101
|
Guo J, Pereira TJ, Mori Y, Gonzalez Medina M, Breen DM, Dalvi PS, Zhang H, McCole DF, McBurney MW, Heximer SP, Tsiani EL, Dolinsky VW, Giacca A. Resveratrol Inhibits Neointimal Growth after Arterial Injury in High-Fat-Fed Rodents: The Roles of SIRT1 and AMPK. J Vasc Res 2020; 57:325-340. [PMID: 32777783 DOI: 10.1159/000509217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
We have shown that both insulin and resveratrol (RSV) decrease neointimal hyperplasia in chow-fed rodents via mechanisms that are in part overlapping and involve the activation of endothelial nitric oxide synthase (eNOS). However, this vasculoprotective effect of insulin is abolished in high-fat-fed insulin-resistant rats. Since RSV, in addition to increasing insulin sensitivity, can activate eNOS via pathways that are independent of insulin signaling, such as the activation of sirtuin 1 (SIRT1) and AMP-activated kinase (AMPK), we speculated that unlike insulin, the vasculoprotective effect of RSV would be retained in high-fat-fed rats. We found that high-fat feeding decreased insulin sensitivity and increased neointimal area and that RSV improved insulin sensitivity (p < 0.05) and decreased neointimal area in high-fat-fed rats (p < 0.05). We investigated the role of SIRT1 in the effect of RSV using two genetic mouse models. We found that RSV decreased neointimal area in high-fat-fed wild-type mice (p < 0.05), an effect that was retained in mice with catalytically inactive SIRT1 (p < 0.05) and in heterozygous SIRT1-null mice. In contrast, the effect of RSV was abolished in AMKPα2-null mice. Thus, RSV decreased neointimal hyperplasia after arterial injury in both high-fat-fed rats and mice, an effect likely not mediated by SIRT1 but by AMPKα2.
Collapse
Affiliation(s)
- June Guo
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Troy J Pereira
- Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yusaku Mori
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | | | - Danna M Breen
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Prasad S Dalvi
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Biology Department, Morosky College of Health Professions and Sciences, Gannon University, Erie, Pennsylvania, USA
| | - Hangjun Zhang
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Declan F McCole
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Michael W McBurney
- Program in Cancer Therapeutics, Ottawa Hospital Research Institute, Departments of Medicine and Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Scott P Heximer
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Evangelia L Tsiani
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Vernon W Dolinsky
- Department of Pharmacology and Therapeutics, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada, .,Department of Medicine, University of Toronto, Toronto, Ontario, Canada, .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada, .,Banting and Best Diabetes Centre, University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada,
| |
Collapse
|
102
|
Ziman B, Karabinis P, Barghouth P, Oviedo NJ. Sirtuin-1 regulates organismal growth by altering feeding behavior and intestinal morphology in planarians. J Cell Sci 2020; 133:jcs239467. [PMID: 32265271 PMCID: PMC7272345 DOI: 10.1242/jcs.239467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 03/19/2020] [Indexed: 01/03/2023] Open
Abstract
Nutrient availability upon feeding leads to an increase in body size in the planarian Schmidtea mediterranea However, it remains unclear how food consumption integrates with cell division at the organismal level. Here, we show that the NAD-dependent protein deacetylases sirtuins are evolutionarily conserved in planarians, and specifically demonstrate that the homolog of human sirtuin-1 (SIRT1) (encoded by Smed-Sirt-1), regulates organismal growth by impairing both feeding behavior and intestinal morphology. Disruption of Smed-Sirt-1 with RNAi or pharmacological inhibition of Sirtuin-1 leads to reduced animal growth. Conversely, enhancement of Sirtuin-1 activity with resveratrol accelerates growth. Differences in growth rates were associated with changes in the amount of time taken to locate food and overall food consumption. Furthermore, Smed-Sirt-1(RNAi) animals displayed reduced cell death and increased stem cell proliferation accompanied by impaired expression of intestinal lineage progenitors and reduced branching of the gut. Taken together, our findings indicate that Sirtuin-1 is a crucial metabolic hub capable of controlling animal behavior, tissue renewal and morphogenesis of the adult intestine.
Collapse
Affiliation(s)
- Benjamin Ziman
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Peter Karabinis
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Paul Barghouth
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
- Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| |
Collapse
|
103
|
Mishra JS, Zhao H, Hattis S, Kumar S. Elevated Glucose and Insulin Levels Decrease DHA Transfer across Human Trophoblasts via SIRT1-Dependent Mechanism. Nutrients 2020; 12:nu12051271. [PMID: 32365792 PMCID: PMC7284516 DOI: 10.3390/nu12051271] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/30/2022] Open
Abstract
Gestational diabetes mellitus (GDM) results in reduced docosahexaenoic acid (DHA) transfer to the fetus, likely due to placental dysfunction. Sirtuin-1 (SIRT1) is a nutrient sensor and regulator of lipid metabolism. This study investigated whether the high glucose and insulin condition of GDM regulates DHA transfer and expression of fatty acid transporters and if this effect is related to SIRT1 expression and function. Syncytialized primary human trophoblasts were treated with and without glucose (25 mmol/L) and insulin (10-7 mol/L) for 72 h to mimic the insulin-resistance conditions of GDM pregnancies. In control conditions, DHA transfer across trophoblasts increased in a time- and dose-dependent manner. Exposure to GDM conditions significantly decreased DHA transfer, but increased triglyceride accumulation and fatty acid transporter expression (CD36, FABP3, and FABP4). GDM conditions significantly suppressed SIRT1 mRNA and protein expression. The SIRT1 inhibitor decreased DHA transfer across control trophoblasts, and recombinant SIRT1 and SIRT1 activators restored the decreased DHA transport induced by GDM conditions. The results demonstrate a novel role of SIRT1 in the regulation of DHA transfer across trophoblasts. The suppressed SIRT1 expression and the resultant decrease in placental DHA transfer caused by high glucose and insulin levels suggest new insights of molecular mechanisms linking GDM to fetal DHA deficiency.
Collapse
Affiliation(s)
- Jay S. Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Hanjie Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Sari Hattis
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (J.S.M.); (H.Z.); (S.H.)
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
- Correspondence: ; Tel.: +1-608-265-1046
| |
Collapse
|
104
|
Hudec M, Dankova P, Solc R, Bettazova N, Cerna M. Epigenetic Regulation of Circadian Rhythm and Its Possible Role in Diabetes Mellitus. Int J Mol Sci 2020; 21:E3005. [PMID: 32344535 PMCID: PMC7215839 DOI: 10.3390/ijms21083005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
This review aims to summarize the knowledge about the relationship between circadian rhythms and their influence on the development of type 2 diabetes mellitus (T2DM) and metabolic syndrome. Circadian rhythms are controlled by internal molecular feedback loops that synchronize the organism with the external environment. These loops are affected by genetic and epigenetic factors. Genetic factors include polymorphisms and mutations of circadian genes. The expression of circadian genes is regulated by epigenetic mechanisms that change from prenatal development to old age. Epigenetic modifications are influenced by the external environment. Most of these modifications are affected by our own life style. Irregular circadian rhythm and low quality of sleep have been shown to increase the risk of developing T2DM and other metabolic disorders. Here, we attempt to provide a wide description of mutual relationships between epigenetic regulation, circadian rhythm, aging process and highlight new evidences that show possible therapeutic advance in the field of chrono-medicine which will be more important in the upcoming years.
