DOI QR코드

DOI QR Code

Investigating the role of Sirtuins in cell reprogramming

  • Shin, Jaein (Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University) ;
  • Kim, Junyeop (Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University) ;
  • Park, Hanseul (Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University) ;
  • Kim, Jongpil (Laboratory of Stem Cells and Cell Reprogramming, Department of Biomedical Engineering (BKplus21 team), Dongguk University)
  • Received : 2018.07.05
  • Published : 2018.10.31

Abstract

Cell reprogramming has been considered a powerful technique in the regenerative medicine field. In addition to diverse its strengths, cell reprogramming technology also has several drawbacks generated during the process of reprogramming. Telomere shortening caused by the cell reprogramming process impedes the efficiency of cell reprogramming. Transcription factors used for reprogramming alter genomic contents and result in genetic mutations. Additionally, defective mitochondria functioning such as excessive mitochondrial fission leads to the limitation of pluripotency and ultimately reduces the efficiency of reprogramming. These problems including genomic instability and impaired mitochondrial dynamics should be resolved to apply cell reprograming in clinical research and to address efficiency and safety concerns. Sirtuin (NAD+-dependent histone deacetylase) has been known to control the chromatin state of the telomere and influence mitochondria function in cells. Recently, several studies reported that Sirtuins could control for genomic instability in cell reprogramming. Here, we review recent findings regarding the role of Sirtuins in cell reprogramming. And we propose that the manipulation of Sirtuins may improve defects that result from the steps of cell reprogramming.

Keywords

References

  1. Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676 https://doi.org/10.1016/j.cell.2006.07.024
  2. Rando TA and Chang HY (2012) Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 148, 46-57 https://doi.org/10.1016/j.cell.2012.01.003
  3. Yu J, Vodyanik MA, Smuga-Otto K et al (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920 https://doi.org/10.1126/science.1151526
  4. Blasco MA, Serrano M and Fernandez-Capetillo O (2011) Genomic instability in iPS: time for a break. EMBO J 30, 991-993 https://doi.org/10.1038/emboj.2011.50
  5. Suva ML, Riggi N and Bernstein BE (2013) Epigenetic reprogramming in cancer. Science 339, 1567-1570 https://doi.org/10.1126/science.1230184
  6. Ruiz S, Panopoulos AD, Herrerias A et al (2011) A high proliferation rate is required for cell reprogramming and maintenance of human embryonic stem cell identity. Curr Biol 21, 45-52 https://doi.org/10.1016/j.cub.2010.11.049
  7. Lopez-Otin C, Blasco MA, Partridge L, Serrano M and Kroemer G (2013) The hallmarks of aging. Cell 153, 1194-1217 https://doi.org/10.1016/j.cell.2013.05.039
  8. Marion RM, Strati K, Li H et al (2009) Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 4, 141-154 https://doi.org/10.1016/j.stem.2008.12.010
  9. Weissbein U, Benvenisty N and Ben-David U (2014) Quality control: Genome maintenance in pluripotent stem cells. J Cell Biol 204, 153-163 https://doi.org/10.1083/jcb.201310135
  10. Folmes CD, Nelson TJ, Martinez-Fernandez A et al (2011) Somatic oxidative bioenergetics transitions into pluripotencydependent glycolysis to facilitate nuclear reprogramming. Cell Metab 14, 264-271 https://doi.org/10.1016/j.cmet.2011.06.011
