMed Central  PubMed  Google Scholar 

  7. Turner BM. Histone acetylation and an epigenetic code. Bioessays. 2000;22:836–45.

    CAS  PubMed  Google Scholar 

  8. Barneda-Zahonero B, Parra M. Histone deacetylases and cancer. Mol Oncol. 2012;6:579–89.

    CAS  PubMed  Google Scholar 

  9. Delcuve GP, Khan DH, Davie JR. Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenet. 2012;4:5.

    CAS  Google Scholar 

  10. Sengupta N, Seto E. Regulation of histone deacetylase activities. J Cell Biochem. 2004;93:57–67.

    CAS  PubMed  Google Scholar 

  11. Smith JS, Brachmann CB, Celic I, Kenna MA, Muhammad S, Starai VJ, Avalos JL, Escalante-Semerena JC, Grubmeyer C, Wolberger C, Boeke JD. A phylogenetically conserved NAD+-dependent protein deacetylase activity in the Sir2 protein family. Proc Natl Acad Sci USA. 2000;97:6658–63.

    PubMed Central  CAS  PubMed  Google Scholar 

  12. Vlaicu SI, Tegla CA, Cudrici CD, Fosbrink M, Nguyen V, Azimzadeh P, Rus V, Chen H, Mircea PA, Shamsuddin A, Rus H. Epigenetic modifications induced by RGC-32 in colon cancer. Exp Mol Pathol. 2010;88:67–76.

    PubMed Central  CAS  PubMed  Google Scholar 

  13. Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, Janle EM, Lobo J, Ferruzzi MG, Davies P, Marambaud P. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J Biol Chem. 2010;285:9100–13.

    PubMed Central  CAS  PubMed  Google Scholar 

  14. Shore D, Squire M, Nasmyth KA. Characterization of two genes required for the position-effect control of yeast mating-type genes. EMBO J. 1984;3:2817–23.

    PubMed Central  CAS  PubMed  Google Scholar 

  15. Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999;13:2570–80.

    PubMed Central  CAS  PubMed  Google Scholar 

  16. Rine J, Herskowitz I. Four genes responsible for a position effect on expression from HML and HMR in Saccharomyces cerevisiae. Genetics. 1987;116:9–22.

    PubMed Central  CAS  PubMed  Google Scholar 

  17. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003;425:191–6.

    CAS  PubMed  Google Scholar 

  18. Frankel S, Ziafazeli T, Rogina B. dSir2 and longevity in Drosophila. Exp Gerontol. 2011;46:391–6.

    PubMed Central  CAS  PubMed  Google Scholar 

  19. Balcerczyk A, Pirola L. Therapeutic potential of activators and inhibitors of sirtuins. Biofactors. 2010;36:383–93.

    CAS  PubMed  Google Scholar 

  20. Klar AJ, Fogel S, Macleod K. MAR1-a regulator of the HMa and HMalpha loci in Saccharomyces cerevisiae. Genetics. 1979;93:37–50.

    PubMed Central  CAS  PubMed  Google Scholar 

  21. Baur JA. Biochemical effects of SIRT1 activators. Biochim Biophys Acta (BBA)-Proteins Proteomics. 2010;1804:1626–34.

    CAS  Google Scholar 

  22. Frye RA. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem Biophys Res Commun. 2000;273:793–8.

    CAS  PubMed  Google Scholar 

  23. Frye RA. Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity. Biochem Biophys Res Commun. 1999;260:273–9.

    CAS  PubMed  Google Scholar 

  24. Gray SG, Dangond F. Rationale for the use of histone deacetylase inhibitors as a dual therapeutic modality in multiple sclerosis. Epigenetics. 2006;1:67–75.

    PubMed  Google Scholar 

  25. Tanno M, Sakamoto J, Miura T, Shimamoto K, Horio Y. Nucleocytoplasmic shuttling of the NAD+-dependent histone deacetylase SIRT1. J Biol Chem. 2007;282:6823–32.

    CAS  PubMed  Google Scholar 

  26. Badea TC, Niculescu FI, Soane L, Shin ML, Rus H. Molecular cloning and characterization of RGC-32, a novel gene induced by complement activation in oligodendrocytes. J Biol Chem. 1998;273:26977–81.