Collapse
Affiliation(s)
- Michael Hudec
- Department of Medical Genetics, Third Faculty of Medicine, Charles University; Ruská 87, 100 00 Prague, Czech Republic; (N.B.); (M.C.)
| | - Pavlina Dankova
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University; Viničná 7, 128 00 Prague, Czech Republic; (P.D.); (R.S.)
| | - Roman Solc
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University; Viničná 7, 128 00 Prague, Czech Republic; (P.D.); (R.S.)
| | - Nardjas Bettazova
- Department of Medical Genetics, Third Faculty of Medicine, Charles University; Ruská 87, 100 00 Prague, Czech Republic; (N.B.); (M.C.)
| | - Marie Cerna
- Department of Medical Genetics, Third Faculty of Medicine, Charles University; Ruská 87, 100 00 Prague, Czech Republic; (N.B.); (M.C.)
| |
Collapse
|
105
|
Yan Q, Huang H, Lu S, Ou B, Feng J, Shan W, Li H, Wang Z, Hong A, Ma Y. PACAP ameliorates fertility in obese male mice via PKA/CREB pathway‐dependent Sirt1 activation and p53 deacetylation. J Cell Physiol 2020; 235:7465-7483. [DOI: 10.1002/jcp.29651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/04/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Qiuxia Yan
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
- Center for Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's HospitalQingyuan China
| | - Hongke Huang
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Shiyin Lu
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Biqian Ou
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Jia Feng
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Wailan Shan
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Huixian Li
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Zixian Wang
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - An Hong
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| | - Yi Ma
- Department of Cellular BiologyInstitute of BiomedicineNational Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan UniversityGuangzhou China
| |
Collapse
|
106
|
Hypothalamic NAD +-Sirtuin Axis: Function and Regulation. Biomolecules 2020; 10:biom10030396. [PMID: 32143417 PMCID: PMC7175325 DOI: 10.3390/biom10030396] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
The rapidly expanding elderly population and obesity endemic have become part of continuing global health care problems. The hypothalamus is a critical center for the homeostatic regulation of energy and glucose metabolism, circadian rhythm, and aging-related physiology. Nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase sirtuins are referred to as master metabolic regulators that link the cellular energy status to adaptive transcriptional responses. Mounting evidence now indicates that hypothalamic sirtuins are essential for adequate hypothalamic neuronal functions. Owing to the NAD+-dependence of sirtuin activity, adequate hypothalamic NAD+ contents are pivotal for maintaining energy homeostasis and circadian physiology. Here, we comprehensively review the regulatory roles of the hypothalamic neuronal NAD+-sirtuin axis in a normal physiological context and their changes in obesity and the aging process. We also discuss the therapeutic potential of NAD+ biology-targeting drugs in aging/obesity-related metabolic and circadian disorders.
Collapse
|
107
|
Ma L, Wang R, Wang H, Zhang Y, Zhao Z. Long-term caloric restriction activates the myocardial SIRT1/AMPK/PGC-1α pathway in C57BL/6J male mice. Food Nutr Res 2020; 64:3668. [PMID: 32082101 PMCID: PMC7007760 DOI: 10.29219/fnr.v64.3668] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 12/22/2019] [Accepted: 01/07/2020] [Indexed: 01/23/2023] Open
Abstract
Background Caloric restriction (CR) can help in improving heart function. There is as yet no consensus on the mechanism of the effect of CR. Silent mating-type information regulation 1 (SIRT1), adenosine monophosphate-activated protein kinase (AMPK), and mTOR are key players in metabolic stress management. We aimed to explore the effect of CR on the myocardial SIRT1/AMPK/mTOR pathway in mice. Methods Thirty-six 6-week-old male C57BL/6J mice were randomly divided into three groups: normal control group (NC group, n = 12), high-energy group (HE group, n = 12) and CR group (n = 12) according to different diets. After 11 months, western blot was used to examine proteins such as p-AMPK, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), SIRT1, and p-mTOR, whereas real-time PCR was used to examine the expression of AMPK, PGC-1α, and SIRT1 transcripts. Results Compared to the HE group, the CR group displayed increased expression of myocardial p-AMPK protein, SIRT1 protein and mRNA, and PGC-1a mRNA. However, no difference was observed in the expression of p-mTOR protein and mTOR mRNA in the myocardium among the three groups. Conclusions CR improves the SIRT1/AMPK/PGC-1α pathway in mice myocardium with no effect on the mTOR pathway.
Collapse
Affiliation(s)
- Lina Ma
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China.,Central Laboratory, Xuanwu Hospital Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Hongjuan Wang
- Department of Biochemistry and Molecular Biology, Yanjing Medical College, Capital Medical University, Beijing, China
| | - Yaxin Zhang
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Zhiwei Zhao
- Central Laboratory, Xuanwu Hospital Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
108
|
Yang X, Sun X, Wu J, Ma J, Si P, Yin L, Zhang Y, Yan LJ, Zhang C. Regulation of the SIRT1 signaling pathway in NMDA-induced Excitotoxicity. Toxicol Lett 2020; 322:66-76. [PMID: 31945382 DOI: 10.1016/j.toxlet.2020.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/12/2019] [Accepted: 01/12/2020] [Indexed: 12/20/2022]
Abstract
Silent Information Regulator 1 (SIRT1), an NAD+-dependent deacetylase, contributes to the neuroprotective effect. However, intracellular signaling pathways that affect SIRT1 function remain unknown. It is well known that N-methyl-D-aspartate (NMDA) receptor activation induces calcium influx which then activates PKC, and SIRT1 is a mRNA target for HuR protein. We hypothesize that Ca2+-PKC-HuR-SIRT1 pathway modulates SIRT1 function. The present study is to investigate the potential pathway of SIRT1 in the SH-SY5Y cell line as an in vitro model of NMDA-induced neurotoxicity. The results showed that: (1) SIRT1 levels were downregulated in NMDA model; (2) NMDA induced an increase in serine phosphorylation of HuR, while inhibition of serine phosphorylation of HuR increased SIRT1 levels, promoting cell survival; (3) PKC inhibitor (Gö 6976) reversed NMDA insults and also suppressed serine phosphorylation of HuR; (4) 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular calcium chelator, fully reversed NMDA insults and also inhibited PKC activity evoked by NMDA. These results indicate that intracellular elevated Ca2+ activates PKC, which phosphorylates HuR and then promotes SIRT1 mRNA decay and subsequent neuronal death in NMDA model. Therefore, the study suggests that inhibition of Ca2+-PKC-HuR-SIRT1 pathway could be an effective strategy for preventing certain neurological diseases related to NMDA excitotoxicity.