  11. Banito A, Rashid ST, Acosta JC et al (2009) Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev 23, 2134-2139 https://doi.org/10.1101/gad.1811609
  12. Kimura H, Hayashi-Takanaka Y, Stasevich TJ and Sato Y (2015) Visualizing posttranslational and epigenetic modifications of endogenous proteins in vivo. Histochem Cell Biol 144, 101-109 https://doi.org/10.1007/s00418-015-1344-0
  13. Xiao Y and Chen J (2013) Proteomics approaches in the identification of molecular signatures of mesenchymal stem cells. Adv Biochem Eng Biotechnol 129, 153-176
  14. Mu WL, Wang YJ, Xu P et al (2015) Sox2 Deacetylation by Sirt1 Is Involved in Mouse Somatic Reprogramming. Stem Cells 33, 2135-2147 https://doi.org/10.1002/stem.2012
  15. Mostoslavsky R, Chua KF, Lombard DB et al (2006) Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 124, 315-329 https://doi.org/10.1016/j.cell.2005.11.044
  16. Laurent LC, Ulitsky I, Slavin I et al (2011) Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture. Cell Stem Cell 8, 106-118 https://doi.org/10.1016/j.stem.2010.12.003
  17. Taapken SM, Nisler BS, Newton MA et al (2011) Karotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells. Nat Biotechnol 29, 313-314 https://doi.org/10.1038/nbt.1835
  18. Araten DJ, Golde DW, Zhang RH et al (2005) A quantitative measurement of the human somatic mutation rate. Cancer Res 65, 8111-8117 https://doi.org/10.1158/0008-5472.CAN-04-1198
  19. Morin GB (1989) The human telomere terminal transferase enzyme is a ribonucleoprotein that synthesizes TTAGGG repeats. Cell 59, 521-529 https://doi.org/10.1016/0092-8674(89)90035-4
  20. Vaziri H, Chapman KB, Guigova A et al (2010) Spontaneous reversal of the developmental aging of normal human cells following transcriptional reprogramming. Regen Med 5, 345-363 https://doi.org/10.2217/rme.10.21
  21. Marion RM and Blasco MA (2010) Telomere rejuvenation during nuclear reprogramming. Curr Opin Genet Dev 20, 190-196 https://doi.org/10.1016/j.gde.2010.01.005
  22. Yehezkel S, Rebibo-Sabbah A, Segev Y et al (2011) Reprogramming of telomeric regions during the generation of human induced pluripotent stem cells and subsequent differentiation into fibroblast-like derivatives. Epigenetics 6, 63-75 https://doi.org/10.4161/epi.6.1.13390
  23. Pasi CE, Dereli-Oz A, Negrini S et al (2011) Genomic instability in induced stem cells. Cell Death Differ 18, 745-753 https://doi.org/10.1038/cdd.2011.9
  24. Lopez-Contreras AJ, Gutierrez-Martinez P, Specks J, Rodrigo-Perez S and Fernandez-Capetillo O (2012) An extra allele of Chk1 limits oncogene-induced replicative stress and promotes transformation. J Exp Med 209, 455-461 https://doi.org/10.1084/jem.20112147
  25. Ruiz S, Lopez-Contreras AJ, Gabut M et al (2015) Limiting replication stress during somatic cell reprogramming reduces genomic instability in induced pluripotent stem cells. Nat Commun 6, 8036 https://doi.org/10.1038/ncomms9036
  26. Mishra P and Chan DC (2014) Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15, 634-646 https://doi.org/10.1038/nrm3877
  27. Ji J, Sharma V, Qi S et al (2014) Antioxidant supplementation reduces genomic aberrations in human induced pluripotent stem cells. Stem Cell Reports 2, 44-51 https://doi.org/10.1016/j.stemcr.2013.11.004
  28. Facucho-Oliveira JM, Alderson J, Spikings EC, Egginton S and St John JC (2007) Mitochondrial DNA replication during differentiation of murine embryonic stem cells. J Cell Sci 120, 4025-4034 https://doi.org/10.1242/jcs.016972