    CAS  PubMed  Google Scholar 

  27. Burgoon MP, Gilden DH, Owens GP. B cells in multiple sclerosis. Front Biosci. 2004;9:786–96.

    PubMed Central  CAS  PubMed  Google Scholar 

  28. Yuan H, Su L, Chen WY. The emerging and diverse roles of sirtuins in cancer: a clinical perspective. Onco Targets Ther. 2013;6:1399–416.

    PubMed Central  CAS  PubMed  Google Scholar 

  29. Paredes S, Villanova L, Chua KF. Molecular pathways: emerging roles of mammalian sirtuin SIRT7 in cancer. Clin Cancer Res. 2014;20:1741–6.

  30. Grozinger CM, Chao ED, Blackwell HE, Moazed D, Schreiber SL. Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J Biol Chem. 2001;276:38837–43.

    CAS  PubMed  Google Scholar 

  31. Chen D, Bruno J, Easlon E, Lin SJ, Cheng HL, Alt FW, Guarente L. Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. 2008;22:1753–7.

    PubMed Central  CAS  PubMed  Google Scholar 

  32. Zu Y, Liu L, Lee MY, Xu C, Liang Y, Man RY, Vanhoutte PM, Wang Y. SIRT1 promotes proliferation and prevents senescence through targeting LKB1 in primary porcine aortic endothelial cells. Circ Res. 2010;106:1384–93.

    CAS  PubMed  Google Scholar 

  33. Wang RH, Sengupta K, Li C, Kim HS, Cao L, Xiao C, Kim S, Xu X, Zheng Y, Chilton B, Jia R, Zheng ZM, Appella E, Wang XW, Ried T, Deng CX. Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice. Cancer Cell. 2008;14:312–23.

    PubMed Central  CAS  PubMed  Google Scholar 

  34. Kim HS, Vassilopoulos A, Wang RH, Lahusen T, Xiao Z, Xu X, Li C, Veenstra TD, Li B, Yu H, Ji J, Wang XW, Park SH, Cha YI, Gius D, Deng CX. SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity. Cancer Cell. 2011;20:487–99.

    PubMed Central  CAS  PubMed  Google Scholar 

  35. Kim HS, Patel K, Muldoon-Jacobs K, Bisht KS, Aykin-Burns N, Pennington JD, van der Meer R, Nguyen P, Savage J, Owens KM, Vassilopoulos A, Ozden O, Park SH, Singh KK, Abdulkadir SA, Spitz DR, Deng CX, Gius D. SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell. 2010;17:41–52.

    PubMed Central  CAS  PubMed  Google Scholar 

  36. Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, Puigserver P, Sinclair DA, Tsai LH. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J. 2007;26:3169–79.

    PubMed Central  CAS  PubMed  Google Scholar 

  37. Webster BR, Lu Z, Sack MN, Scott I. The role of sirtuins in modulating redox stressors. Free Radic Biol Med. 2012;52:281–90.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Hubbard BP, Gomes AP, Dai H, Li J, Case AW, Considine T, Riera TV, Lee JE, Yen ES, Lamming DW, Pentelute BL, Schuman ER, Stevens LA, Ling AJ, Armour SM, Michan S, Zhao H, Jiang Y, Sweitzer SM, Blum CA, Disch JS, Ng PY, Howitz KT, Rolo AP, Hamuro Y, Moss J, Perni RB, Ellis JL, Vlasuk GP, Sinclair DA. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science. 2013;339:1216–9.

    PubMed Central  CAS  PubMed  Google Scholar 

  39. Gibson BA, Kraus WL. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat Rev Mol Cell Biol. 2012;13:411–24.

    CAS  PubMed  Google Scholar 

  40. Virag L. Structure and function of poly(ADP-ribose) polymerase-1: role in oxidative stress-related pathologies. Curr Vasc Pharmacol. 2005;3:209–14.

    CAS  PubMed  Google Scholar 

  41. Nasrin N, Kaushik VK, Fortier E, Wall D, Pearson KJ, de Cabo R, Bordone L. JNK1 phosphorylates SIRT1 and promotes its enzymatic activity. PLoS One. 2009;4:e8414.

    PubMed Central  PubMed  Google Scholar 

  42. Vinciguerra M, Santini MP, Martinez C, Pazienza V, Claycomb WC, Giuliani A, Rosenthal N. mIGF-1/JNK1/SirT1 signaling confers protection against oxidative stress in the heart. Aging Cell. 2012;11:139–49.