Collapse
Affiliation(s)
- Xiaorong Yang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China.
| | - Xuefei Sun
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China; The People's Hospital of Funing, Qinhuangdao 066300, Hebei Province, PR China
| | - Jinzi Wu
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jinteng Ma
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China
| | - Peipei Si
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050071, Hebei Province, PR China
| | - Litian Yin
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China
| | - Yu Zhang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Ce Zhang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and the Department of Physiology, Shanxi Medical University, #56 Xin Jian South Road, Taiyuan 030001, Shanxi Province, PR China
| |
Collapse
|
109
|
Li B, Li M, Li X, Li H, Lai Y, Huang S, He X, Si X, Zheng H, Liao W, Liao Y, Bin J. Sirt1-inducible deacetylation of p21 promotes cardiomyocyte proliferation. Aging (Albany NY) 2019; 11:12546-12567. [PMID: 31881009 PMCID: PMC6949046 DOI: 10.18632/aging.102587] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022]
Abstract
Inducing cardiomyocyte proliferation is a hopeful approach for cardiac regeneration following myocardial infarction. Previous studies have shown that p21 inhibits the cardiomyocyte proliferation and cardiac regeneration. Deacetylation of p21 by Sirt1 deacetylase may reduce p21 abundance and remove p21-induced cell cycle arrest. However, whether p21 deacetylation and Sirt1 deacetylate control cardiomyocyte proliferation is unclear. Here, we show that acetylation of p21 induces cardiomyocyte proliferation arrest, whereas blocking the acetylation of p21 increases cardiomyocyte proliferation. P21 can be acetylated by Sirt1, and Sirt1 activate p21 ubiquitination through deacetylation. Additionally, overexpression of Sirt1 induces EdU-, pH3-, and Aurora B-positive cardiomyocytes in neonatal and adult mice. In contrast, depletion of Sirt1 reduces cardiomyocyte proliferation in vitro and in vivo. Moreover, Sirt1 protects cardiac function, reduces cardiac remodeling, inhibits cardiomyocyte apoptosis, and attenuates cardiomyocyte hypertrophy post-myocardial infarction. These results suggest that Sirt1-induced p21 deacetylation plays an essential role in cardiomyocyte proliferation and that it could be a novel therapeutic strategy for myocardial infarction.
Collapse
Affiliation(s)
- Bing Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,School of Medicine, Guizhou University, Guiyang, Guizhou 550025, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Mengsha Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinzhong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Hairui Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanxian Lai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Senlin Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Xiang He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Xiaoyun Si
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| |
Collapse
|
110
|
Nevoral J, Landsmann L, Stiavnicka M, Hosek P, Moravec J, Prokesova S, Rimnacova H, Koutna E, Klein P, Hoskova K, Zalmanova T, Fenclova T, Petr J, Kralickova M. Epigenetic and non-epigenetic mode of SIRT1 action during oocyte meiosis progression. J Anim Sci Biotechnol 2019; 10:67. [PMID: 31413827 PMCID: PMC6688279 DOI: 10.1186/s40104-019-0372-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
Background SIRT1 histone deacetylase acts on many epigenetic and non-epigenetic targets. It is thought that SIRT1 is involved in oocyte maturation; therefore, the importance of the ooplasmic SIRT1 pool for the further fate of mature oocytes has been strongly suggested. We hypothesised that SIRT1 plays the role of a signalling molecule in mature oocytes through selected epigenetic and non-epigenetic regulation. Results We observed SIRT1 re-localisation in mature oocytes and its association with spindle microtubules. In mature oocytes, SIRT1 distribution shows a spindle-like pattern, and spindle-specific SIRT1 action decreases α-tubulin acetylation. Based on the observation of the histone code in immature and mature oocytes, we suggest that SIRT1 is mostly predestined for an epigenetic mode of action in the germinal vesicles (GVs) of immature oocytes. Accordingly, BML-278-driven trimethylation of lysine K9 in histone H3 in mature oocytes is considered to be a result of GV epigenetic transformation. Conclusions Taken together, our observations point out the dual spatiotemporal SIRT1 action in oocytes, which can be readily switched from the epigenetic to non-epigenetic mode of action depending on the progress of meiosis. Electronic supplementary material The online version of this article (10.1186/s40104-019-0372-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jan Nevoral
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic.,2Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, 301 66 Pilsen, Czech Republic
| | - Lukas Landsmann
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic.,3Faculty of Science, Charles University, Albertov 2038/6, 128 00 Prague, Czech Republic
| | - Miriam Stiavnicka
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Petr Hosek
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Jiri Moravec
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Sarka Prokesova
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic.,4Faculty of Agriculture, Food and Natural Resources, Czech University of Life Sciences in Prague, Kamycka 129, 165 00 Praha-Suchdol, Czech Republic.,5Institute of Animal Science, Pratelstvi 815/107, 104 00, Prague 10-Uhrineves, Czech Republic
| | - Hedvika Rimnacova
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Eliska Koutna
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic.,4Faculty of Agriculture, Food and Natural Resources, Czech University of Life Sciences in Prague, Kamycka 129, 165 00 Praha-Suchdol, Czech Republic
| | - Pavel Klein
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Kristyna Hoskova
- 5Institute of Animal Science, Pratelstvi 815/107, 104 00, Prague 10-Uhrineves, Czech Republic
| | - Tereza Zalmanova
- 5Institute of Animal Science, Pratelstvi 815/107, 104 00, Prague 10-Uhrineves, Czech Republic
| | - Tereza Fenclova
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic
| | - Jaroslav Petr
- 5Institute of Animal Science, Pratelstvi 815/107, 104 00, Prague 10-Uhrineves, Czech Republic
| | - Milena Kralickova
- 1Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, 323 00 Pilsen, Czech Republic.,2Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, 301 66 Pilsen, Czech Republic
| |
Collapse
|
111
|
Zhang W, Feng Y, Guo Q, Guo W, Xu H, Li X, Yi F, Guan Y, Geng N, Wang P, Cao L, O'Rourke BP, Jo J, Kwon J, Wang R, Song X, Lee IH, Cao L. SIRT1 modulates cell cycle progression by regulating CHK2 acetylation-phosphorylation. Cell Death Differ 2019; 27:482-496. [PMID: 31209362 PMCID: PMC7206007 DOI: 10.1038/s41418-019-0369-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 04/26/2019] [Accepted: 05/27/2019] [Indexed: 12/21/2022] Open
Abstract
Both the stress-response protein, SIRT1, and the cell cycle checkpoint kinase, CHK2, play critical roles in aging and cancer via the modulation of cellular homeostasis and the maintenance of genomic integrity. However, the underlying mechanism linking the two pathways remains elusive. Here, we show that SIRT1 functions as a modifier of CHK2 in cell cycle control. Specifically, SIRT1 interacts with CHK2 and deacetylates it at lysine 520 residue, which suppresses CHK2 phosphorylation, dimerization, and thus activation. SIRT1 depletion induces CHK2 hyperactivation-mediated cell cycle arrest and subsequent cell death. In vivo, genetic deletion of Chk2 rescues the neonatal lethality of Sirt1−/− mice, consistent with the role of SIRT1 in preventing CHK2 hyperactivation. Together, these results suggest that CHK2 mediates the function of SIRT1 in cell cycle progression, and may provide new insights into modulating cellular homeostasis and maintaining genomic integrity in the prevention of aging and cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yanling Feng
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Qiqiang Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Wendong Guo
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Hongde Xu
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Xiaoman Li
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Fei Yi
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yi Guan
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Nanxi Geng
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Pingyuan Wang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Longyue Cao
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Brian P O'Rourke
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Juhyeon Jo
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea
| | - Jiyun Kwon
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea
| | - Ruihong Wang
- Faculty of Health Science, University of Macau, Macau, China
| | - Xiaoyu Song
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| | - In Hye Lee
- Department of Life Science, College of Natural Science Office #106, Science building C, Ewha Womans University 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, South Korea.