  29. Moussaieff A, Rouleau M, Kitsberg D et al (2015) Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab 21, 392-402 https://doi.org/10.1016/j.cmet.2015.02.002
  30. Prigione A, Fauler B, Lurz R, Lehrach H and Adjaye J (2010) The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells 28, 721-733 https://doi.org/10.1002/stem.404
  31. Mandal S, Lindgren AG, Srivastava AS, Clark AT and Banerjee U (2011) Mitochondrial function controls proliferation and early differentiation potential of embryonic stem cells. Stem Cells 29, 486-495 https://doi.org/10.1002/stem.590
  32. Sathananthan H, Pera M and Trounson A (2002) The fine structure of human embryonic stem cells. Reprod Biomed Online 4, 56-61 https://doi.org/10.1016/S1472-6483(10)61916-5
  33. Han H, Irimia M, Ross PJ et al (2013) MBNL proteins repress ES-cell-specific alternative splicing and reprogramming. Nature 498, 241-245 https://doi.org/10.1038/nature12270
  34. Wang L, Ye X, Zhao Q et al (2014) Drp1 is dispensable for mitochondria biogenesis in induction to pluripotency but required for differentiation of embryonic stem cells. Stem Cells Dev 23, 2422-2434 https://doi.org/10.1089/scd.2014.0059
  35. Otera H and Mihara K (2011) Molecular mechanisms and physiologic functions of mitochondrial dynamics. J Biochem 149, 241-251 https://doi.org/10.1093/jb/mvr002
  36. Ramanathan A and Schreiber SL (2009) Direct control of mitochondrial function by mTOR. Proc Natl Acad Sci U S A 106, 22229-22232 https://doi.org/10.1073/pnas.0912074106
  37. Wang S, Xia P, Ye B, Huang G, Liu J and Fan Z (2013) Transient activation of autophagy via Sox2-mediated suppression of mTOR is an important early step in reprogramming to pluripotency. Cell Stem Cell 13, 617-625 https://doi.org/10.1016/j.stem.2013.10.005
  38. Greer EL, Dowlatshahi D, Banko MR et al (2007) An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol 17, 1646-1656 https://doi.org/10.1016/j.cub.2007.08.047
  39. Wang Z, Zang C, Rosenfeld JA et al (2008) Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet 40, 897-903 https://doi.org/10.1038/ng.154
  40. Lee S, Park JR, Seo MS et al (2009) Histone deacetylase inhibitors decrease proliferation potential and multilineage differentiation capability of human mesenchymal stem cells. Cell Prolif 42, 711-720 https://doi.org/10.1111/j.1365-2184.2009.00633.x
  41. Michishita E, McCord RA, Berber E et al (2008) SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature 452, 492-496 https://doi.org/10.1038/nature06736
  42. Chen T, Shen L, Yu J et al (2011) Rapamycin and other longevity-promoting compounds enhance the generation of mouse induced pluripotent stem cells. Aging Cell 10, 908-911 https://doi.org/10.1111/j.1474-9726.2011.00722.x
  43. Huangfu D, Maehr R, Guo W et al (2008) Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol 26, 795-797 https://doi.org/10.1038/nbt1418
  44. Vaquero A, Scher M, Lee D, Erdjument-Bromage H, Tempst P and Reinberg D (2004) Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol Cell 16, 93-105 https://doi.org/10.1016/j.molcel.2004.08.031
  45. Zhang ZN, Chung SK, Xu Z and Xu Y (2014) Oct4 maintains the pluripotency of human embryonic stem cells by inactivating p53 through Sirt1-mediated deacetylation. Stem Cells 32, 157-165 https://doi.org/10.1002/stem.1532
