Nutritional Influences on Epigenetics, Aging and Disease

Anna Meiliana, Nurrani Mustika Dewi, Andi Wijaya

Abstract


BACKGROUND: Altered epigenetics is regarded to play quite a role in many chronic diseases including cancer, diabetes, obesity, dyslipidemia, hypertension and neurodegeneration, hence nutrition suggested to contribute in epigenetics and disease.

CONTENT: Histone modifications, as a part of epigenetics mechanisms, depend on metabolites which acts as cofactors or substrates. Fluctuating levels of specific metabolites become the direct and rapid mechanisms to influence gene activity. Therefore, these metabolites may have a role as gatekeepers of chromatin, in chromatin landscape modulation as a response to key nutritional cues. Chemical modifications of histones and DNA have a critical role in epigenetic gene regulation including histone acetylation, and DNA methylation. Some enzymes add or remove such chemical modifications, and suggested to be sensitive to changes in intracellular metabolism, such as mutations in the metabolic enzymes succinate dehydrogenase (SDH), fumarate hydratase (FH) and isocitrate dehydrogenase (IDH) can result in cancer.

SUMMARY: As a response to their nutrient environment, organisms tend to rapidly alter their gene expression. Many evidences showed an epigenetic regulation of chromatin is coupled to the changes on metabolites levels due to this kind of response. These metabolites will lead the recruitment of transcriptional regulatory complexes to DNA, thus clearly influencing the dynamic chromatin landscape.

KEYWORDS: metabolites, enzymes, epigenetics, chromatin, nutrition


Full Text:

PDF

References


Waddington CH. The epigenotype 1942. Int J Epidemiol. 2012; 41: 10-3, CrossRef.

Waddington CH. Canalization of development and the inheritance of acquired characters. Nature. 1942; 150: 563-5, CrossRef.

Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016; 17: 487-500, CrossRef.

Kaelin WG, McKnight SL. Influence of metabolism on epigenetics and disease. Cell. 2013; 153: 56-69, CrossRef.

Borrelli E, Nestler EJ, Allis CD, Sassone-Corsi P. Decoding the epigenetic language of neuronal plasticity. Neuron. 2008; 60: 961-74, CrossRef.

Katada S, Imhof A, Sassone-Corsi P. Connecting threads: epigenetics and metabolism. Cell. 2012; 148: 24-8, CrossRef.

Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science. 2009; 324: 654-7, CrossRef.

Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. 2009; 324: 1076-80, CrossRef.

Zaman S, Lippman SI, Zhao X, Broach JR. How saccharomyces responds to nutrients. Annu Rev Genet. 2008; 42: 27-81, CrossRef.

Zaman S, Lippman SI, Schneper L, Slonim N, Broach JR. Glucose regulates transcription in yeast through a network of signaling pathways. Mol Syst Biol. 2009; 5: 245, CrossRef.

Lee KK, Workman JL. Histone acetyltransferase complexes: one size doesn’t fit all. Nat Rev Mol Cell Biol. 2007; 8: 284-95, CrossRef.

Shahbazian MD, Grunstein M. Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem. 2007; 76: 75-100, CrossRef.

Takahashi H, McCaffery JM, Irizarry RA, Boeke JD. Nucleocytosolic acetyl-coenzyme A synthetase is required for histone acetylation and global transcription. Mol Cell. 2006; 23: 207-17, CrossRef.

Cai L, Sutter BM, Li B, Tu BP. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes. Mol Cell. 2011; 42: 426-37, CrossRef.

Henry RA, Kuo YM, Bhattacharjee V, Yen TJ, Andrews AJ. Changing the selectivity of p300 by acetyl-CoA modulation of histone acetylation. ACS Chem Biol. 2015; 10: 146-56, CrossRef.

Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov. 2014; 13: 673-91, CrossRef.

Guedes-Dias P, Oliveira JM. Lysine deacetylases and mitochondrial dynamics in neurodegeneration. Biochim Biophys Acta. 2013; 1832: 1345-59, CrossRef.

Kaochar S, Tu BP. Gatekeepers of chromatin: small metabolites elicit big changes in gene expression. Trends Biochem Sci. 2012; 37: 477-83, CrossRef.

Haigis MC, Sinclair DA. Mammalian sirtuins: biological insights and disease relevance. Annu Rev Pathol. 2010; 5: 253-95, CrossRef.