| | - Liu Cao
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
112
|
Choi I, Rickert E, Fernandez M, Webster NJG. SIRT1 in Astrocytes Regulates Glucose Metabolism and Reproductive Function. Endocrinology 2019; 160:1547-1560. [PMID: 31127273 PMCID: PMC6542483 DOI: 10.1210/en.2019-00223] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
Abstract
Sirtuin 1 (Sirt1) is an NAD-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, evidence suggests that SIRT1 in neurons plays a role in the central regulation of energy balance and reproduction, but no studies have addressed the contribution of astrocytes. We show here that overexpression of SIRT1 in astrocytes causes markedly increased food intake, body weight gain, and glucose intolerance, but expression of a deacetylase-deficient SIRT1 mutant decreases food intake and body weight and improves glucose tolerance, particularly in female mice. Paradoxically, the effect of these SIRT1 mutants on insulin tolerance was reversed, with overexpression showing greater insulin sensitivity. The mice overexpressing SIRT1 were more active, generated more heat, and had elevated oxygen consumption, possibly in compensation for the increased food intake. The female overexpressing mice were also more sensitive to diet-induced obesity. Reproductively, the mice expressing the deacetylase-deficient SIRT1 mutant had impaired estrous cycles, decreased LH surges, and fewer corpora lutea, indicating decreased ovulation. The GnRH neurons were responsive to kisspeptin stimulation, but hypothalamic expression of Kiss1 was reduced in the mutant mice. Our results showed that SIRT1 signaling in astrocytes can contribute to metabolic and reproductive regulation independent of SIRT1 effects in neurons.
Collapse
Affiliation(s)
- Irene Choi
- VA San Diego Healthcare System, San Diego, California
| | - Emily Rickert
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California
| | - Marina Fernandez
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California
| | - Nicholas J G Webster
- VA San Diego Healthcare System, San Diego, California
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California
- Moores Cancer Center, University of California San Diego, La Jolla, California
- Correspondence: Nicholas J. G. Webster, PhD, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093. E-mail:
| |
Collapse
|
113
|
Wang H, Hu Z, Wu J, Mei Y, Zhang Q, Zhang H, Miao D, Sun W. Sirt1 Promotes Osteogenic Differentiation and Increases Alveolar Bone Mass via Bmi1 Activation in Mice. J Bone Miner Res 2019; 34:1169-1181. [PMID: 30690778 DOI: 10.1002/jbmr.3677] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 12/27/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022]
Abstract
Sirtuin 1 (Sirt1), a protein deacetylase, is a novel target for bone metabolism. To investigate whether overexpression of Sirt1 in mandibular mesenchymal stem cells (M-MSCs) increased alveolar bone mass in vivo, we generated Sirt1 transgenic mice (Sirt1TG ), with Sirt1 gene expression driven by the Prx1 gene, which represents the mesenchymal lineage. Our results demonstrated that overexpression of Sirt1 in M-MSCs increased the alveolar bone volume in 1-month-old, 9-month-old, and 18-month-old Sirt1TG mice compared with age-matched wild-type (WT) mice, and in ovariectomized Sirt1TG mice compared with ovariectomized WT mice by stimulating M-MSC differentiation into osteoblasts. Treatment with resveratrol, a Sirt1 activator, increased Sirt1 binding with Bmi1 and reduced Bmi1 acetylation in a dose-dependent manner demonstrated in M-MSC cultures. Both treatment with resveratrol in M-MSC cultures and overexpressed Sirt1 in M-MSCs ex vivo cultures increased nuclear translocation of Bmi1. Furthermore, we demonstrated that deletion of Bmi1 blocked the increased alveolar bone volume in Sirt1TG mice. The Sirt1 activator resveratrol inhibited human MSC senescence and promoted their differentiation into osteoblasts, which were associated with upregulating the expression levels of Sirt1 and nuclear translocation of Bmi1. The present results suggested that Sirt1 promotes MSC proliferation and osteogenic differentiation, inhibits MSC senescence to increase alveolar bone volume by promoting the deacetylation and nuclear translocation of Bmi1. Thus, our study elucidated the mechanism by which Sirt1 increases alveolar bone mass, and these findings are important for the clinical application of the Sirt1 activator resveratrol for the promotion of alveolar bone formation and prevention of alveolar bone loss. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zixuan Hu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Jun Wu
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yukun Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Hengwei Zhang
- Center for Musculoskeletal Research (CMSR), Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dengshun Miao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
114
|
Kabiljo J, Murko C, Pusch O, Zupkovitz G. Spatio-temporal expression profile of sirtuins during aging of the annual fish Nothobranchius furzeri. Gene Expr Patterns 2019; 33:11-19. [PMID: 31075496 DOI: 10.1016/j.gep.2019.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/24/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022]
Abstract
The founding member of the sirtuin family, yeast Sir2, was the first evolutionarily conserved gene to be identified as a regulator of longevity. Sirtuins constitute a protein family of metabolic sensors, translating changes in NAD + levels into adaptive responses, thereby acting as crucial regulators of the network that controls energy homeostasis and as such determines healthspan. In mammals the sirtuin family comprises seven proteins, SIRT1-SIRT7, which vary in tissue specificity, subcellular localization, enzymatic activity and targets. Here, we report the identification and a detailed spatio-temporal expression profile of sirtuin genes in the short-lived fish Nothobranchius furzeri, from embryogenesis to late adulthood, mapping its entire life cycle. Database exploration of the recently published N. furzeri genome revealed eight orthologues corresponding to the seven known mammalian sirtuins, including two copies of the sirt5 gene. Phylogenetic analysis showed high cross species similarity of individual sirtuins in both their overall amino acid sequence and catalytic domain, suggesting a high degree of functional conservation. Moreover, we show that N. furzeri sirtuins exhibit ubiquitous and wide tissue distribution with a unique spatial expression pattern for each individual member of this enzyme family. Specifically, we observed a transcriptional down-regulation of several sirtuin genes with age, most significantly sirt1, sirt5a, sirt6 and sirt7 in a wide range of functionally distinct tissues. Overall, this spatio-temporal expression analysis provides the foundation for future research, both into genetic and pharmacological manipulation of this important group of enzymes in Nothobranchius furzeri, an emerging model organism for aging research.
Collapse
Affiliation(s)
- Julijan Kabiljo
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, 1090, Austria.
| | - Christina Murko
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, 1090, Austria.
| | - Oliver Pusch
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, 1090, Austria.
| | - Gordin Zupkovitz
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
115
|
Lin JB, Kubota S, Mostoslavsky R, Apte RS. Role of Sirtuins in Retinal Function Under Basal Conditions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1074:561-567. [PMID: 29721988 DOI: 10.1007/978-3-319-75402-4_68] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sirtuins are NAD+-dependent enzymes that govern cellular homeostasis by regulating the acylation status of their diverse target proteins. We recently demonstrated that both rod and cone photoreceptors rely on NAMPT-mediated NAD+ biosynthesis to meet their energetic requirements. Moreover, we found that this NAD+-dependent retinal homeostasis relies, in part, on maintenance of optimal activity of the mitochondrial sirtuins and of SIRT3 in particular. Nonetheless, it is unknown whether other sirtuin family members also play important roles in retinal homeostasis. Our results suggest that SIRT1, SIRT2, SIRT4, and SIRT6 are dispensable for retinal survival at baseline, as individual deletion of each of these sirtuins does not cause retinal degeneration by fundus biomicroscopy or retinal dysfunction by ERG. These findings have significant implications and inform future studies investigating the mechanisms underlying the central role of NAD+ biosynthesis in retinal survival and function.
