DOI QR코드

DOI QR Code

Sirtuins in Cancer: a Balancing Act between Genome Stability and Metabolism

  • Jeong, Seung Min (Department of Biochemistry, College of Medicine, The Catholic University of Korea) ;
  • Haigis, Marcia C. (Department of Cell Biology, Harvard Medical School)
  • Received : 2015.06.12
  • Accepted : 2015.08.31
  • Published : 2015.09.30

Abstract

Genomic instability and altered metabolism are key features of most cancers. Recent studies suggest that metabolic reprogramming is part of a systematic response to cellular DNA damage. Thus, defining the molecules that fine-tune metabolism in response to DNA damage will enhance our understanding of molecular mechanisms of tumorigenesis and have profound implications for the development of strategies for cancer therapy. Sirtuins have been established as critical regulators in cellular homeostasis and physiology. Here, we review the emerging data revealing a pivotal function of sirtuins in genome maintenance and cell metabolism, and highlight current advances about the phenotypic consequences of defects in these critical regulators in tumorigenesis. While many questions should be addressed about the regulation and context-dependent functions of sirtuins, it appears clear that sirtuins may provide a promising, exciting new avenue for cancer therapy.

Keywords

References

  1. Abbas, T., and Dutta, A. (2009). p21 in cancer: intricate networks and multiple activities. Nat. Rev. Cancer 9, 400-414. https://doi.org/10.1038/nrc2657
  2. Abraham, R.T. (2001). Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev. 15, 2177-2196. https://doi.org/10.1101/gad.914401
  3. Ahn, B.H., Kim, H.S., Song, S., Lee, I.H., Liu, J., Vassilopoulos, A., Deng, C.X. and Finkel, T. (2008). A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc. Natl. Acad. Sci. USA 105, 14447-14452. https://doi.org/10.1073/pnas.0803790105
  4. Ahuja, N., Schwer, B., Carobbio, S., Waltregny, D., North, B.J., Castronovo, V., Maechler, P. and Verdin, E. (2007). Regulation of insulin secretion by SIRT4, a mitochondrial ADPribosyltransferase. J. Biol. Chem. 282, 33583-33592. https://doi.org/10.1074/jbc.M705488200
  5. Alexander, A., and Walker, C.L. (2011). The role of LKB1 and AMPK in cellular responses to stress and damage. FEBS Lett. 585, 952-957. https://doi.org/10.1016/j.febslet.2011.03.010
  6. Alhazzazi, T.Y., Kamarajan, P., Joo, N., Huang, J.Y., Verdin, E., D'Silva, N.J., and Kapila, Y.L. (2011). Sirtuin-3 (SIRT3)., a novel potential therapeutic target for oral cancer. Cancer 117, 1670-1678. https://doi.org/10.1002/cncr.25676
  7. Anderson, K.A., and Hirschey, M.D. (2012). Mitochondrial protein acetylation regulates metabolism. Essays Biochem. 52, 23-35. https://doi.org/10.1042/bse0520023
  8. Barber, M.F., Michishita-Kioi, E., Xi, Y., Tasselli, L., Kioi, M., Moqtaderi, Z., Tennen, R.I., Paredes, S., Young, N.L., Chen, K., et al. (2012). SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature 487, 114-118.
  9. Bauer, I., Grozio, A., Lasiglie, D., Basile, G., Sturla, L., Magnone, M., Sociali, G., Soncini, D., Caffa, I., Poggi, A., et al. (2012). The NAD+-dependent histone deacetylase SIRT6 promotes cytokine production and migration in pancreatic cancer cells by regulating Ca2+ responses. J. Biol. Chem. 287, 40924-40937. https://doi.org/10.1074/jbc.M112.405837
  10. Bell, E.L., 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
  11. Bell, E.L., Emerling, B.M., Ricoult, S.J., and Guarente, L. (2011). SirT3 suppresses hypoxia inducible factor 1alpha and tumor growth by inhibiting mitochondrial ROS production. Oncogene 30, 2986-2996. https://doi.org/10.1038/onc.2011.37
  12. Bordone, L., Motta, M.C., Picard, F., Robinson, A., Jhala, U.S., Apfeld, J., McDonagh, T., Lemieux, M., McBurney, M., Szilvasi, A., et al. (2006). Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 4, e31.