    CAS  PubMed  Google Scholar 

  43. Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab. 2014;25:138–45.

    CAS  PubMed  Google Scholar 

  44. Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, Hong HK, Chong JL, Buhr ED, Lee C, Takahashi JS, Imai S, Bass J. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009;324:651–4.

    PubMed Central  CAS  PubMed  Google Scholar 

  45. Vaziri H, Dessain SK, Eaton EN, Imai SI, Frye RA, Pandita TK, Guarente L, Weinberg RA. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–59.

    CAS  PubMed  Google Scholar 

  46. Chong ZZ, Lin SH, Maiese K. The NAD+ precursor nicotinamide governs neuronal survival during oxidative stress through protein kinase B coupled to FOXO3a and mitochondrial membrane potential. J Cereb Blood Flow Metab. 2004;24:728–43.

    CAS  PubMed  Google Scholar 

  47. Tanner KG, Landry J, Sternglanz R, Denu JM. Silent information regulator 2 family of NAD- dependent histone/protein deacetylases generates a unique product, 1-O-acetyl-ADP-ribose. Proc Natl Acad Sci USA. 2000;97:14178–82.

    PubMed Central  CAS  PubMed  Google Scholar 

  48. Hubbard BP, Sinclair DA. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci. 2014;35:146–54.

    CAS  PubMed  Google Scholar 

  49. Tegla CA, Azimzadeh P, Andrian-Albescu M, Martin A, Cudrici CD, Trippe R III, Sugarman A, Chen H, Boodhoo D, Vlaicu SI, Royal W III, Bever C, Rus V, Rus H. SIRT1 is decreased during relapses in patients with multiple sclerosis. Exp Mol Pathol. 2014;96:139–48.

    CAS  PubMed  Google Scholar 

  50. Oberdoerffer P, Michan S, McVay M, Mostoslavsky R, Vann J, Park SK, Hartlerode A, Stegmuller J, Hafner A, Loerch P, Wright SM, Mills KD, Bonni A, Yankner BA, Scully R, Prolla TA, Alt FW, Sinclair DA. SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell. 2008;135:907–18.

    PubMed Central  CAS  PubMed  Google Scholar 

  51. Sauve AA, Celic I, Avalos J, Deng H, Boeke JD, Schramm VL. Chemistry of gene silencing: the mechanism of NAD+-dependent deacetylation reactions. Biochemistry. 2001;40:15456–63.

    CAS  PubMed  Google Scholar 

  52. Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem. 2002;277:45099–107.

    CAS  PubMed  Google Scholar 

  53. Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012;13:225–38.

    CAS  PubMed  Google Scholar 

  54. Zschoernig B, Mahlknecht U. SIRTUIN 1: regulating the regulator. Biochem Biophys Res Commun. 2008;376:251–5.

    CAS  PubMed  Google Scholar 

  55. Wang C, Chen L, Hou X, Li Z, Kabra N, Ma Y, Nemoto S, Finkel T, Gu W, Cress WD, Chen J. Interactions between E2F1 and SirT1 regulate apoptotic response to DNA damage. Nat Cell Biol. 2006;8:1025–31.

    CAS  PubMed  Google Scholar 

  56. Ng F, Tang BL. Sirtuins’ modulation of autophagy. J Cell Physiol. 2013;228:2262–70.

    CAS  PubMed  Google Scholar 

  57. Xiong S, Salazar G, Patrushev N, Alexander RW. FoxO1 mediates an autofeedback loop regulating SIRT1 expression. J Biol Chem. 2011;286:5289–99.

    PubMed Central  CAS  PubMed  Google Scholar 

  58. Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science. 2004;306:2105–8.

    CAS  PubMed  Google Scholar 

  59. Sasaki T, Maier B, Koclega KD, Chruszcz M, Gluba W, Stukenberg PT, Minor W, Scrable H. Phosphorylation regulates SIRT1 function. PLoS One. 2008;3:e4020.

    PubMed Central  PubMed  Google Scholar 

  60. Revollo JR, Li X. The ways and means that fine tune Sirt1 activity. Trends Biochem Sci. 2013;38:160–7.

    PubMed Central  CAS  PubMed  Google Scholar 

  61. North BJ, Verdin E. Mitotic regulation of SIRT2 by cyclin-dependent kinase 1-dependent phosphorylation. J Biol Chem. 2007;282:19546–55.