Collapse
Affiliation(s)
- Jonathan B Lin
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.,Neuroscience Graduate Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Shunsuke Kubota
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Raul Mostoslavsky
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Rajendra S Apte
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
116
|
Lin C, Liu M, Zhu X, Zhang M, Xu S, Wang D, Zhao Y. Cloning and expression of the lifespan-associated protein Sir2 from Daphnia pulex. Comp Biochem Physiol B Biochem Mol Biol 2019; 231:1-10. [DOI: 10.1016/j.cbpb.2019.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 11/15/2022]
|
117
|
Abstract
The sirtuin family of NAD+-dependent protein deacetylases promotes longevity and counteracts age-related diseases. One of the major targets of Sirtuins are the FoxO family of transcription factors. FoxOs play a major role in the adaptation of cells to a variety of stressors such as oxidative stress and growth factor deprivation. Studies with murine models of cell-specific loss- or gain-of-function of Sirtuins or FoxOs and with Sirtuin1 stimulators have provided novel insights into the function and signaling of these proteins on the skeleton. These studies have revealed that both Sirtuins and FoxOs acting directly in cartilage and bone cells are critical for normal skeletal development, homeostasis and that their dysregulation might contribute to skeletal disease. Deacetylation of FoxOs by Sirt1 in osteoblasts and osteoclasts stimulates bone formation and inhibits bone resorption, making Sirt1 ligands promising therapeutic agents for diseases of low bone mass. While a similar link has not been established in chondrocytes, Sirt1 and FoxOs both have chondroprotective actions, suggesting that Sirt1 activators may have similar efficacy in preventing cartilage degeneration due to aging or injury. In this review we summarize these advances and discuss their implications for the pathogenesis of age-related osteoporosis and osteoarthritis.
Collapse
Affiliation(s)
- Maria Almeida
- Department of Medicine, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopedics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Ryan M Porter
- Department of Medicine, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Orthopedics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
118
|
Zainabadi K. Drugs targeting SIRT1, a new generation of therapeutics for osteoporosis and other bone related disorders? Pharmacol Res 2019; 143:97-105. [PMID: 30862606 DOI: 10.1016/j.phrs.2019.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 12/16/2022]
Abstract
With an aging population and limited treatment options, osteoporosis currently represents a significant public health challenge. Recent animal studies indicate that longevity-associated SIRT1 may serve as an attractive pharmacological target for the treatment of osteoporosis and other bone related disorders. Pre-clinical studies demonstrate that mice treated with SIRT1 agonists show protection against age-related, post-menopausal, and disuse models of osteoporosis. Conversely, SIRT1 knockout models display low bone mass phenotypes associated with increased bone resorption and decreased bone formation. This review summarizes recent animal and human experimental data showing that pharmacological activation of SIRT1 may act in a manner that current treatments do not, namely by treating the imbalance in bone remodeling that is the root cause of osteoporosis and other bone disorders.
Collapse
Affiliation(s)
- Kayvan Zainabadi
- Glenn Center for the Science of Aging, Department of Biology, Koch Institute, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
119
|
Teng L, Fan L, Peng Y, He X, Chen H, Duan H, Yang F, Lin D, Lin Z, Li H, Shao B. Carnosic Acid Mitigates Early Brain Injury After Subarachnoid Hemorrhage: Possible Involvement of the SIRT1/p66shc Signaling Pathway. Front Neurosci 2019; 13:26. [PMID: 30890904 PMCID: PMC6411796 DOI: 10.3389/fnins.2019.00026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022] Open
Abstract
Carnosic acid (CA) has been reported to exhibit a variety of bioactivities including antioxidation, neuroprotection, and anti-inflammation; however, the impact of CA on subarachnoid hemorrhage (SAH) has never been elucidated. The current study was undertaken to explore the role of CA in early brain injury (EBI) secondary to SAH and the underlying mechanisms. Adult male Sprague-Dawley rats were perforated to mimic a clinical aneurysm with SAH. CA or vehicle was administered intravenously immediately after the SAH occurred. Mortality, SAH grade, neurologic function scores, brain water content, Evans blue extravasation, and the levels of reactive oxygen species (ROS) levels in the ipsilateral cortex were determined 24 h after the SAH occurred. Western blot, immunofluorescence, Fluoro-Jade C (FJC) and TUNEL staining were also performed. Our results showed that CA decreased ROS levels, alleviated brain edema and blood-brain barrier permeability, reduced neuronal cell death, and promoted neurologic function improvement. To probe into the potential mechanisms. We showed that CA increased SIRT1, MnSOD, and Bcl-2 expression, as well as decreased p66shc, Bax, and cleaved caspase-3 expression. Interestingly, sirtinol, a selective inhibitor of SIRT1, abolished the anti-apoptotic effects of CA. Taken together, these data revealed that CA has a neuroprotective role in EBI secondary to SAH. The potential mechanism may involve suppression of neuronal apoptosis through the SIRT1/p66shc signaling pathway. CA may provide a promising therapeutic regimen for management of SAH.
Collapse
Affiliation(s)
- Lingfang Teng
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Linfeng Fan
- Department of Pediatric Surgery, Capital Institute of Pediatrics, Beijing, China
| | - Yujiang Peng
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Xijun He
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Huihui Chen
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Hongyu Duan
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Fan Yang
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Da Lin
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Zheng Lin
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Huiyong Li
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Bo Shao
- Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
120
|
Allas L, Boumédiene K, Baugé C. Epigenetic dynamic during endochondral ossification and articular cartilage development. Bone 2019; 120:523-532. [PMID: 30296494 DOI: 10.1016/j.bone.2018.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022]
Abstract
Within the last decade epigenetics has emerged as fundamental regulator of numerous cellular processes, including those orchestrating embryonic and fetal development. As such, epigenetic factors play especially crucial roles in endochondral ossification, the process by which bone tissue is created, as well during articular cartilage formation. In this review, we summarize the recent discoveries that characterize how DNA methylation, histone post-translational modifications and non-coding RNA (e.g., miRNA and lcnRNA) epigenetically regulate endochondral ossification and chondrogenesis.
Collapse
Affiliation(s)
- Lyess Allas
- Normandie Univ, UNICAEN, EA7451 BioConnecT, Caen, France
| | | | | |
Collapse
|
121
|
Fang Y, Tang S, Li X. Sirtuins in Metabolic and Epigenetic Regulation of Stem Cells. Trends Endocrinol Metab 2019; 30:177-188. [PMID: 30630664 PMCID: PMC6382540 DOI: 10.1016/j.tem.2018.12.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/30/2018] [Accepted: 12/16/2018] [Indexed: 02/08/2023]
Abstract
Sirtuins are highly conserved NAD+-dependent enzymes that are capable of removing a wide range of lipid lysine acyl-groups from protein substrates in a NAD+-dependent manner. These NAD+-dependent activities enable sirtuins to monitor cellular energy status and modulate gene transcription, genome stability, and energy metabolism in response to environmental signals. Consequently, sirtuins are important for cell survival, stress resistance, proliferation, and differentiation. In recent years, sirtuins are increasingly recognized as crucial regulators of stem cell biology in addition to their well-known roles in metabolism and aging. This review article highlights our current knowledge on sirtuins in stem cells, including their functions in pluripotent stem cells, embryogenesis, and development as well as their roles in adult stem cell maintenance, regeneration, and aging.