  13. Bradbury, C.A., Khanim, F.L., Hayden, R., Bunce, C.M., White, D.A., Drayson, M.T., Craddock, C., and Turner, B.M. (2005). Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia 19, 1751-1759. https://doi.org/10.1038/sj.leu.2403910
  14. Canto, C., Gerhart-Hines, Z., Feige, J.N., Lagouge, M., Noriega, L., Milne, J.C., Elliott, P.J., Puigserver, P., and Auwerx, J. (2009). AMPK regulates energy expenditure by modulating $NAD^+$ metabolism and SIRT1 activity. Nature 458, 1056-1060. https://doi.org/10.1038/nature07813
  15. Chen, J., Chan, A.W., To, K.F., Chen, W., Zhang, Z., Ren, J., Song, C., Cheung, Y.S., Lai, P.B., Cheng, S.H., et al. (2013). SIRT2 overexpression in hepatocellular carcinoma mediates epithelial to mesenchymal transition by protein kinase B/glycogen synthase kinase-3beta/beta-catenin signaling. Hepatology 57, 2287-2298. https://doi.org/10.1002/hep.26278
  16. Ciccia, A., and Elledge, S.J. (2010). The DNA damage response: making it safe to play with knives. Mol. Cell 40, 179-204. https://doi.org/10.1016/j.molcel.2010.09.019
  17. Cosentino, C., Grieco, D., and Costanzo, V. (2011). ATM activates the pentose phosphate pathway promoting anti-oxidant defence and DNA repair. EMBO J. 30, 546-555. https://doi.org/10.1038/emboj.2010.330
  18. Csibi, A., Fendt, S.M., Li, C., Poulogiannis, G., Choo, A.Y., Chapski, D.J., Jeong, S.M., Dempsey, J.M., Parkhitko, A., Morrison, T., et al. (2013). The mTORC1 pathway stimulates glutamine metabolism and cell proliferation by repressing SIRT4. Cell 153, 840-854. https://doi.org/10.1016/j.cell.2013.04.023
  19. Currie, E., Schulze, A., Zechner, R., Walther, T.C., and Farese, R.V., Jr. (2013). Cellular fatty acid metabolism and cancer. Cell Metabol. 18, 153-161. https://doi.org/10.1016/j.cmet.2013.05.017
  20. Dan, L., Klimenkova, O., Klimiankou, M., Klusman, J.H., van den Heuvel-Eibrink, M.M., Reinhardt, D., Welte, K., and Skokowa, J. (2012). The role of sirtuin 2 activation by nicotinamide phosphoribosyltransferase in the aberrant proliferation and survival of myeloid leukemia cells. Haematologica 97, 551-559. https://doi.org/10.3324/haematol.2011.055236
  21. Dang, C.V. (2010). Rethinking the Warburg effect with Myc micromanaging glutamine metabolism. Cancer Res. 70, 859-862. https://doi.org/10.1158/0008-5472.CAN-09-3556
  22. DeBerardinis, R.J., Mancuso, A., Daikhin, E., Nissim, I., Yudkoff, M., Wehrli, S., and Thompson, C.B. (2007). Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. USA 104, 19345-19350. https://doi.org/10.1073/pnas.0709747104
  23. Deng, C.X. (2006). BRCA1: cell cycle checkpoint, genetic instability, DNA damage response and cancer evolution. Nucleic Acids Res. 34, 1416-1426. https://doi.org/10.1093/nar/gkl010
  24. Dominy, J.E., Jr., Lee, Y., Jedrychowski, M.P., Chim, H., Jurczak, M.J., Camporez, J.P., Ruan, H.B., Feldman, J., Pierce, K., Mostoslavsky, R., et al. (2012). The deacetylase Sirt6 activates the acetyltransferase GCN5 and suppresses hepatic gluconeogenesis. Mol. Cell 48, 900-913. https://doi.org/10.1016/j.molcel.2012.09.030
  25. Dryden, S.C., Nahhas, F.A., Nowak, J.E., Goustin, A.S., and Tainsky, M.A. (2003). Role for human SIRT2 NAD-dependent deacetylase activity in control of mitotic exit in the cell cycle. Mol. Cell. Biol. 23, 3173-3185. https://doi.org/10.1128/MCB.23.9.3173-3185.2003
  26. Du, J., Zhou, Y., Su, X., Yu, J.J., Khan, S., Jiang, H., Kim, J., Woo, J., Kim, J.H., Choi, B.H., et al. (2011). Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science 334, 806-809. https://doi.org/10.1126/science.1207861
  27. Eng, C.H., Yu, K., Lucas, J., White, E., and Abraham, R.T. (2010). Ammonia derived from glutaminolysis is a diffusible regulator of autophagy. Sci. Signal. 3, ra31.
  28. Fan, W., and Luo, J. (2010). SIRT1 regulates UV-induced DNA repair through deacetylating XPA. Mol. Cell 39, 247-258. https://doi.org/10.1016/j.molcel.2010.07.006
  29. Finkel, T., Deng, C.X., and Mostoslavsky, R. (2009). Recent progress in the biology and physiology of sirtuins. Nature 460, 587-591. https://doi.org/10.1038/nature08197
  30. Finley, L.W., Carracedo, A., Lee, J., Souza, A., Egia, A., Zhang, J., Teruya-Feldstein, J., Moreira, P.I., Cardoso, S.M., Clish, C.B., et al. (2011). SIRT3 opposes reprogramming of cancer cell metabolism through HIF1alpha destabilization. Cancer Cell 19, 416-428. https://doi.org/10.1016/j.ccr.2011.02.014