Guarente L. Calorie restriction and sirtuins revisited. Genes Dev. 2013; 27: 2072-85, CrossRef.

Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013; 339: 211-4, CrossRef.

Cahill GF Jr, Herrera MG, Morgan AP, Soeldner JS, Steinke J, Levy PL, et al. Hormone-fuel interrelationships during fasting. J Clin Invest. 1966; 45: 1751-69, CrossRef.

Cahill GF Jr. Fuel metabolism in starvation. Annu Rev Nutr. 2006; 26: 1-22, CrossRef.

Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell. 2012; 48: 612-26, CrossRef.

Jackson V, Shires A, Chalkley R, Granner DK. Studies on highly metabolically active acetylation and phosphorylation of histones. J Biol Chem. 1975; 250: 4856–4863, PMID.

Waterborg JH. Dynamics of histone acetylation in vivo. A function for acetylation turnover? Biochem Cell Biol. 2002; 80: 363-78, CrossRef.

Martinez-Pastor B, Cosentino C, Mostoslavsky R. A tale of metabolites: the cross-talk between chromatin and energy metabolism. Cancer Discov. 2013; 3: 497-501, CrossRef.

Comerford SA, Huang Z, Du X, Wang Y, Cai L, Witkiewicz AK, et al. Acetate dependence of tumors. Cell. 2014; 159: 1591-602, CrossRef.

Mashimo T, Pichumani K, Vemireddy V, Hatanpaa KJ, Singh DK, Sirasanagandla S, et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell. 2014; 159: 1603-14, CrossRef.

Kogure M, Takawa M, Saloura V, Sone K, Piao L, Ueda K, et al. The oncogenic polycomb histone methyltransferase EZH2 methylates lysine 120 on histone H2B and competes ubiquitination. Neoplasia. 2013; 15: 1251-61, CrossRef.

Strahl BD, Grant PA, Briggs SD, Sun ZW, Bone JR, Caldwell JA, et al. Set2 is a nucleosomal histone H3-selective methyltransferase that mediates transcriptional repression. Mol Cell Biol. 2002; 22: 1298-306, CrossRef.

van Leeuwen F, Gafken PR, Gottschling DE. Dot1p modulates silencing in yeast by methylation of the nucleosome core. Cell. 2002; 109: 745-56, CrossRef.

Edwards CR, Dang W, Berger SL. Histone H4 lysine 20 of Saccharomyces cerevisiae is monomethylated and functions in subtelomeric silencing. Biochemistry. 2011; 50: 10473-83, CrossRef.

Green EM, Mas G, Young NL, Garcia BA, Gozani O. Methylation of H4 lysines 5, 8 and 12 by yeast Set5 calibrates chromatin stress responses. Nat Struct Mol Biol. 2012; 19: 361-3, CrossRef.

Sadhu MJ, Guan Q, Li F, Sales-Lee J, Iavarone AT, Hammond MC, et al. Nutritional control of epigenetic processes in yeast and human cells. Genetics. 2013; 195: 831-44,

Huang Z, Cal L, Tu BP. Dietary control of chromatin. Curr Opin Cell Biol. 2015; 34: 69-74, CrossRef.

Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004; 119: 941-53,

Klose RJ, Kallin EM, Zhang Y. JmjC-domain-containing proteins and histone demethylation. Nat Rev Genet. 2006; 7: 715-27, CrossRef.

Tsukada Y, Fang J, Erdjument-Bromage H, Warren ME, Borchers CH, Tempst P, et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature. 2006; 439: 811-6, CrossRef.

Hojfeldt JW, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov. 2013; 12: 917-30, CrossRef.

Zee BM, Levin RS, Xu B, LeRoy G, Wingreen NS, Garcia BA. In vivo residue-specific histone methylation dynamics. J Biol Chem. 2010; 285: 3341-50, CrossRef.

Carey BW, Finley LW, Cross JR, Allis CD, Thompson CB. Intracellular alpha-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature. 2014; 518: 413-6, CrossRef.

Shyh-Chang N, Locasale JW, Lyssiotis CA, Zheng Y, Teo RY, Ratanasirintrawoot S, et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science. 2013; 339: 222-6, CrossRef.

Hino S, Sakamoto A, Nagaoka K, Anan K, Wang Y, Mimasu S, et al. FAD-dependent lysine-specific demethylase-1 regulates cellular energy expenditure. Nat Commun. 2012; 3: 758, CrossRef.