    CAS  PubMed  Google Scholar 

  62. Guo X, Williams JG, Schug TT, Li X. DYRK1A and DYRK3 promote cell survival through phosphorylation and activation of SIRT1. J Biol Chem. 2010;285:13223–32.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. Hou J, Chong ZZ, Shang YC, Maiese K. Early apoptotic vascular signaling is determined by Sirt1 through nuclear shuttling, forkhead trafficking, bad, and mitochondrial caspase activation. Curr Neurovasc Res. 2010;7:95–112.

    PubMed Central  CAS  PubMed  Google Scholar 

  64. Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795–800.

    CAS  PubMed  Google Scholar 

  65. Hou J, Wang S, Shang YC, Chong ZZ, Maiese K. Erythropoietin employs cell longevity pathways of SIRT1 to foster endothelial vascular integrity during oxidant stress. Curr Neurovasc Res. 2011;8:220–35.

    PubMed Central  CAS  PubMed  Google Scholar 

  66. Yang Y, Fu W, Chen J, Olashaw N, Zhang X, Nicosia SV, Bhalla K, Bai W. SIRT1 sumoylation regulates its deacetylase activity and cellular response to genotoxic stress. Nat Cell Biol. 2007;9:1253–62.

    PubMed Central  CAS  PubMed  Google Scholar 

  67. Kim EJ, Kho JH, Kang MR, Um SJ. Active regulator of SIRT1 cooperates with SIRT1 and facilitates suppression of p53 activity. Mol Cell. 2007;28:277–90.

    CAS  PubMed  Google Scholar 

  68. Kim JE, Chen J, Lou Z. DBC1 is a negative regulator of SIRT1. Nature. 2008;451:583–6.

    CAS  PubMed  Google Scholar 

  69. Alcain FJ, Villalba JM. Sirtuin activators. Expert Opin Ther Pat. 2009;19:403–14.

    CAS  PubMed  Google Scholar 

  70. Alcain FJ, Villalba JM. Sirtuin inhibitors. Expert Opin Ther Pat. 2009;19:283–94.

    CAS  PubMed  Google Scholar 

  71. Chan MM. Antimicrobial effect of resveratrol on dermatophytes and bacterial pathogens of the skin. Biochem Pharmacol. 2002;63:99–104.

    CAS  PubMed  Google Scholar 

  72. Ramadori G, Lee CE, Bookout AL, Lee S, Williams KW, Anderson J, Elmquist JK, Coppari R. Brain SIRT1: anatomical distribution and regulation by energy availability. J Neurosci. 2008;28:9989–96.

    PubMed Central  CAS  PubMed  Google Scholar 

  73. Zhang F, Wang S, Gan L, Vosler PS, Gao Y, Zigmond MJ, Chen J. Protective effects and mechanisms of sirtuins in the nervous system. Prog Neurobiol. 2011;95:373–95.

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Donmez G. The neurobiology of sirtuins and their role in neurodegeneration. Trends Pharmacol Sci. 2012;33:494–501.

    CAS  PubMed  Google Scholar 

  75. Tang BL, Chua CEL. SIRT1 and neuronal diseases. Mol Asp Med. 2008;29:187–200.

    CAS  Google Scholar 

  76. Sakamoto J, Miura T, Shimamoto K, Horio Y. Predominant expression of Sir2alpha, an NAD-dependent histone deacetylase, in the embryonic mouse heart and brain. FEBS Lett. 2004;556:281–6.

    CAS  PubMed  Google Scholar 

  77. Hisahara S, Chiba S, Matsumoto H, Tanno M, Yagi H, Shimohama S, Sato M, Horio Y. Histone deacetylase SIRT1 modulates neuronal differentiation by its nuclear translocation. Proc Natl Acad Sci USA. 2008;105:15599–604.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. Nakae J, Cao Y, Daitoku H, Fukamizu A, Ogawa W, Yano Y, Hayashi Y. The LXXLL motif of murine forkhead transcription factor FoxO1 mediates Sirt1-dependent transcriptional activity. J Clin Investig. 2006;116:2473–83.