  31. Ford, E., Voit, R., Liszt, G., Magin, C., Grummt, I., and Guarente, L. (2006). Mammalian Sir2 homolog SIRT7 is an activator of RNA polymerase I transcription. Genes Dev. 20, 1075-1080. https://doi.org/10.1101/gad.1399706
  32. Frescas, D., Valenti, L., and Accili, D. (2005). Nuclear trapping of the forkhead transcription factor FoxO1 via Sirt-dependent deacetylation promotes expression of glucogenetic genes. J. Biol. Chem. 280, 20589-20595. https://doi.org/10.1074/jbc.M412357200
  33. Fu, X., Wan, S., Lyu, Y.L., Liu, L.F., and Qi, H. (2008). Etoposide induces ATM-dependent mitochondrial biogenesis through AMPK activation. PLoS One 3, e2009. https://doi.org/10.1371/journal.pone.0002009
  34. Gatenby, R.A., and Gillies, R.J. (2004). Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891-899. https://doi.org/10.1038/nrc1478
  35. Gerhart-Hines, Z., Rodgers, J.T., Bare, O., Lerin, C., Kim, S.H., Mostoslavsky, R., Alt, F.W., Wu, Z., and Puigserver, P. (2007). Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J. 26, 1913-1923. https://doi.org/10.1038/sj.emboj.7601633
  36. Guarente, L. (2013). Calorie restriction and sirtuins revisited. Genes Dev. 27, 2072-2085. https://doi.org/10.1101/gad.227439.113
  37. Guarente, L., and Kenyon, C. (2000). Genetic pathways that regulate ageing in model organisms. Nature 408, 255-262. https://doi.org/10.1038/35041700
  38. Haigis, M.C., and Guarente, L.P. (2006). Mammalian sirtuins--emerging roles in physiology, aging, and calorie restriction. Genes Dev. 20, 2913-2921. https://doi.org/10.1101/gad.1467506
  39. Haigis, M.C., and Sinclair, D.A. (2010). Mammalian sirtuins: biological insights and disease relevance. Ann. Rev. Pathol. 5, 253-295. https://doi.org/10.1146/annurev.pathol.4.110807.092250
  40. Haigis, M.C., Mostoslavsky, R., Haigis, K.M., Fahie, K., Christodoulou, D.C., Murphy, A.J., Valenzuela, D.M., Yancopoulos, G.D., Karow, M., Blander, G., et al. (2006). SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell 126, 941-954. https://doi.org/10.1016/j.cell.2006.06.057
  41. Hallows, W.C., Yu, W., Smith, B.C., Devries, M.K., Ellinger, J.J., Someya, S., Shortreed, M.R., Prolla, T., Markley, J.L., Smith, L.M., et al. (2011). Sirt3 promotes the urea cycle and fatty acid oxidation during dietary restriction. Mol. Cell 41, 139-149. https://doi.org/10.1016/j.molcel.2011.01.002
  42. Hallows, W.C., Yu, W., and Denu, J.M. (2012). Regulation of glycolytic enzyme phosphoglycerate mutase-1 by Sirt1 proteinmediated deacetylation. J. Biol. Chem. 287, 3850-3858. https://doi.org/10.1074/jbc.M111.317404
  43. Hanahan, D., and Weinberg, R.A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646-674. https://doi.org/10.1016/j.cell.2011.02.013
  44. Hebert, A.S., Dittenhafer-Reed, K.E., Yu, W., Bailey, D.J., Selen, E.S., Boersma, M.D., Carson, J.J., Tonelli, M., Balloon, A.J., Higbee, A.J., et al. (2013). Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial protein acetylome. Mol. Cell 49, 186-199. https://doi.org/10.1016/j.molcel.2012.10.024
  45. Herranz, D., Maraver, A., Canamero, M., Gomez-Lopez, G., Inglada-Perez, L., Robledo, M., Castelblanco, E., Matias-Guiu, X., and Serrano, M. (2013). SIRT1 promotes thyroid carcinogenesis driven by PTEN deficiency. Oncogene 32, 4052-4056. https://doi.org/10.1038/onc.2012.407
  46. Hirschey, M.D., Shimazu, T., Goetzman, E., Jing, E., Schwer, B., Lombard, D.B., Grueter, C.A., Harris, C., Biddinger, S., Ilkayeva, O.R., et al. (2010). SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature 464, 121-125. https://doi.org/10.1038/nature08778
  47. Hou, H., Chen, W., Zhao, L., Zuo, Q., Zhang, G., Zhang, X., Wang, H., Gong, H., Li, X., Wang, M., et al. (2012). Cortactin is associated with tumour progression and poor prognosis in prostate cancer and SIRT2 other than HADC6 may work as facilitator in situ. J. Clin. Pathol. 65, 1088-1096. https://doi.org/10.1136/jclinpath-2012-200940
  48. Houtkooper, R.H., Pirinen, E., and Auwerx, J. (2012). Sirtuins as regulators of metabolism and healthspan. Nat. Rev. Mol. Cell Biol. 13, 225-238. https://doi.org/10.1038/nrn3209
  49. Hubbi, M.E., Hu, H., Kshitiz, Gilkes, D.M., and Semenza, G.L. (2013). Sirtuin-7 inhibits the activity of hypoxia-inducible factors. J. Biol. Chem. 288, 20768-20775. https://doi.org/10.1074/jbc.M113.476903
  50. Huffman, D.M., Grizzle, W.E., Bamman, M.M., Kim, J.S., Eltoum, I.A., Elgavish, A., and Nagy, T.R. (2007). SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 67, 6612-6618. https://doi.org/10.1158/0008-5472.CAN-07-0085