Berger S, Sassone-Corsi P. Metabolic signaling to chromatin. Cold Spring Harb Perspect Biol. 2016; 8: a019463, CrossRef.

Metallo CM, Vander Heiden MG. Metabolism strikes back: metabolic flux regulates cell signaling. Genes Dev. 2010; 24: 2717-22, CrossRef.

Wellen KE, Thompson CB. Cellular metabolic stress: considering how cells respond to nutrient excess. Mol Cell. 2010; 40: 323-32,

Guan KL, Xiong Y. Regulation of intermediary metabolism by protein acetylation. Trends Biochem Sci. 2011; 36: 108-16, CrossRef.

Wellen KE, Thompson GB. A two-way street: reciprocal regulation of metabolism and signaling. Nat Rev Mol Cell Biol. 2012; 13: 270-6, CrossRef.

Gerken T, Girard CA, Tung YC, Webby CJ, Saudek V, Hewitson KS, et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science. 2007; 318: 1469-72, CrossRef.

Lucchesi JC, Kuroda MI. Dosage compensation in Drosophila. Cold Spring Harb Perspect Biol. 2015; 7: a019398, CrossRef.

Smemo S, Tena JJ, Kim K-H, Gamazon ER, Sakabe NJ, Gómez-Marín C, et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature. 2014; 507: 371-5, CrossRef.

Shi Y, Shi Y. Metabolic enzymes and coenzymes in transcription–a direct link between metabolism and transcription? Trends Genet. 2004; 20: 445-52, CrossRef.

Amon J, Titgemeyer F, Burkovski A. Common patterns — unique features: nitrogen metabolism and regulation in gram-positive bacteria. FEMS Microbiol Rev. 2010; 34: 588-605, CrossRef.

Wilson CJ, Zhan H, Swint-Kruse l, Matthews KS. The lactose repressor system: paradigms for regulation, allosteric behavior and protein folding. Cell Mol life Sci. 2007; 64: 3-16, CrossRef.

Asher G, Schibler U. Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab. 2011; 13: 125-37, CrossRef.

Feige JN, auwerx J. Transcriptional coregulators in the control of energy homeostasis. Trends Cell Biol. 2007; 17: 292-301, CrossRef.

Jenuwein T, Allis CD. Translating the histone code. Science. 2001; 293: 1074-80, CrossRef.

Chiacchiera F, Piunti A, Pasini D. Epigenetic methylation and their connection with metabolism. Cell Mol Life Sci. 2013; 70: 1495-508, CrossRef.

Menzies KJ, Zhang H, Katsyuba E, Auwerx J. Protein acetylation in metabolism — metabolites and cofactors. Nat Rev Endocrinol. 2016; 12: 43-60, CrossRef.

Su X, Wellen KE, Rabinowitz JD. Metabolic control of methylation and acetylation. Curr Opin Chem Biol. 2016; 30: 52-60, CrossRef.

Gao L, Chiou W, Tang H, Cheng X, Camp HS, Burns DJ. Simultaneous quantification of malonyl-CoA and several other short-chain acyl-CoAs in animal tissues by ion-pairing reversed-phase HPLC/MS. J Chromatogr B Analyt Technol Biomed Life Sci. 2007; 853: 303-13, CrossRef.

Michan S. Calorie restriction and NAD(+)/sirtuin counteract the hallmarks of aging. Front Biosci. 2014; 19: 1300-19, CrossRef.

Cheng Z, Guo S, Copps K, Dong X, Kollipara R, Rodgers JT, et al. Foxo1 integrates insulin signaling with mitochondrial function in the liver. Nat Med. 2009; 15: 1307-11, CrossRef.

Braidy N, Guillemin GJ, Mansour H, Chan-Ling T, Poljak A, Grant R. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One. 2011; 6: e19194, CrossRef.

Yoshino J, Mills KF, Yoon MJ, Imai S. Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 2011; 14: 528-36, CrossRef.

Canto C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, et al. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 2012; 15: 838-47, CrossRef.

Kraus D, Yang Q, Kong D, Banks AS, Zhang L, Rodgers JT, et al. Nicotinamide N-methyltransferase knockdown protects against diet-induced obesity. Nature. 2014; 508: 258-62, CrossRef.