    PubMed Central  CAS  PubMed  Google Scholar 

  79. Araki T, Sasaki Y, Milbrandt J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science. 2004;305:1010–3.

    CAS  PubMed  Google Scholar 

  80. Gan L, Qiao S, Lan X, Chi L, Luo C, Lien L, Yan Liu Q, Liu R. Neurogenic responses to amyloid-beta plaques in the brain of Alzheimer’s disease-like transgenic (pPDGF-APPSw, Ind) mice. Neurobiol Dis. 2008;29:71–80.

    PubMed Central  CAS  PubMed  Google Scholar 

  81. Luo J, Su F, Chen D, Shiloh A, Gu W. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature. 2000;408:377–81.

    CAS  PubMed  Google Scholar 

  82. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, Mayo MW. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–80.

    PubMed Central  CAS  PubMed  Google Scholar 

  83. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M, Guarente L. Mammalian SIRT1 represses forkhead transcription factors. Cell. 2004;116:551–63.

    CAS  PubMed  Google Scholar 

  84. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA, Alt FW, Greenberg ME. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 2004;303:2011–5.

    CAS  PubMed  Google Scholar 

  85. Herskovits AZ, Guarente L. Sirtuin deacetylases in neurodegenerative diseases of aging. Cell Res. 2013;23:746–58.

    PubMed Central  CAS  PubMed  Google Scholar 

  86. Alzheimer’s Association. 2012 Alzheimer’s disease facts and figures. Alzheimers Dement. 2012;8:131–68.

    Google Scholar 

  87. Castellani RJ, Rolston RK, Smith MA. Alzheimer disease. Dis Mon. 2010;56:484–546.

    PubMed Central  PubMed  Google Scholar 

  88. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6.

    CAS  PubMed  Google Scholar 

  89. Wirths O, Multhaup G, Bayer TA. A modified beta-amyloid hypothesis: intraneuronal accumulation of the beta-amyloid peptide—the first step of a fatal cascade. J Neurochem. 2004;91:513–20.

    CAS  PubMed  Google Scholar 

  90. Bell KF, Ducatenzeiler A, Ribeiro-da-Silva A, Duff K, Bennett DA, Cuello AC. The amyloid pathology progresses in a neurotransmitter-specific manner. Neurobiol Aging. 2006;27:1644–57.

    CAS  PubMed  Google Scholar 

  91. Jang JH, Surh YJ. Protective effect of resveratrol on beta-amyloid-induced oxidative PC12 cell death. Free Radic Biol Med. 2003;34:1100–10.

    CAS  PubMed  Google Scholar 

  92. Conte A, Pellegrini S, Tagliazucchi D. Synergistic protection of PC12 cells from beta-amyloid toxicity by resveratrol and catechin. Brain Res Bull. 2003;62:29–38.

    CAS  PubMed  Google Scholar 

  93. Han YS, Zheng WH, Bastianetto S, Chabot JG, Quirion R. Neuroprotective effects of resveratrol against beta-amyloid-induced neurotoxicity in rat hippocampal neurons: involvement of protein kinase C. Br J Pharmacol. 2004;141:997–1005.

    PubMed Central  CAS  PubMed  Google Scholar 

  94. Karuppagounder SS, Pinto JT, Xu H, Chen HL, Beal MF, Gibson GE. Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer’s disease. Neurochem Int. 2009;54:111–8.

    PubMed Central  CAS  PubMed  Google Scholar 

  95. Anekonda TS, Reddy PH. Neuronal protection by sirtuins in Alzheimer’s disease. J Neurochem. 2006;96:305–13.

    CAS  PubMed  Google Scholar 

  96. Min SW, Sohn PD, Cho SH, Swanson RA, Gan L. Sirtuins in neurodegenerative diseases: an update on potential mechanisms. Front Aging Neurosci. 2013;5:53.

    PubMed Central  PubMed  Google Scholar 

  97. Julien C, Tremblay C, Emond V, Lebbadi M, Salem N Jr, Bennett DA, Calon F. Sirtuin 1 reduction parallels the accumulation of tau in Alzheimer disease. J Neuropathol Exp Neurol. 2009;68:48–58.

    PubMed Central  CAS  PubMed  Google Scholar 

  98. de Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5:525–35.

    PubMed  Google Scholar 

  99. Blesa J, Phani S, Jackson-Lewis V, Przedborski S. Classic and new animal models of Parkinson’s disease. J Biomed Biotechnol. 2012;2012:845618.