  46. Etchegaray JP, Chavez L, Huang Y et al (2015) The histone deacetylase SIRT6 controls embryonic stem cell fate via TET-mediated production of 5-hydroxymethylcytosine. Nat Cell Biol 17, 545-557 https://doi.org/10.1038/ncb3147
  47. Peng CH, Cherng JY, Chiou GY et al (2011) Delivery of Oct4 and SirT1 with cationic polyurethanes-short branch PEI to aged retinal pigment epithelium. Biomaterials 32, 9077-9088 https://doi.org/10.1016/j.biomaterials.2011.08.008
  48. Liu PY, Xu N, Malyukova A et al (2013) The histone deacetylase SIRT2 stabilizes Myc oncoproteins. Cell Death Differ 20, 503-514 https://doi.org/10.1038/cdd.2012.147
  49. Mao B, Zhao G, Lv X et al (2011) Sirt1 deacetylates c-Myc and promotes c-Myc/Max association. Int J Biochem Cell Biol 43, 1573-1581 https://doi.org/10.1016/j.biocel.2011.07.006
  50. Vaziri H, Dessain SK, Ng Eaton E et al (2001) hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 107, 149-159 https://doi.org/10.1016/S0092-8674(01)00527-X
  51. Han MK, Song EK, Guo Y, Ou X, Mantel C and Broxmeyer HE (2008) SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2, 241-251 https://doi.org/10.1016/j.stem.2008.01.002
  52. Katada S, Imhof A and Sassone-Corsi P (2012) Connecting threads: epigenetics and metabolism. Cell 148, 24-28 https://doi.org/10.1016/j.cell.2012.01.001
  53. Ryall JG, Dell'Orso S, Derfoul A et al (2015) The NAD(+)-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell 16, 171-183 https://doi.org/10.1016/j.stem.2014.12.004
  54. Zwaans BM and Lombard DB (2014) Interplay between sirtuins, MYC and hypoxia-inducible factor in cancerassociated metabolic reprogramming. Dis Model Mech 7, 1023-1032 https://doi.org/10.1242/dmm.016287
  55. Tennen RI, Bua DJ, Wright WE and Chua KF (2011) SIRT6 is required for maintenance of telomere position effect in human cells. Nat Commun 2, 433 https://doi.org/10.1038/ncomms1443
  56. Kugel S and Mostoslavsky R (2014) Chromatin and beyond: the multitasking roles for SIRT6. Trends Biochem Sci 39, 72-81 https://doi.org/10.1016/j.tibs.2013.12.002
  57. Michishita E, McCord RA, Boxer LD et al (2009) Cell cycle-dependent deacetylation of telomeric histone H3 lysine K56 by human SIRT6. Cell Cycle 8, 2664-2666 https://doi.org/10.4161/cc.8.16.9367
  58. Toiber D, Erdel F, Bouazoune K et al (2013) SIRT6 recruits SNF2H to DNA break sites, preventing genomic instability through chromatin remodeling. Mol Cell 51, 454-468 https://doi.org/10.1016/j.molcel.2013.06.018
  59. Galleano I, Schiedel M, Jung M, Madsen AS and Olsen CA (2016) A Continuous, Fluorogenic Sirtuin 2 Deacylase Assay: Substrate Screening and Inhibitor Evaluation. J Med Chem 59, 1021-1031 https://doi.org/10.1021/acs.jmedchem.5b01532
  60. Baur JA (2010) Biochemical effects of SIRT1 activators. Biochim Biophys Acta 1804, 1626-1634 https://doi.org/10.1016/j.bbapap.2009.10.025
  61. Gu XS, Wang ZB, Ye Z et al (2014) Resveratrol, an activator of SIRT1, upregulates AMPK and improves cardiac function in heart failure. Genet Mol Res 13, 323-335 https://doi.org/10.4238/2014.January.17.17
  62. Chaudhary N and Pfluger PT (2009) Metabolic benefits from Sirt1 and Sirt1 activators. Curr Opin Clin Nutr Metab Care 12, 431-437 https://doi.org/10.1097/MCO.0b013e32832cdaae
  63. Bruckbauer A, Zemel MB, Thorpe T et al (2012) Synergistic effects of leucine and resveratrol on insulin sensitivity and fat metabolism in adipocytes and mice. Nutr Metab (Lond) 9, 77 https://doi.org/10.1186/1743-7075-9-77