Ulanovskaya OA, Zuhl AM, Cravatt BF. NNMT promotes epigenetic remodeling in cancer by creating a metabolic methylation sink. Nat Chem Biol. 2013; 9: 300-6, CrossRef.

Kim do Y, Davis LM, Sullivan PG, Maalouf M, Simeone TA, van Brederode J, et al. Ketone bodies are protective against oxidative stress in neocortical neurons. J Neurochem. 2007; 101: 1316-26, CrossRef.

Chang P, Zuckermann AM, Williams S, Close AJ, Cano-Jaimez M, McEvoy JP, et al. Seizure control by derivatives of medium chain fatty acids associated with the ketogenic diet show novel branching-point structure for enhanced potency. J Pharmacol Exp Ther. 2014; 352: 43-52, CrossRef.

Luger K, Mäder AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997; 389: 251-60, CrossRef.

Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000; 403: 41-5, CrossRef.

Tan M, Luo H, Lee S, Jin F, Yang JS, Montellier E, et al. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell. 2011; 146: 1016-28, CrossRef.

Huang H, Sabari BR, Garcia BA, Allis CD, Zhao Y. SnapShot: Histone Modifications. Cell. 2014; 159: 458-458.e1, CrossRef.

Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 2014; 15: 703-8, CrossRef.

Baylin SB, Jones PA. A decade of exploring the cancer epigenome — biological and translational implications. Nat Rev Cancer. 2011; 11: 726-34, CrossRef.

Wu H, Zhang Y. Mechanisms and functions of Tet protein-mediated 5-methylcytosine oxidation. Genes Dev. 2011; 25: 2436-52, CrossRef.

Ruthenburg AJ, Li H, Patel DJ, David Allis C. Multivalent engagement of chromatin modifications by linked binding modules. Nat Rev Mol Cell Biol. 2007; 8: 983-94, CrossRef.

Chi P, Allis CD, Wang GG. Covalent histone modifications — miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 2010; 10: 457-69, CrossRef.

Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012; 150: 12-27, CrossRef.

Cheng X. Structural and functional coordination of DNA and histone methylation. Cold Spring Harb Perspect Biol. 2014; 6: a018747, CrossRef.

Berger SL. The complex language of chromatin regulation during transcription. Nature. 2007; 447: 407-12, CrossRef.

Grillo MA, Colombatto S. S-adenosylmethionine and its products. Amino Acids. 2008; 34: 187-93, CrossRef.

Mentch SJ, Mehrmohamadi M, Huang L, Liu X, Gupta D, Mattocks D, et al. Histone methylation dynamics and gene regulation occur through the sensing of one-carbon metabolism. Cell Metab. 2015; 22: 861-73, CrossRef.

Shiraki N, Shiraki Y, Tsuyama T, Obata F, Miura M, Nagae G, et al. Methionine regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab. 2014; 19: 780-94, CrossRef.

Janke R, Dodson AE, Rine J. Metabolism and epigenetics. Annu Rev Cell Dev Biol. 2015; 31: 473-96, CrossRef.

Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc Natl Acad Sci USA. 2007; 104: 13056-61, CrossRef.

Katoh Y, Ikura T, Hoshikawa Y, Tashiro S, Ito T, Ohta M, et al. Methionine adenosyltransferase II serves as a transcriptional corepressor of Maf oncoprotein. Mol Cell. 2011; 41: 554-66, CrossRef.

Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci USA. 1964; 51: 786-94, CrossRef.

Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009; 324: 651-4, CrossRef.

Li X, Egervari G, Wang Y, Berger SL, Lu Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat Rev Mol Cell Biol. 2018; 19: 563-78, CrossRef.

Yan H, Simola DF, Bonasio R, Liebig J, Berger SL, Reinberg D. Eusocial insects as emerging models for behavioural epigenetics. Nat Rev Genet. 2014; 15: 677-88, CrossRef.

Gut P, Verdin E. The nexus of chromatin regulation and intermediary metabolism. Nature. 2013; 502: 489-98, CrossRef.

Lempradl A, Pospisilik JA, Penninger JM. Exploring the emerging complexity in transcriptional regulation of energy homeostasis. Nat Rev Genet. 2015; 16: 665-81, CrossRef.

Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nature Rev Mol Cell Biol. 2011; 12: 21-35, CrossRef.

Cantó C, Auwerx J. AMP-activated protein kinase and its downstream transcriptional pathways. Cell Mol Life Sci. 2010; 67: 3407-23, CrossRef.