    PubMed Central  PubMed  Google Scholar 

  100. Wu Y, Li X, Zhu JX, Xie W, Le W, Fan Z, Jankovic J, Pan T. Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson’s disease. Neurosignals. 2011;19:163–74.

    PubMed Central  CAS  PubMed  Google Scholar 

  101. van Ham TJ, Thijssen KL, Breitling R, Hofstra RM, Plasterk RH, Nollen EA. C. elegans model identifies genetic modifiers of alpha-synuclein inclusion formation during aging. PLoS Genet. 2008;4:e1000027.

    PubMed Central  PubMed  Google Scholar 

  102. Mudo G, Makela J, Di Liberto V, Tselykh TV, Olivieri M, Piepponen P, Eriksson O, Malkia A, Bonomo A, Kairisalo M, Aguirre JA, Korhonen L, Belluardo N, Lindholm D. Transgenic expression and activation of PGC-1alpha protect dopaminergic neurons in the MPTP mouse model of Parkinson’s disease. Cell Mol Life Sci. 2012;69:1153–65.

    CAS  PubMed  Google Scholar 

  103. Donmez G, Arun A, Chung CY, McLean PJ, Lindquist S, Guarente L. SIRT1 protects against alpha-synuclein aggregation by activating molecular chaperones. J Neurosci. 2012;32:124–32.

    PubMed Central  CAS  PubMed  Google Scholar 

  104. Sepers MD, Raymond LA. Mechanisms of synaptic dysfunction and excitotoxicity in Huntington’s disease. Drug Discov Today. 2014;19:990–6.

  105. Chen Y, Carter RL, Cho IK, Chan AW. Cell-based therapies for Huntington’s disease. Drug Discov Today. 2014;19:980–4.

  106. Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell. 2006;127:59–69.

    CAS  PubMed  Google Scholar 

  107. Jeong H, Cohen DE, Cui L, Supinski A, Savas JN, Mazzulli JR, Yates JR III, Bordone L, Guarente L, Krainc D. Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat Med. 2011;18:159–65.

    PubMed  Google Scholar 

  108. Jiang M, Wang J, Fu J, Du L, Jeong H, West T, Xiang L, Peng Q, Hou Z, Cai H, Seredenina T, Arbez N, Zhu S, Sommers K, Qian J, Zhang J, Mori S, Yang XW, Tamashiro KL, Aja S, Moran TH, Luthi-Carter R, Martin B, Maudsley S, Mattson MP, Cichewicz RH, Ross CA, Holtzman DM, Krainc D, Duan W. Neuroprotective role of Sirt1 in mammalian models of Huntington’s disease through activation of multiple Sirt1 targets. Nat Med. 2011;18:153–8.

    PubMed  Google Scholar 

  109. Parker JA, Vazquez-Manrique RP, Tourette C, Farina F, Offner N, Mukhopadhyay A, Orfila AM, Darbois A, Menet S, Tissenbaum HA, Neri C. Integration of beta-catenin, sirtuin, and FOXO signaling protects from mutant huntingtin toxicity. J Neurosci. 2012;32:12630–40.

    PubMed Central  CAS  PubMed  Google Scholar 

  110. Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502–17.

    CAS  PubMed  Google Scholar 

  111. Frohman EM, Racke MK, Raine CS. Multiple sclerosis—the plaque and its pathogenesis. N Engl J Med. 2006;354:942–55.

    CAS  PubMed  Google Scholar 

  112. Tegla CA, Cudrici C, Rus V, Ito T, Vlaicu S, Singh A, Rus H. Neuroprotective effects of the complement terminal pathway during demyelination: implications for oligodendrocyte survival. J Neuroimmunol. 2009;213:3–11.

    PubMed Central  CAS  PubMed  Google Scholar 

  113. Rangachari M, Kuchroo VK. Using EAE to better understand principles of immune function and autoimmune pathology. J Autoimmun. 2013;45:31–9.

    PubMed Central  CAS  PubMed  Google Scholar 

  114. Michan S, Sinclair D. Sirtuins in mammals: insights into their biological function. Biochem J. 2007;404:1–13.

    PubMed Central  CAS  PubMed  Google Scholar 

  115. Shindler KS, Ventura E, Rex TS, Elliott P, Rostami A. SIRT1 activation confers neuroprotection in experimental optic neuritis. Investig Ophthalmol Vis Sci. 2007;48:3602–9.