  51. Jeong, J., Juhn, K., Lee, H., Kim, S.H., Min, B.H., Lee, K.M., Cho, M.H., Park, G.H., and Lee, K.H. (2007). SIRT1 promotes DNA repair activity and deacetylation of Ku70. Exp. Mol. Med. 39, 8-13. https://doi.org/10.1038/emm.2007.2
  52. Jeong, S.M., Xiao, C., Finley, L.W., Lahusen, T., Souza, A.L., Pierce, K., Li, Y.H., Wang, X., Laurent, G., German, N.J., et al. (2013). SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell 23, 450-463. https://doi.org/10.1016/j.ccr.2013.02.024
  53. Jeong, S.M., Lee, A., Lee, J., and Haigis, M.C. (2014). SIRT4 protein suppresses tumor formation in genetic models of Mycinduced B cell lymphoma. J. Biol. Chem. 289, 4135-4144. https://doi.org/10.1074/jbc.M113.525949
  54. Jeong, S.M., Lee, J., Finley, L.W., Schmidt, P.J., Fleming, M.D., and Haigis, M.C. (2015). SIRT3 regulates cellular iron metabolism and cancer growth by repressing iron regulatory protein 1. Oncogene 34, 2115-2124. https://doi.org/10.1038/onc.2014.124
  55. Jiang, W., Wang, S., Xiao, M., Lin, Y., Zhou, L., Lei, Q., Xiong, Y., Guan, K.L., and Zhao, S. (2011). Acetylation regulates gluconeogenesis by promoting PEPCK1 degradation via recruiting the UBR5 ubiquitin ligase. Mol. Cell 43, 33-44. https://doi.org/10.1016/j.molcel.2011.04.028
  56. Jing, E., Gesta, S., and Kahn, C.R. (2007). SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation. Cell Metabol. 6, 105-114. https://doi.org/10.1016/j.cmet.2007.07.003
  57. Kaidi, A., Weinert, B.T., Choudhary, C., and Jackson, S.P. (2010). Human SIRT6 promotes DNA end resection through CtIP deacetylation. Science 329, 1348-1353. https://doi.org/10.1126/science.1192049
  58. Kanfi, Y., Peshti, V., Gil, R., Naiman, S., Nahum, L., Levin, E., Kronfeld-Schor, N., and Cohen, H.Y. (2010). SIRT6 protects against pathological damage caused by diet-induced obesity. Aging Cell 9, 162-173. https://doi.org/10.1111/j.1474-9726.2009.00544.x
  59. Khongkow, M., Olmos, Y., Gong, C., Gomes, A.R., Monteiro, L.J., Yague, E., Cavaco, T.B., Khongkow, P., Man, E.P., Laohasinnarong, S., et al. (2013). SIRT6 modulates paclitaxel and epirubicin resistance and survival in breast cancer. Carcinogenesis 34, 1476-1486. https://doi.org/10.1093/carcin/bgt098
  60. Kim, H.S., Patel, K., Muldoon-Jacobs, K., Bisht, K.S., Aykin-Burns, N., Pennington, J.D., van der Meer, R., Nguyen, P., Savage, J., Owens, K.M., et al. (2010a). SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell 17, 41-52. https://doi.org/10.1016/j.ccr.2009.11.023
  61. Kim, H.S., Xiao, C., Wang, R.H., Lahusen, T., Xu, X., Vassilopoulos, A., Vazquez-Ortiz, G., Jeong, W.I., Park, O., Ki, S.H., et al. (2010b). Hepatic-specific disruption of SIRT6 in mice results in fatty liver formation due to enhanced glycolysis and triglyceride synthesis. Cell Metabol. 12, 224-236. https://doi.org/10.1016/j.cmet.2010.06.009
  62. Kim, H.S., Vassilopoulos, A., Wang, R.H., Lahusen, T., Xiao, Z., Xu, X., Li, C., Veenstra, T.D., Li, B., Yu, H., et al. (2011). SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity. Cancer Cell 20, 487-499. https://doi.org/10.1016/j.ccr.2011.09.004
  63. Kim, J.K., Noh, J.H., Jung, K.H., Eun, J.W., Bae, H.J., Kim, M.G., Chang, Y.G., Shen, Q., Park, W.S., Lee, J.Y., et al. (2013). Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology 57, 1055-1067. https://doi.org/10.1002/hep.26101
  64. Kiran, S., Oddi, V., and Ramakrishna, G. (2015). Sirtuin 7 promotes cellular survival following genomic stress by attenuation of DNA damage, SAPK activation and p53 response. Exp. Cell Res. 331, 123-141. https://doi.org/10.1016/j.yexcr.2014.11.001
  65. Krishnan, J., Danzer, C., Simka, T., Ukropec, J., Walter, K.M., Kumpf, S., Mirtschink, P., Ukropcova, B., Gasperikova, D., Pedrazzini, T., et al. (2012). Dietary obesity-associated Hif1alpha activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD+ system. Genes Dev. 26, 259-270. https://doi.org/10.1101/gad.180406.111
  66. Lan, F., Cacicedo, J.M., Ruderman, N., and Ido, Y. (2008). SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation. J. Biol. Chem. 283, 27628-27635. https://doi.org/10.1074/jbc.M805711200
  67. Lapenna, S., and Giordano, A. (2009). Cell cycle kinases as therapeutic targets for cancer. Nat. Rev. Drug Discov. 8, 547-566. https://doi.org/10.1038/nrd2907
  68. Laurent, G., German, N.J., Saha, A.K., de Boer, V.C., Davies, M., Koves, T.R., Dephoure, N., Fischer, F., Boanca, G., Vaitheesvaran, B., et al. (2013). SIRT4 coordinates the balance between lipid synthesis and catabolism by repressing malonyl CoA decarboxylase. Mol. Cell 50, 686-698. https://doi.org/10.1016/j.molcel.2013.05.012