Collapse
Affiliation(s)
- Yi Fang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; These authors contributed equally to this work
| | - Shuang Tang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; Current address: Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; These authors contributed equally to this work
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
122
|
Li J, Huang J, Lu J, Guo Z, Li Z, Gao H, Wang P, Luo W, Cai S, Hu Y, Guo K, Wang L, Li Z, Wang M, Zhang X, Liu P. Sirtuin 1 represses PKC-ζ activity through regulating interplay of acetylation and phosphorylation in cardiac hypertrophy. Br J Pharmacol 2019; 176:416-435. [PMID: 30414383 PMCID: PMC6329629 DOI: 10.1111/bph.14538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/20/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of PKC-ζ is closely linked to the pathogenesis of cardiac hypertrophy. PKC-ζ can be activated by certain lipid metabolites such as phosphatidylinositol (3,4,5)-trisphosphate and ceramide. However, its endogenous negative regulators are not well defined. Here, the role of the sirtuin1-PKC-ζ signalling axis and the underlying molecular mechanisms were investigated in cardiac hypertrophy. EXPERIMENTAL APPROACH Cellular hypertrophy in cultures of cardiac myocytes, from neonatal Sprague-Dawley rats, was monitored by measuring cell surface area and the mRNA levels of hypertrophic biomarkers. Interaction between sirtuin1 and PKC-ζ was investigated by co-immunoprecipitation and confocal immunofluorescence microscopy. Sirtuin1 activation was enhanced by resveratrol treatment or Ad-sirtuin1 transfection. A model of cardiac hypertrophy in Sprague-Dawley rats was established by abdominal aortic constriction surgery or induced by isoprenaline in vivo. KEY RESULTS Overexpression of PKC-ζ led to cardiac hypertrophy and increased activity of NF-κB, ERK1/2 and ERK5, which was ameliorated by sirtuin1 overexpression. Enhancement of sirtuin1 activity suppressed acetylation of PKC-ζ, hindered its binding to phosphoinositide-dependent kinase 1 and inhibited PKC-ζ phosphorylation in cardiac hypertrophy. Consequently, the downstream pathways of PKC-ζ' were suppressed in cardiac hypertrophy. This regulation loop suggests a new role for sirtuin1 in mediation of cardiac hypertrophy. CONCLUSIONS AND IMPLICATIONS Sirtuin1 is an endogenous negative regulator for PKC-ζ and mediates its activity via regulating the acetylation and phosphorylation in the pathogenesis of cardiac hypertrophy. Targeting the sirtuin1-PKC-ζ signalling axis may suggest a novel therapeutic approach against cardiac hypertrophy.
Collapse
Affiliation(s)
- Jingyan Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Junying Huang
- College of Life SciencesGuangzhou UniversityGuangzhouGuangdongChina
| | - Jing Lu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhen Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hui Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
- Department of Pharmacology, School of MedicineJishou UniversityJishouChina
| | - Panxia Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Wenwei Luo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Sidong Cai
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuehuai Hu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Kaiteng Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Luping Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhenzhen Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Minghui Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaolei Zhang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
123
|
Cakouros D, Gronthos S. Epigenetic Regulation of Bone Marrow Stem Cell Aging: Revealing Epigenetic Signatures associated with Hematopoietic and Mesenchymal Stem Cell Aging. Aging Dis 2019; 10:174-189. [PMID: 30705777 PMCID: PMC6345334 DOI: 10.14336/ad.2017.1213] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022] Open
Abstract
In this review we explore the importance of epigenetics as a contributing factor for aging adult stem cells. We summarize the latest findings of epigenetic factors deregulated as adult stem cells age and the consequence on stem cell self-renewal and differentiation, with a focus on adult stem cells in the bone marrow. With the latest whole genome bisulphite sequencing and chromatin immunoprecipitations we are able to decipher an emerging pattern common for adult stem cells in the bone marrow niche and how this might correlate to epigenetic enzymes deregulated during aging. We begin by briefly discussing the initial observations in yeast, drosophila and Caenorhabditis elegans (C. elegans) that led to the breakthrough research that identified the role of epigenetic changes associated with lifespan and aging. We then focus on adult stem cells, specifically in the bone marrow, which lends strong support for the deregulation of DNA methyltransferases, histone deacetylases, acetylates, methyltransferases and demethylases in aging stem cells, and how their corresponding epigenetic modifications influence gene expression and the aging phenotype. Given the reversible nature of epigenetic modifications we envisage “epi” targeted therapy as a means to reprogram aged stem cells into their younger counterparts.
Collapse
Affiliation(s)
- Dimitrios Cakouros
- 1Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,2South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- 1Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,2South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
124
|
Taormina G, Russo A, Latteri MA, Mirisola MG. Mitochondrion at the Crossroad Between Nutrients and Epigenome. Front Endocrinol (Lausanne) 2019; 10:673. [PMID: 31636605 PMCID: PMC6787768 DOI: 10.3389/fendo.2019.00673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Epigenetic profile is the link between the regulation of nuclear gene expression and the environment. The most important factors capable of significantly affecting the cellular environment are the amount and quality of nutrients available. Mitochondria are both involved in the production of some of the molecules capable of directly affecting the epigenome and have a critical role in the conversion of nutrients into usable energy. Carbohydrate and fats are converted into ATP, acetyl-CoA, SAM, and NADH. These high-energy substrates are, in turn, capable of driving the epigenetic profile. We describe substances capable of affecting this mechanism. On the other hand, nutritional interventions capable of reducing calories or significantly impairing the normal Acetyl-CoA production or the SAM-SAH ratio also impact chromatin methylation and histone modification, suggesting a critical role of mitochondria on nutrient-dependent epigenetic profile.
Collapse
|
125
|
Meroni SB, Galardo MN, Rindone G, Gorga A, Riera MF, Cigorraga SB. Molecular Mechanisms and Signaling Pathways Involved in Sertoli Cell Proliferation. Front Endocrinol (Lausanne) 2019; 10:224. [PMID: 31040821 PMCID: PMC6476933 DOI: 10.3389/fendo.2019.00224] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 12/16/2022] Open
Abstract
Sertoli cells are somatic cells present in seminiferous tubules which have essential roles in regulating spermatogenesis. Considering that each Sertoli cell is able to support a limited number of germ cells, the final number of Sertoli cells reached during the proliferative period determines sperm production capacity. Only immature Sertoli cells, which have not established the blood-testis barrier, proliferate. A number of hormonal cues regulate Sertoli cell proliferation. Among them, FSH, the insulin family of growth factors, activin, and cytokines action must be highlighted. It has been demonstrated that cAMP/PKA, ERK1/2, PI3K/Akt, and mTORC1/p70SK6 pathways are the main signal transduction pathways involved in Sertoli cell proliferation. Additionally, c-Myc and hypoxia inducible factor are transcription factors which participate in the induction by FSH of various genes of relevance in cell cycle progression. Cessation of proliferation is a pre-requisite to Sertoli cell maturation accompanied by the establishment of the blood-testis barrier. With respect to this barrier, the participation of androgens, estrogens, thyroid hormones, retinoic acid and opioids has been reported. Additionally, two central enzymes that are involved in sensing cell energy status have been associated with the suppression of Sertoli cell proliferation, namely AMPK and Sirtuin 1 (SIRT1). Among the molecular mechanisms involved in the cessation of proliferation and in the maturation of Sertoli cells, it is worth mentioning the up-regulation of the cell cycle inhibitors p21Cip1, p27Kip, and p19INK4, and of the gap junction protein connexin 43. A decrease in Sertoli cell proliferation due to administration of certain therapeutic drugs and exposure to xenobiotic agents before puberty has been experimentally demonstrated. This review focuses on the hormones, locally produced factors, signal transduction pathways, and molecular mechanisms controlling Sertoli cell proliferation and maturation. The comprehension of how the final number of Sertoli cells in adulthood is established constitutes a pre-requisite to understand the underlying causes responsible for the progressive decrease in sperm production that has been observed during the last 50 years in humans.