    Google Scholar 

  116. Prozorovski T, Schulze-Topphoff U, Glumm R, Baumgart J, Schröter F, Ninnemann O, Siegert E, Bendix I, Brüstle O, Nitsch R, Zipp F, Aktas O. Sirt1 contributes critically to the redox-dependent fate of neural progenitors. Nat Cell Biol. 2008;10:385–94.

    CAS  PubMed  Google Scholar 

  117. Shindler KS, Ventura E, Dutt M, Elliott P, Fitzgerald DC, Rostami A. Oral resveratrol reduces neuronal damage in a model of multiple sclerosis. J Neuro-Ophthalmol. 2010;30:328–39.

    Google Scholar 

  118. Fonseca-Kelly Z, Nassrallah M, Uribe J, Khan RS, Dine K, Dutt M, Shindler KS. Resveratrol neuroprotection in a chronic mouse model of multiple sclerosis. Front Neurol. 2012;3:84.

    PubMed Central  PubMed  Google Scholar 

  119. Imler TJ, Petro TM. Decreased severity of experimental autoimmune encephalomyelitis during resveratrol administration is associated with increased IL-17+IL-10+ T cells, CD4− IFN-γ+ cells, and decreased macrophage IL-6 expression. Int Immunopharmacol. 2009;9:134–43.

    CAS  PubMed  Google Scholar 

  120. Singh NP, Hegde VL, Hofseth LJ, Nagarkatti M, Nagarkatti P. Resveratrol (trans-3,5,4′-trihydroxystilbene) ameliorates experimental allergic encephalomyelitis, primarily via induction of apoptosis in T cells involving activation of aryl hydrocarbon receptor and estrogen receptor. Mol Pharmacol. 2007;72:1508–21.

    CAS  PubMed  Google Scholar 

  121. Nimmagadda VK, Bever CT, Vattikunta NR, Talat S, Ahmad V, Nagalla NK, Trisler D, Judge SIV, Royal W, Chandrasekaran K, Russell JW, Makar TK. Overexpression of SIRT1 protein in neurons protects against experimental autoimmune encephalomyelitis through activation of multiple SIRT1 targets. J Immunol. 2013;190:4595–607.

    PubMed Central  CAS  PubMed  Google Scholar 

  122. Vaquero A, Scher M, Lee D, Erdjument-Bromage H, Tempst P, Reinberg D. Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol Cell. 2004;16:93–105.

    CAS  PubMed  Google Scholar 

  123. Vlaicu SI, Tegla CA, Cudrici CD, Danoff J, Madani H, Sugarman A, Niculescu F, Mircea PA, Rus V, Rus H. Role of C5b-9 complement complex and response gene to complement-32 (RGC-32) in cancer. Immunol Res. 2013;56:109–21.

    CAS  PubMed  Google Scholar 

  124. Tegla CA, Cudrici CD, Azimzadeh P, Singh AK, Trippe R III, Khan A, Chen H, Andrian-Albescu M, Royal W III, Bever C, Rus V, Rus H. Dual role of response gene to complement-32 in multiple sclerosis. Exp Mol Pathol. 2013;94:17–28.

    CAS  PubMed  Google Scholar 

  125. Badea T, Niculescu F, Soane L, Fosbrink M, Sorana H, Rus V, Shin ML, Rus H. RGC-32 increases p34CDC2 kinase activity and entry of aortic smooth muscle cells into S-phase. J Biol Chem. 2002;277:502–8.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank Dr. Deborah McClellan for editing this manuscript. This work was supported in part by a Pilot Project PP 1422 from the MS Society (to H. R.) and a Veterans Administration Merit Award (BX 001458 to H. R.). The MS brain samples were obtained from the National Neurological Research Specimen Bank (West Los Angeles Veterans Administration Hospital, Los Angeles, CA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Horea Rus.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martin, A., Tegla, C.A., Cudrici, C.D. et al. Role of SIRT1 in autoimmune demyelination and neurodegeneration. Immunol Res 61, 187–197 (2015). https://doi.org/10.1007/s12026-014-8557-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12026-014-8557-5

Keywords

Navigation