Houtkooper RH, Williams RW, Auwerx J. Metabolic networks of longevity. Cell. 2010; 142: 9-14, CrossRef.

Fontana L, Partridge L, Longo VD. Extending healthy life span — from yeast to humans. Science. 2010; 328: 321-6, CrossRef.

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, CrossRef.

Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003; 425: 191-6, CrossRef.

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

Braunstein M, Sobel RE, Allis CD, Turner BM, Broach JR. Efficient transcriptional silencing in Saccharomyces cerevisiae requires a heterochromatin histone acetylation pattern. Mol Cell Biol. 1996; 16: 4349-56, CrossRef.

Haigis MC, Mostoslavsky R, Haigis KM, Fahie K, Christodoulou DC, Murphy AJ, et al. SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic β-cells. Cell. 2006; 126: 941-54, CrossRef.

Liszt G, Ford E, Kurtev M, Guarente L. Mouse Sir2 homolog SIRT6 is a nuclear ADP-ribosyltransferase. J Biol Chem. 2005; 280: 21313-20, CrossRef.

Michishita E, McCord RA, Berber E, Kioi M, Padilla-Nash H, Damian M, et al. SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature. 2008; 452: 492-6, CrossRef.

Zhong L, D'Urso A, Toiber D, Sebastian C, Henry RE, Vadysirisack DD, et al. The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1α. Cell. 2010; 140: 280-93, CrossRef.

Nakagawa T, Lomb DJ, Haigis MC, Guarente L. SIRT5 deacetylates carbamoyl phosphate synthetase 1 and regulates the urea cycle. Cell. 2009; 137: 560-70, CrossRef.

Peng C, Lu Z, Xie Z, Cheng Z, Chen Y, Tan M, et al. The first identification of lysine malonylation substrates and its regulatory enzyme. Mol Cell Proteomics. 2011; 10: M111.012658, CrossRef.

Du J, Zhou Y, Su X, Yu JJ, Khan S, Jiang H, et al. Sirt5 is an NAD-dependent protein lysine demalonylase and desuccinylase. Science. 2011; 334: 806-9, CrossRef.

Houtkooper RH, Cantó C, Wanders RJ, Auwerx J. The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. Endocr Rev. 2010; 31: 194-223, CrossRef.

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, CrossRef.

Anderson RM, Bitterman KJ, Wood JG, Medvedik O, Sinclair DA. Nicotinamide and PNC1 govern lifespan extension by calorie restriction in Saccharomyces cerevisiae. Nature. 2003; 423: 181-5, CrossRef.

Kustatscher G, Hothorn M, Pugieux C, Scheffzek K, Ladurner AG. Splicing regulates NAD metabolite binding to histone macroH2A. Nature Struct Mol Biol. 2005; 12: 624-5, CrossRef.

Liou GG, Tanny JC, Kruger RG, Walz T, Moazed D. Assembly of the SIR complex and its regulation by O-acetyl-ADP-ribose, a product of NAD-dependent histone deacetylation. Cell. 2005; 121: 515-27, CrossRef.

Tong L, Denu JM. Function and metabolism of sirtuin metabolite O-acetyl-ADP-ribose. Biochim Biophys Acta. 2010; 1804: 1617-25, CrossRef.

Zhong L, Mostoslavsky R. Fine tuning our cellular factories: sirtuins in mitochondrial biology. Cell Metab. 2011; 13: 621-6, CrossRef.

Verdin E, Hirschey MD, Finley LWS, Haigis MC. Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem Sci. 2010; 35: 669-75, CrossRef.

Lombard DB, Alt FW, Cheng HL, Bunkenborg J, Streeper RS, Mostoslavsky R, et al. Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Mol Cell Biol. 2007; 27: 8807-14, CrossRef.

Hirschey MD, Shimazu T, Goetzman E, Jing E, Schwer B, Lombard DB, et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature. 2010; 464: 121-5, CrossRef.

Hirschey MD, Shimazu T, Jing E, Grueter CA, Collins AM, Aouizerat B, et al. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol Cell. 2011; 21: 177-90, CrossRef.

Shimazu T, Hirschey MD, Hua L, Dittenhafer-Reed KE, Schwer B, Lombard DB, et al. SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metab. 2010; 12: 654-61, CrossRef.

Someya S, Yu W, Hallows WC, Xu J, Vann JM, Leeuwenburgh C, et al. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell. 2010; 143: 802-12, CrossRef.