  69. Li, X., Zhang, S., Blander, G., Tse, J.G., Krieger, M. and Guarente, L. (2007). SIRT1 deacetylates and positively regulates the nuclear receptor LXR. Mol. Cell 28, 91-106. https://doi.org/10.1016/j.molcel.2007.07.032
  70. Li, K., Casta, A., Wang, R., Lozada, E., Fan, W., Kane, S., Ge, Q., Gu, W., Orren, D., and Luo, J. (2008). Regulation of WRN protein cellular localization and enzymatic activities by SIRT1-mediated deacetylation. J. Biol. Chem. 283, 7590-7598. https://doi.org/10.1074/jbc.M709707200
  71. Li, S., Banck, M., Mujtaba, S., Zhou, M.M., Sugrue, M.M., and Walsh, M.J. (2010). p53-induced growth arrest is regulated by the mitochondrial SirT3 deacetylase. PLoS One 5, e10486. https://doi.org/10.1371/journal.pone.0010486
  72. Lim, C.S. (2006). SIRT1: tumor promoter or tumor suppressor? Med. Hypotheses 67, 341-344. https://doi.org/10.1016/j.mehy.2006.01.050
  73. Lim, J.H., Lee, Y.M., Chun, Y.S., Chen, J., Kim, J.E., and Park, J.W. (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
  74. Lin, Y.H., Yuan, J., Pei, H., Liu, T., Ann, D.K., and Lou, Z. (2015). KAP1 deacetylation by SIRT1 promotes non-homologous endjoining repair. PLoS One 10, e0123935. https://doi.org/10.1371/journal.pone.0123935
  75. Liszt, G., Ford, E., Kurtev, M., and Guarente, L. (2005). Mouse Sir2 homolog SIRT6 is a nuclear ADP-ribosyltransferase. J. Biol. Chem. 280, 21313-21320. https://doi.org/10.1074/jbc.M413296200
  76. Liu, P.Y., Xu, N., Malyukova, A., Scarlett, C.J., Sun, Y.T., Zhang, X.D., Ling, D., Su, S.P., Nelson, C., Chang, D.K., et al. (2013). The histone deacetylase SIRT2 stabilizes Myc oncoproteins. Cell Death Differ. 20, 503-514. https://doi.org/10.1038/cdd.2012.147
  77. Liu, T., Lin, Y.H., Leng, W., Jung, S.Y., Zhang, H., Deng, M., Evans, D., Li, Y., Luo, K., Qin, B., et al. (2014). A divergent role of the SIRT1-TopBP1 axis in regulating metabolic checkpoint and DNA damage checkpoint. Mol. Cell 56, 681-695. https://doi.org/10.1016/j.molcel.2014.10.007
  78. Lombard, D.B., Schwer, B., Alt, F.W., and Mostoslavsky, R. (2008). SIRT6 in DNA repair, metabolism and ageing. J. Int. Med. 263, 128-141. https://doi.org/10.1111/j.1365-2796.2007.01902.x
  79. Mao, Z., Hine, C., Tian, X., Van Meter, M., Au, M., Vaidya, A., Seluanov, A., and Gorbunova, V. (2011). SIRT6 promotes DNA repair under stress by activating PARP1. Science 332, 1443-1446. https://doi.org/10.1126/science.1202723
  80. Martinez-Pastor, B., and Mostoslavsky, R. (2012). Sirtuins, metabolism, and cancer. Front. Pharmacol. 3, 22.
  81. Mathias, R.A., Greco, T.M., Oberstein, A., Budayeva, H.G., Chakrabarti, R., Rowland, E.A., Kang, Y., Shenk, T., and Cristea, I.M. (2014). Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell 159, 1615-1625. https://doi.org/10.1016/j.cell.2014.11.046
  82. McCord, R.A., Michishita, E., Hong, T., Berber, E., Boxer, L.D., Kusumoto, R., Guan, S., Shi, X., Gozani, O., Burlingame, A.L., et al. (2009). SIRT6 stabilizes DNA-dependent protein kinase at chromatin for DNA double-strand break repair. Aging 1, 109-121. https://doi.org/10.18632/aging.100011
  83. Michishita, E., McCord, R.A., Berber, E., Kioi, M., Padilla-Nash, H., Damian, M., Cheung, P., Kusumoto, R., Kawahara, T.L., Barrett, J.C., 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
  84. Ming, M., Shea, C.R., Guo, X., Li, X., Soltani, K., Han, W., and He, Y.Y. (2010). Regulation of global genome nucleotide excision repair by SIRT1 through xeroderma pigmentosum C. Proc. Natl. Acad. Sci. USA 107, 22623-22628. https://doi.org/10.1073/pnas.1010377108
  85. Morris, B.J. (2013). Seven sirtuins for seven deadly diseases of aging. Free Radical Biol. Med. 56, 133-171. https://doi.org/10.1016/j.freeradbiomed.2012.10.525
  86. Mostoslavsky, R., Chua, K.F., Lombard, D.B., Pang, W.W., Fischer, M.R., Gellon, L., Liu, P., Mostoslavsky, G., Franco, S., Murphy, M.M., 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
  87. Moynihan, K.A., Grimm, A.A., Plueger, M.M., Bernal-Mizrachi, E., Ford, E., Cras-Meneur, C., Permutt, M.A., and Imai, S. (2005). Increased dosage of mammalian Sir2 in pancreatic beta cells enhances glucose-stimulated insulin secretion in mice. Cell Metabol. 2, 105-117. https://doi.org/10.1016/j.cmet.2005.07.001
  88. Nakagawa, T., Lomb, D.J., Haigis, M.C., and Guarente, L. (2009). SIRT5 Deacetylates carbamoyl phosphate synthetase 1 and regulates the urea cycle. Cell 137, 560-570. https://doi.org/10.1016/j.cell.2009.02.026