Collapse
|
126
|
Rickert E, Fernandez MO, Choi I, Gorman M, Olefsky JM, Webster NJG. Neuronal SIRT1 Regulates Metabolic and Reproductive Function and the Response to Caloric Restriction. J Endocr Soc 2018; 3:427-445. [PMID: 30746504 PMCID: PMC6364627 DOI: 10.1210/js.2018-00318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/19/2018] [Indexed: 01/06/2023] Open
Abstract
Sirt1 is an NAD-dependent, class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. In this study, we generated mice expressing an enzymatically inactive form (N-MUT) or wild-type (WT) SIRT1 (N-OX) in mature neurons. N-OX male and female mice had impaired glucose tolerance, and N-MUT female, but not male, mice had improved glucose tolerance compared with that of WT littermates. Furthermore, glucose tolerance was improved in all mice with caloric restriction (CR) but was greater in the N-OX mice, who had better glucose tolerance than their littermates. At the reproductive level, N-OX females had impaired estrous cycles, with increased cycle length and more time in estrus. LH and progesterone surges were absent on the evening of proestrus in the N-OX mice, suggesting a defect in spontaneous ovulation, which was confirmed by the ovarian histology revealing fewer corpora lutea. Despite this defect, the mice were still fertile when mated to WT mice on the day of proestrus, indicating that the mice could respond to normal pheromonal or environmental cues. When subjected to CR, the N-OX mice went into diestrus arrest earlier than their littermates. Together, these results suggested that the overexpression of SIRT1 rendered the mice more sensitive to the metabolic improvements and suppression of reproductive cycles by CR, which was independent of circadian rhythms.
Collapse
Affiliation(s)
- Emily Rickert
- VA San Diego Healthcare System, San Diego, California.,Department of Medicine, University of California San Diego, La Jolla, California
| | | | - Irene Choi
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Michael Gorman
- Department of Psychology, University of California San Diego, La Jolla, California
| | - Jerrold M Olefsky
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Nicholas J G Webster
- VA San Diego Healthcare System, San Diego, California.,Department of Medicine, University of California San Diego, La Jolla, California.,Moores Cancer Center, University of California San Diego, La Jolla, California
| |
Collapse
|
127
|
Nguyen MT, Somogyvári M, Sőti C. Hsp90 Stabilizes SIRT1 Orthologs in Mammalian Cells and C. elegans. Int J Mol Sci 2018; 19:ijms19113661. [PMID: 30463299 PMCID: PMC6274930 DOI: 10.3390/ijms19113661] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 01/02/2023] Open
Abstract
Sirtuin 1 (SIRT1) othologs are ubiquitous NAD+-dependent deacetylases that act as nutrient sensors and modulate metabolism and stress responses in diverse organisms. Both mammalian SIRT1 and Caenorhabditis elegans SIR-2.1 have been implicated in dietary restriction, longevity, and healthspan. Hsp90 is an evolutionarily conserved molecular chaperone that stabilizes a plethora of signaling ’client’ proteins and regulates fundamental biological processes. Here we report that Hsp90 is required for conformational stabilization of SIRT1 and SIR-2.1. We find that inhibition of Hsp90 by geldanamycin (GA) induces the depletion of mammalian SIRT1 protein in a concentration and time dependent manner in COS-7 and HepG2 cells. In contrast to SIRT1, SIRT2 level remains unchanged by GA treatment, reflecting a specific Hsp90 SIRT1 interaction. Hsp90 inhibition leads to the destabilization and proteasomal degradation of SIRT1. Moreover, we observe a GA-sensitive physical interaction between SIRT1 and Hsp90 by immunoprecipitation. We also demonstrate that hsp-90 gene silencing also induces SIR-2.1 protein depletion and proteasomal degradation in C. elegans. Our findings identify metazoan SIRT1 orthologs as Hsp90 clients and reveal a novel crosstalk between the proteostasis and nutrient signaling networks, which may have implications in various age related diseases.
Collapse
Affiliation(s)
- Minh Tu Nguyen
- Department of Medical Chemistry, Semmelweis University, H-1094 Budapest, Hungary.
| | - Milán Somogyvári
- Department of Medical Chemistry, Semmelweis University, H-1094 Budapest, Hungary.
| | - Csaba Sőti
- Department of Medical Chemistry, Semmelweis University, H-1094 Budapest, Hungary.
| |
Collapse
|
128
|
Peng J, Zhou Y, Hong Z, Wu Y, Cai A, Xia M, Deng Z, Yang Y, Song T, Xiong J, Wei H, Peng J. Maternal eicosapentaenoic acid feeding promotes placental angiogenesis through a Sirtuin-1 independent inflammatory pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:147-157. [PMID: 30445165 DOI: 10.1016/j.bbalip.2018.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/21/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022]
Abstract
Maternal overnutrition or obesity is associated with a wide range of metabolic disorders and may impair placental angiogenesis. Previous studies have shown that n-3 polyunsaturated fatty acids (PUFA) promote fetal growth in both rodents and humans. Whether n-3 PUFA impacts on placental angiogenesis in vivo remains unclear. Sirtuin-1 (SIRT1) is a protein deacetylase that plays an important role in regulating inflammation and endothelial function. Little information is available on a putative role of SIRT1 in placental angiogenesis. The goal of this study was to examine the capability of eicosapentaenoic acid (EPA) to regulate angiogenesis and inflammation in SIRT1-deficient placentas. In the present study, male and female SIRT1+/- mice were mated overnight, then primiparous SIRT1+/- mice were fed a 60% kcal HFD or equienergy EPA diet (4.4% EPA-ethyl ester). We found that the EPA diet significantly improved maternal insulin sensitivity and decreased plasma levels of inflammatory factors IL-6 and TNFα concentration. Moreover, EPA treatment promoted fetus growth and placental angiogenesis, and inhibited the hypoxia inducible factor-1α(HIF1α) pathway. SIRT1 deficiency exhibited an opposite effect, leading to decrease in placental angiogenesis and fetal weight. No significant effect was observed between diet and genotype. Here, we reported for the first time that EPA treatment increased the expression of placental inflammatory genes and promoted translocation of NFκB into the nucleus. On the contrary, SIRT1-deficient placentas showed a decreased inflammation state. Together, these data demonstrate a previously unknown role of EPA to promote placental angiogenesis through a SIRT1 independent inflammatory pathway.