  64. Chauhan D, Bandi M, Singh AV et al (2011) Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells. Br J Haematol 155, 588-598 https://doi.org/10.1111/j.1365-2141.2011.08888.x
  65. Lahusen TJ and Deng CX (2015) SRT1720 induces lysosomal-dependent cell death of breast cancer cells. Mol Cancer Ther 14, 183-192
  66. Maya JD, Morello A, Repetto Y et al (2001) Trypanosoma cruzi: inhibition of parasite growth and respiration by oxazolo(thiazolo)pyridine derivatives and its relationship to redox potential and lipophilicity. Exp Parasitol 99, 1-6 https://doi.org/10.1006/expr.2001.4638
  67. Vu CB, Bemis JE, Disch JS et al (2009) Discovery of imidazo[1,2-b]thiazole derivatives as novel SIRT1 activators. J Med Chem 52, 1275-1283 https://doi.org/10.1021/jm8012954
  68. Bonkowski MS and Sinclair DA (2016) Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol 17, 679-690 https://doi.org/10.1038/nrm.2016.93
  69. Camins A, Sureda FX, Junyent F et al (2010) Sirtuin activators: designing molecules to extend life span. Biochim Biophys Acta 1799, 740-749 https://doi.org/10.1016/j.bbagrm.2010.06.005
  70. Botta G, De Santis LP and Saladino R (2012) Current advances in the synthesis and antitumoral activity of SIRT1-2 inhibitors by modulation of p53 and pro-apoptotic proteins. Curr Med Chem 19, 5871-5884 https://doi.org/10.2174/092986712804143303
  71. Villalba JM and Alcain FJ (2012) Sirtuin activators and inhibitors. Biofactors 38, 349-359 https://doi.org/10.1002/biof.1032
  72. Verdin E, Hirschey MD, Finley LW and Haigis MC (2010) Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem Sci 35, 669-675 https://doi.org/10.1016/j.tibs.2010.07.003
  73. Wang TT, Schoene NW, Kim EK and Kim YS (2013) Pleiotropic effects of the sirtuin inhibitor sirtinol involves concentration-dependent modulation of multiple nuclear receptor-mediated pathways in androgen-responsive prostate cancer cell LNCaP. Mol Carcinog 52, 676-685 https://doi.org/10.1002/mc.21906
  74. Peck B, Chen CY, Ho KK et al (2010) SIRT inhibitors induce cell death and p53 acetylation through targeting both SIRT1 and SIRT2. Mol Cancer Ther 9, 844-855 https://doi.org/10.1158/1535-7163.MCT-09-0971
  75. Naia L and Rego AC (2015) Sirtuins: double players in Huntington's disease. Biochim Biophys Acta 1852, 2183-2194 https://doi.org/10.1016/j.bbadis.2015.07.003
  76. Chen J, Zhou Y, Mueller-Steiner S et al (2005) SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem 280, 40364-40374 https://doi.org/10.1074/jbc.M509329200
  77. Risitano R, Curro M, Cirmi S et al (2014) Flavonoid fraction of Bergamot juice reduces LPS-induced inflammatory response through SIRT1-mediated NF-kappaB inhibition in THP-1 monocytes. PLoS One 9, e107431 https://doi.org/10.1371/journal.pone.0107431
  78. Polyakova O, Borman S, Grimley R, Vamathevan J, Hayes B and Solari R (2012) Identification of novel interacting partners of Sirtuin6. PLoS One 7, e51555 https://doi.org/10.1371/journal.pone.0051555
  79. Mopert K, Hajek P, Frank S, Chen C, Kaufmann J and Santel A (2009) Loss of Drp1 function alters OPA1 processing and changes mitochondrial membrane organization. Exp Cell Res 315, 2165-2180 https://doi.org/10.1016/j.yexcr.2009.04.016
  80. Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE and Chan DC (2003) Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160, 189-200 https://doi.org/10.1083/jcb.200211046
  81. Chen H, Chomyn A and Chan DC (2005) Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280, 26185-26192 https://doi.org/10.1074/jbc.M503062200