  89. Negrini, S., Gorgoulis, V.G., and Halazonetis, T.D. (2010). Genomic instability--an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220-228. https://doi.org/10.1038/nrm2858
  90. North, B.J., and Verdin, E. (2007). Mitotic regulation of SIRT2 by cyclin-dependent kinase 1-dependent phosphorylation. J. Biol. Chem. 282, 19546-19555. https://doi.org/10.1074/jbc.M702990200
  91. Oberdoerffer, P., Michan, S., McVay, M., Mostoslavsky, R., Vann, J., Park, S.K., Hartlerode, A., Stegmuller, J., Hafner, A., Loerch, P., et al. (2008). SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 135, 907-918. https://doi.org/10.1016/j.cell.2008.10.025
  92. Palacios, J.A., Herranz, D., De Bonis, M.L., Velasco, S., Serrano, M., and Blasco, M.A. (2010). SIRT1 contributes to telomere maintenance and augments global homologous recombination. J. Cell Biol. 191, 1299-1313. https://doi.org/10.1083/jcb.201005160
  93. Park, J., Chen, Y., Tishkoff, D.X., Peng, C., Tan, M., Dai, L., Xie, Z., Zhang, Y., Zwaans, B.M., Skinner, M.E., et al. (2013). SIRT5-mediated lysine desuccinylation impacts diverse metabolic pathways. Mol. Cell 50, 919-930. https://doi.org/10.1016/j.molcel.2013.06.001
  94. Picard, F., Kurtev, M., Chung, N., Topark-Ngarm, A., Senawong, T., Machado De Oliveira, R., Leid, M., McBurney, M.W., and Guarente, L. (2004). Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature 429, 771-776. https://doi.org/10.1038/nature02583
  95. Polletta, L., Vernucci, E., Carnevale, I., Arcangeli, T., Rotili, D., Palmerio, S., Steegborn, C., Nowak, T., Schutkowski, M., Pellegrini, L., et al. (2015). SIRT5 regulation of ammoniainduced autophagy and mitophagy. Autophagy 11, 253-270. https://doi.org/10.1080/15548627.2015.1009778
  96. Ponugoti, B., Kim, D.H., Xiao, Z., Smith, Z., Miao, J., Zang, M., Wu, S.Y., Chiang, C.M., Veenstra, T.D., and Kemper, J.K. (2010). SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J. Biol. Chem. 285, 33959-33970. https://doi.org/10.1074/jbc.M110.122978
  97. Qiu, X., Brown, K., Hirschey, M.D., Verdin, E., and Chen, D. (2010). Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metabol. 12, 662-667. https://doi.org/10.1016/j.cmet.2010.11.015
  98. Rodgers, J.T., Lerin, C., Haas, W., Gygi, S.P., Spiegelman, B.M., and Puigserver, P. (2005). Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 434, 113-118. https://doi.org/10.1038/nature03354
  99. Ryu, D., Jo, Y.S., Lo Sasso, G., Stein, S., Zhang, H., Perino, A., Lee, J.U., Zeviani, M., Romand, R., Hottiger, M.O., et al. (2014). A SIRT7-dependent acetylation switch of GABPbeta1 controls mitochondrial function. Cell Metabol. 20, 856-869. https://doi.org/10.1016/j.cmet.2014.08.001
  100. Santos, C.R., and Schulze, A. (2012). Lipid metabolism in cancer. FEBS J. 279, 2610-2623. https://doi.org/10.1111/j.1742-4658.2012.08644.x
  101. Sapkota, G.P., Deak, M., Kieloch, A., Morrice, N., Goodarzi, A.A., Smythe, C., Shiloh, Y., Lees-Miller, S.P., and Alessi, D.R. (2002). Ionizing radiation induces ataxia telangiectasia mutated kinase (ATM).-mediated phosphorylation of LKB1/STK11 at Thr-366. Biochem. J. 368, 507-516. https://doi.org/10.1042/bj20021284
  102. Sawada, M., Sun, W., Hayes, P., Leskov, K., Boothman, D.A., and Matsuyama, S. (2003). Ku70 suppresses the apoptotic translocation of Bax to mitochondria. Nat. Cell Biol. 5, 320-329. https://doi.org/10.1038/ncb950
  103. Sebastian, C., Zwaans, B.M., Silberman, D.M., Gymrek, M., Goren, A., Zhong, L., Ram, O., Truelove, J., Guimaraes, A.R., Toiber, D., et al. (2012). The histone deacetylase SIRT6 is a tumor suppressor that controls cancer metabolism. Cell 151, 1185-1199. https://doi.org/10.1016/j.cell.2012.10.047
  104. Seo, K.S., Park, J.H., Heo, J.Y., Jing, K., Han, J., Min, K.N., Kim, C., Koh, G.Y., Lim, K., Kang, G.Y., et al. (2015). SIRT2 regulates tumour hypoxia response by promoting HIF-1alpha hydroxylation. Oncogene 34, 1354-1362. https://doi.org/10.1038/onc.2014.76
  105. Serrano, L., Martinez-Redondo, P., Marazuela-Duque, A., Vazquez, B.N., Dooley, S.J., Voigt, P., Beck, D.B., Kane-Goldsmith, N., Tong, Q., Rabanal, R.M., et al. (2013). The tumor suppressor SirT2 regulates cell cycle progression and genome stability by modulating the mitotic deposition of H4K20 methylation. Genes Dev. 27, 639-653. https://doi.org/10.1101/gad.211342.112
  106. Shimazu, T., Hirschey, M.D., Hua, L., Dittenhafer-Reed, K.E., Schwer, B., Lombard, D.B., Li, Y., Bunkenborg, J., Alt, F.W., Denu, J.M., et al. (2010). SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metabol. 12, 654-661. https://doi.org/10.1016/j.cmet.2010.11.003