Ahn BH, Kim H-S, Song S, Lee IH, Liu J, Vassilopoulos A, et al. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci USA. 2008; 105: 14447-52, CrossRef.

Finley LW, Haas W, Desquiret-Dumas V, Wallace DC, Procaccio V, Gygi SP, et al. Succinate dehydrogenase is a direct target of sirtuin 3 deacetylase activity. PLoS ONE. 2011; 6: e23295, CrossRef.

Jing E, Emanuelli B, Hirschey MD, Boucher J, Lee KY, Lombard D, et al. Sirtuin-3 (Sirt3) regulates skeletal muscle metabolism and insulin signaling via altered mitochondrial oxidation and reactive oxygen species production. Proc Natl Acad Sci USA. 2011; 108: 14608-13, CrossRef.

Lavu S, Boss O, Elliott PJ, Lambert PD. Sirtuins — novel therapeutic targets to treat age-associated diseases. Nature Rev Drug Discov. 2008; 7: 841-53, CrossRef.

Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α. Cell. 2006; 127: 1109-22, CrossRef.

Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; 444: 337-42, CrossRef.

Pearson KJ, Baur JA, Lewis KN, Peshkin L, Price NL, Labinskyy N, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008; 8: 157-68, CrossRef.

Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011; 14: 612-22, CrossRef.

Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, Gagne DJ, et al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007; 450: 712-6, CrossRef.

Feige JN, Lagouge M, Canto C, Strehle A, Houten SM, Milne JC, et al. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 2008; 8: 347-58, CrossRef.

Minor RK, Baur JA, Gomes AP, Ward TM, Csiszar A, Mercken EM, et al. SRT1720 improves survival and healthspan of obese mice. Sci Rep. 2011; 1: 70, CrossRef.

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

Brunet A, Rando TA. Interaction between epigenetic and metabolism in aging stem cells. Curr Opin Cell Biol. 2017; 45: 1-7, CrossRef.

Goodell MA, Rando TA. Stem cells and healthy aging. Science. 2015; 350: 1199-204, CrossRef.

Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 2007; 5: e201, CrossRef.

Liu L, Cheung TH, Charville GW, Hurgo BM, Leavitt T, Shih J, et al. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013; 4: 189-204, CrossRef.

Avgustinova A, Benitah SA. Epigenetic control of adult stem cell function. Nat Rev Mol Cell Biol. 2016; 17: 643-58, CrossRef.

Mayle A, Yang L, Rodriguez B, Zhou T, Chang E, Curry CV, et al. Dnmt3a loss predisposes murine hematopoietic stem cells to malignant transformation. Blood. 2015; 125: 629-38, CrossRef.

Challen GA, Sun D, Mayle A, Jeong M, Luo M, Rodriguez B, et al. Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells. Cell Stem Cell. 2014; 15: 350-364, CrossRef.

Ugarte F, Sousae R, Cinquin B, Martin EW, Krietsch J, Sanchez G, et al. Progressive chromatin condensation and H3K9 methylation regulate the differentiation of embryonic and hematopoietic stem cells. Stem Cell Rep. 2015; 5: 728-40, CrossRef.

Boonsanay V, Zhang T, Georgieva A, Kostin S, Qi H, Yuan X, et al. Regulation of skeletal muscle stem cell quiescence by Suv4-20h1-dependent facultative heterochromatin formation. Cell Stem Cell. 2016; 18: 229-42, CrossRef.

Benayoun BA, Pollina EA, Ucar D, Mahmoudi S, Karra K, Wong ED, et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell. 2014; 158: 673-88, CrossRef.

Ren R, Ocampo A, Liu GH, Belmonte CI. Regulation of stem cell aging by metabolism and epigenetics. Cell Metab. 2017, 26: 460-74, CrossRef.

Chandel NS, Jasper H, Ho TT, Passegue E. Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing. Nat Cell Biol. 2016; 18: 823-32, CrossRef.

Wu J, Ocampo A, Izpisua Belmonte JC. Cellular metabolism and induced pluripotency. Cell. 2016; 166: 1371-85, CrossRef.




DOI: https://doi.org/10.18585/inabj.v11i1.780

Copyright (c) 2019 The Prodia Education and Research Institute

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

 

Indexed by:

                  

               

                   

 

 

The Prodia Education and Research Institute