  82. Christiansen EG (1949) Orientation of the mitochondria during mitosis. Nature 163, 361
  83. Cho DH, Nakamura T and Lipton SA (2010) Mitochondrial dynamics in cell death and neurodegeneration. Cell Mol Life Sci 67, 3435-3447 https://doi.org/10.1007/s00018-010-0435-2
  84. Wilkerson DC and Sankar U (2011) Mitochondria: a sulfhydryl oxidase and fission GTPase connect mitochondrial dynamics with pluripotency in embryonic stem cells. Int J Biochem Cell Biol 43, 1252-1256 https://doi.org/10.1016/j.biocel.2011.05.005
  85. Cipolat S, Martins de Brito O, Dal Zilio B and Scorrano L (2004) OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad Sci U S A 101, 15927-15932 https://doi.org/10.1073/pnas.0407043101
  86. Son MJ, Kwon Y, Son MY et al (2015) Mitofusins deficiency elicits mitochondrial metabolic reprogramming to pluripotency. Cell Death Differ 22, 1957-1969 https://doi.org/10.1038/cdd.2015.43
  87. Tanno M, Sakamoto J, Miura T, Shimamoto K and Horio Y (2007) Nucleocytoplasmic shuttling of the NAD+-dependent histone deacetylase SIRT1. J Biol Chem 282, 6823-6832 https://doi.org/10.1074/jbc.M609554200
  88. Bell EL and Guarente L (2011) The SirT3 divining rod points to oxidative stress. Mol Cell 42, 561-568 https://doi.org/10.1016/j.molcel.2011.05.008
  89. Giralt A, Hondares E, Villena JA et al (2011) Peroxisome proliferator-activated receptor-gamma coactivator-1alpha controls transcription of the Sirt3 gene, an essential component of the thermogenic brown adipocyte phenotype. J Biol Chem 286, 16958-16966 https://doi.org/10.1074/jbc.M110.202390
  90. Olichon A, Baricault L, Gas N et al (2003) Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278, 7743-7746 https://doi.org/10.1074/jbc.C200677200
  91. Park SH, Ozden O, Jiang H et al (2011) Sirt3, mitochondrial ROS, ageing, and carcinogenesis. Int J Mol Sci 12, 6226-6239 https://doi.org/10.3390/ijms12096226
  92. Ou X, Lee MR, Huang X, Messina-Graham S and Broxmeyer HE (2014) SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells 32, 1183-1194 https://doi.org/10.1002/stem.1641
  93. Yoshii SR and Mizushima N (2015) Autophagy machinery in the context of mammalian mitophagy. Biochim Biophys Acta 1853, 2797-2801 https://doi.org/10.1016/j.bbamcr.2015.01.013
  94. Eiyama A and Okamoto K (2015) PINK1/Parkin-mediated mitophagy in mammalian cells. Curr Opin Cell Biol 33, 95-101 https://doi.org/10.1016/j.ceb.2015.01.002
  95. Koh H, Kim H, Kim MJ, Park J, Lee HJ and Chung J (2012) Silent information regulator 2 (Sir2) and Forkhead box O (FOXO) complement mitochondrial dysfunction and dopaminergic neuron loss in Drosophila PTEN-induced kinase 1 (PINK1) null mutant. J Biol Chem 287, 12750-12758 https://doi.org/10.1074/jbc.M111.337907
  96. Lim JH, Lee YM, Chun YS, Chen J, Kim JE and Park JW (2010) Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxia-inducible factor 1alpha. Mol Cell 38, 864-878 https://doi.org/10.1016/j.molcel.2010.05.023
  97. Qiu X, Brown K, Hirschey MD, Verdin E and Chen D (2010) Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab 12, 662-667 https://doi.org/10.1016/j.cmet.2010.11.015
  98. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A and Gupta MP (2009) Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest 119, 2758-2771
  99. Morigi M, Perico L, Rota C et al (2015) Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury. J Clin Invest 125, 715-726 https://doi.org/10.1172/JCI77632