  107. Shin, J., He, M., Liu, Y., Paredes, S., Villanova, L., Brown, K., Qiu, X., Nabavi, N., Mohrin, M., Wojnoonski, K., et al. (2013). SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Cell Rep. 5, 654-665. https://doi.org/10.1016/j.celrep.2013.10.007
  108. Someya, S., Yu, W., Hallows, W.C., Xu, J., Vann, J.M., Leeuwenburgh, C., Tanokura, M., Denu, J.M. and Prolla, T.A. (2010). Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 143, 802-812. https://doi.org/10.1016/j.cell.2010.10.002
  109. Su, T.T. (2006). Cellular responses to DNA damage: one signal, multiple choices. Annu. Rev. Genet. 40, 187-208. https://doi.org/10.1146/annurev.genet.40.110405.090428
  110. Sundaresan, N.R., Samant, S.A., Pillai, V.B., Rajamohan, S.B., and Gupta, M.P. (2008). SIRT3 is a stress-responsive deacetylase in cardiomyocytes that protects cells from stress-mediated cell death by deacetylation of Ku70. Mol. Cell. Biol. 28, 6384-6401. https://doi.org/10.1128/MCB.00426-08
  111. Tao, R., Coleman, M.C., Pennington, J.D., Ozden, O., Park, S.H., Jiang, H., Kim, H.S., Flynn, C.R., Hill, S., Hayes McDonald, W., et al. (2010). Sirt3-mediated deacetylation of evolutionarily conserved lysine 122 regulates MnSOD activity in response to stress. Mol. Cell 40, 893-904. https://doi.org/10.1016/j.molcel.2010.12.013
  112. Tennant, D.A., Duran, R.V., and Gottlieb, E. (2010). Targeting metabolic transformation for cancer therapy. Nat. Rev. Cancer 10, 267-277. https://doi.org/10.1038/nrc2817
  113. Tong, X., Zhao, F., and Thompson, C.B. (2009). The molecular determinants of de novo nucleotide biosynthesis in cancer cells. Curr. Opin. Genet. Dev. 19, 32-37. https://doi.org/10.1016/j.gde.2009.01.002
  114. Vakhrusheva, O., Braeuer, D., Liu, Z., Braun, T., and Bober, E. (2008a). Sirt7-dependent inhibition of cell growth and proliferation might be instrumental to mediate tissue integrity during aging. J. Physiol. Pharmacol. 59 Suppl 9, 201-212.
  115. Vakhrusheva, O., Smolka, C., Gajawada, P., Kostin, S., Boettger, T., Kubin, T., Braun, T., and Bober, E. (2008b). Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ.Res. 102, 703-710. https://doi.org/10.1161/CIRCRESAHA.107.164558
  116. Vander Heiden, M.G., Cantley, L.C., and Thompson, C.B. (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029-1033. https://doi.org/10.1126/science.1160809
  117. Vaquero, A., Scher, M.B., Lee, D.H., Sutton, A., Cheng, H.L., Alt, F.W., Serrano, L., Sternglanz, R., and Reinberg, D. (2006). SirT2 is a histone deacetylase with preference for histone H4 Lys 16 during mitosis. Genes Dev. 20, 1256-1261. https://doi.org/10.1101/gad.1412706
  118. Verdin, E., Hirschey, M.D., Finley, L.W., and Haigis, M.C. (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
  119. Wang, F., Nguyen, M., Qin, F.X., and Tong, Q. (2007). SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell 6, 505-514. https://doi.org/10.1111/j.1474-9726.2007.00304.x
  120. Wang, R.H., Sengupta, K., Li, C., Kim, H.S., Cao, L., Xiao, C., Kim, S., Xu, X., Zheng, Y., Chilton, B., et al. (2008). Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice. Cancer Cell 14, 312-323. https://doi.org/10.1016/j.ccr.2008.09.001
  121. Wang, J.B., Erickson, J.W., Fuji, R., Ramachandran, S., Gao, P., Dinavahi, R., Wilson, K.F., Ambrosio, A.L., Dias, S.M., Dang, C.V., et al. (2010). Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 18, 207-219. https://doi.org/10.1016/j.ccr.2010.08.009
  122. Warburg, O. (1956). On the origin of cancer cells. Science 123, 309-314. https://doi.org/10.1126/science.123.3191.309
  123. Wise, D.R., and Thompson, C.B. (2010). Glutamine addiction: a new therapeutic target in cancer. Trends Biochem. Sci. 35, 427-433. https://doi.org/10.1016/j.tibs.2010.05.003
  124. Yang, C., Sudderth, J., Dang, T., Bachoo, R.M., McDonald, J.G. and DeBerardinis, R.J. (2009). Glioblastoma cells require glutamate dehydrogenase to survive impairments of glucose metabolism or Akt signaling. Cancer Res. 69, 7986-7993. https://doi.org/10.1158/0008-5472.CAN-09-2266
  125. Yoshizawa, T., Karim, M.F., Sato, Y., Senokuchi, T., Miyata, K., Fukuda, T., Go, C., Tasaki, M., Uchimura, K., Kadomatsu, T., et al. (2014). SIRT7 controls hepatic lipid metabolism by regulating the ubiquitin-proteasome pathway. Cell Metabol. 19, 712-721. https://doi.org/10.1016/j.cmet.2014.03.006
  126. Yuan, H., Wang, Z., Li, L., Zhang, H., Modi, H., Horne, D., Bhatia, R., and Chen, W. (2012). Activation of stress response gene SIRT1 by BCR-ABL promotes leukemogenesis. Blood 119, 1904-1914. https://doi.org/10.1182/blood-2011-06-361691
  127. Zhong, L., D'Urso, A., Toiber, D., Sebastian, C., Henry, R.E., Vadysirisack, D.D., Guimaraes, A., Marinelli, B., Wikstrom, J.D., Nir, T., et al. (2010). The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1alpha. Cell 140, 280-293. https://doi.org/10.1016/j.cell.2009.12.041

Cited by

  1. Mitochondrial Sirtuins and Molecular Mechanisms of Aging vol.23, pp.4, 2017, https://doi.org/10.1016/j.molmed.2017.02.005
  2. Sirtuins Expression and Their Role in Retinal Diseases vol.2017, 2017, https://doi.org/10.1155/2017/3187594
  3. Thienopyrimidinone Based Sirtuin-2 (SIRT2)-Selective Inhibitors Bind in the Ligand Induced Selectivity Pocket vol.60, pp.5, 2017, https://doi.org/10.1021/acs.jmedchem.6b01690
  4. SIRT4 regulates cancer cell survival and growth after stress vol.470, pp.2, 2016, https://doi.org/10.1016/j.bbrc.2016.01.078
  5. Alpha-Ketoglutarate as a Molecule with Pleiotropic Activity: Well-Known and Novel Possibilities of Therapeutic Use vol.65, pp.1, 2017, https://doi.org/10.1007/s00005-016-0406-x
  6. Sirtuins in metabolism, DNA repair and cancer vol.35, pp.1, 2016, https://doi.org/10.1186/s13046-016-0461-5
  7. Lactate dehydrogenase inhibition: exploring possible applications beyond cancer treatment vol.8, pp.6, 2016, https://doi.org/10.4155/fmc.16.10
  8. Therapeutic role of sirtuins in neurodegenerative disease and their modulation by polyphenols vol.73, 2017, https://doi.org/10.1016/j.neubiorev.2016.11.022
  9. Inhibitors of Trypanosoma cruzi Sir2 related protein 1 as potential drugs against Chagas disease vol.12, pp.1, 2018, https://doi.org/10.1371/journal.pntd.0006180
  10. Hsp90 Stabilizes SIRT1 Orthologs in Mammalian Cells and C. elegans vol.19, pp.11, 2018, https://doi.org/10.3390/ijms19113661
  11. Metabolism and Epigenetic Interplay in Cancer: Regulation and Putative Therapeutic Targets vol.9, pp.1664-8021, 2018, https://doi.org/10.3389/fgene.2018.00427
  12. Engineering Lineage Potency and Plasticity of Stem Cells using Epigenetic Molecules vol.8, pp.1, 2018, https://doi.org/10.1038/s41598-018-34511-7
  13. A 5′ AMP-Activated Protein Kinase Enzyme Activator, Compound 59, Induces Autophagy and Apoptosis in Human Oral Squamous Cell Carcinoma vol.123, pp.1, 2018, https://doi.org/10.1111/bcpt.12976
  14. The sirtuin 1/2 inhibitor tenovin-1 induces a nonlinear apoptosis-inducing factor-dependent cell death in a p53 null Ewing’s sarcoma cell line vol.36, pp.3, 2018, https://doi.org/10.1007/s10637-017-0541-1
  15. Dual Tumor Suppressor and Tumor Promoter Action of Sirtuins in Determining Malignant Phenotype vol.10, pp.1663-9812, 2019, https://doi.org/10.3389/fphar.2019.00038
  16. The Glycolytic Switch in Tumors: How Many Players Are Involved? vol.8, pp.17, 2015, https://doi.org/10.7150/jca.21125
  17. The sirtuin family in cancer vol.18, pp.18, 2019, https://doi.org/10.1080/15384101.2019.1634953
  18. Regulation of Adaptive Immune Cells by Sirtuins vol.10, pp.None, 2015, https://doi.org/10.3389/fendo.2019.00466
  19. SIRT5 downregulation is associated with poor prognosis in glioblastoma vol.24, pp.4, 2015, https://doi.org/10.3233/cbm-182197
  20. Sirtuins and SIRT6 in Carcinogenesis and in Diet vol.20, pp.19, 2015, https://doi.org/10.3390/ijms20194945
  21. Sirt4: A Multifaceted Enzyme at the Crossroads of Mitochondrial Metabolism and Cancer vol.10, pp.None, 2020, https://doi.org/10.3389/fonc.2020.00474
  22. Expression of SIRT1, SIRT3 and SIRT6 Genes for Predicting Survival in Triple-Negative and Hormone Receptor-Positive Subtypes of Breast Cancer vol.26, pp.4, 2015, https://doi.org/10.1007/s12253-020-00873-5
  23. Sirtuin 1 and Sirtuin 3 in Granulosa Cell Tumors vol.22, pp.4, 2015, https://doi.org/10.3390/ijms22042047
  24. Elucidation of Structural Determinants Delineates the Residues Playing Key Roles in Differential Dynamics and Selective Inhibition of Sirt1-3 vol.61, pp.3, 2015, https://doi.org/10.1021/acs.jcim.0c01193
  25. Structure-based design, synthesis, and biological evaluation of novel piperine-resveratrol hybrids as antiproliferative agents targeting SIRT-2 vol.11, pp.41, 2015, https://doi.org/10.1039/d1ra04061h
  26. One Omics Approach Does Not Rule Them All: The Metabolome and the Epigenome Join Forces in Haematological Malignancies vol.5, pp.4, 2015, https://doi.org/10.3390/epigenomes5040022