Collapse
Affiliation(s)
- Jie Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Zhang Hong
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yinghui Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Anle Cai
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Mao Xia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Zhao Deng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yang Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jia Xiong
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, PR China.
| |
Collapse
|
129
|
Fujita Y, Yamashita T. Sirtuins in Neuroendocrine Regulation and Neurological Diseases. Front Neurosci 2018; 12:778. [PMID: 30416425 PMCID: PMC6213750 DOI: 10.3389/fnins.2018.00778] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Silent information regulator 1 (SIRT1) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Sirtuin was originally studied as the lifespan-extending gene, silent information regulator 2 (SIRT2) in budding yeast. There are seven mammalian homologs of sirtuin (SIRT1–7), and SIRT1 is the closest homolog to SIRT2. SIRT1 modulates various key targets via deacetylation. In addition to histones, these targets include transcription factors, such as forkhead box O (FOXO), Ku70, p53, NF-κB, PPAR-gamma co-activator 1-alpha (PGC-1α), and peroxisome proliferator-activated receptor γ (PPARγ). SIRT1 has many biological functions, including aging, cell survival, differentiation, and metabolism. Genetic and physiological analyses in animal models have shown beneficial roles for SIRT1 in the brain during both development and adulthood. Evidence from in vivo and in vitro studies have revealed that SIRT1 regulates the cellular fate of neural progenitors, axon elongation, dendritic branching, synaptic plasticity, and endocrine function. In addition to its importance in physiological processes, SIRT1 has also been implicated in protection of neurons from degeneration in models of neurological diseases, such as traumatic brain injury and Alzheimer’s disease. In this review, we focus on the role of SIRT1 in the neuroendocrine system and neurodegenerative diseases. We also discuss the potential therapeutic implications of targeting the sirtuin pathway.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
130
|
Gorga A, Rindone GM, Regueira M, Pellizzari EH, Camberos MC, Cigorraga SB, Riera MF, Galardo MN, Meroni SB. Effect of resveratrol on Sertoli cell proliferation. J Cell Biochem 2018; 119:10131-10142. [DOI: 10.1002/jcb.27350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Affiliation(s)
- A Gorga
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - GM Rindone
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - M Regueira
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - EH Pellizzari
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MC Camberos
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - SB Cigorraga
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MF Riera
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - MN Galardo
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| | - SB Meroni
- Centro de Investigaciones Endocrinológicas, “Dr César Bergadá,” CONICET‐FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez Buenos Aires Argentina
| |
Collapse
|
131
|
Lin S, Xing H, Zang T, Ruan X, Wo L, He M. Sirtuins in mitochondrial stress: Indispensable helpers behind the scenes. Ageing Res Rev 2018; 44:22-32. [PMID: 29580919 DOI: 10.1016/j.arr.2018.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/16/2018] [Accepted: 03/22/2018] [Indexed: 12/26/2022]
Abstract
Mitochondria play an essential part in guaranteeing normal cellular physiological functions through providing ATP and participating in diverse processes and signaling pathways. Recently, more and more studies have revealed the vital roles of mitochondria in coping with stressors in the aging process, metabolic disturbances and neurological disorders. Mitochondrial stress responses, including the mitochondrial unfolded protein response (UPRmt), antioxidant defense, mitochondrial fission, mitochondrial fusion and mitophagy, are induced to maintain cellular integrity in response to stress. The sirtuin family, a group of NAD+-dependent deacetylases, has been the focus of much attention in recent years for their multiple regulatory functions, especially in aging and metabolism. Recent reports validated the significant link between mitochondrial stress responses and the sirtuin family, which may help to elucidate the pathogenesis and therapies for diseases such as Alzheimer's disease or Parkinson's disease. This review will summarize recent related studies and illuminate the interplay between sirtuins and mitochondrial stress.
Collapse
|
132
|
Assali DR, Hsu CT, Gunapala KM, Aguayo A, McBurney M, Steele AD. Food anticipatory activity on a calorie-restricted diet is independent of Sirt1. PLoS One 2018; 13:e0199586. [PMID: 29940007 PMCID: PMC6016907 DOI: 10.1371/journal.pone.0199586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 06/11/2018] [Indexed: 11/19/2022] Open
Abstract
A number of studies have demonstrated that the Sirtuin family member, Sirt1, is a key integrator of growth, metabolism, and lifespan. Sirt1 directly interacts with and deacetylates key regulators of the circadian clock, positioning it to be an important link between feeding and circadian rhythms. In fact, one study suggests that Sirt1 is necessary for behavioral anticipation of limited daily food availability, a circadian process termed food anticipatory activity (FAA). In their study, mice overexpressing Sirt1 had enhanced FAA, while mice lacking Sirt1 had little to no FAA. Based on the supposition that Sirt1 was indeed required for FAA, we sought to use Sirt1 deletion to map the neural circuitry responsible for FAA. We began by inactivating Sirt1 using the cell-type specific Cre-driver lines proopiomelanocortin, but after observing no effect on body weight loss or FAA we then moved on to more broadly neuronal Cre drivers Ca2+/calmodulin-dependent protein kinase II and nestin. As neither of these neuronal deletions of Sirt1 had impaired FAA, we then tested 1) a broad postnatal tamoxifen-inducible deletion, 2) a complete, developmental knockout of Sirt1, and 3) a gene replacement, catalytically inactive, form of Sirt1; but all of these mice had FAA similar to controls. Therefore, our findings suggest that FAA is completely independent of Sirt1.
Collapse
Affiliation(s)
- Dina R. Assali
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Cynthia T. Hsu
- Division of Biology, California Institute of Technology, Pasadena, CA, United States of America
| | - Keith M. Gunapala
- Division of Biology, California Institute of Technology, Pasadena, CA, United States of America
| | - Antonio Aguayo
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Michael McBurney
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Andrew D. Steele
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| |
Collapse
|
133
|
Gat-Yablonski G, De Luca F. Effect of Nutrition on Statural Growth
. Horm Res Paediatr 2018; 88:46-62. [PMID: 28365689 DOI: 10.1159/000456547] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
In children, proper growth and development are often regarded as a surrogate marker for good health. A complex system controls the initiation, rate, and cessation of growth, and thus gives a wonderful example of the interactions between genetics, epigenetics, and environmental factors (especially stress and nutrition). Malnutrition is considered a leading cause of growth attenuation in children. This review summarizes our current knowledge regarding the mechanisms linking nutrition and skeletal growth, including systemic factors, such as insulin, growth hormone, insulin-like growth factor-1, fibroblast growth factor-21, etc., and local mechanisms, including mTOR, miRNAs, and epigenetics. Studying the molecular mechanisms regulating skeletal growth may lead to the establishment of better nutritional and therapeutic regimens for more effective linear growth in children with malnutrition and growth abnormalities.
.
Collapse
Affiliation(s)
- Galia Gat-Yablonski
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Children's Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Francesco De Luca
- Section of Endocrinology and Diabetes, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
134
|
Pham J, Arul Nambi Rajan K, Li P, Parast MM. The role of Sirtuin1-PPARγ axis in placental development and function. J Mol Endocrinol 2018; 60:R201-R212. [PMID: 29467141 PMCID: PMC8584848 DOI: 10.1530/jme-17-0315] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
Abstract
Placental development is important for proper in utero growth and development of the fetus, as well as maternal well-being during pregnancy. Abnormal differentiation of placental epithelial cells, called trophoblast, is at the root of multiple pregnancy complications, including miscarriage, the maternal hypertensive disorder preeclampsia and intrauterine growth restriction. The ligand-activated nuclear receptor, PPARγ, and nutrient sensor, Sirtuin-1, both play a role in numerous pathways important to cell survival and differentiation, metabolism and inflammation. However, each has also been identified as a key player in trophoblast differentiation and placental development. This review details these studies, and also describes how various stressors, including hypoxia and inflammation, alter the expression or activity of PPARγ and Sirtuin-1, thereby contributing to placenta-based pregnancy complications.
Collapse
Affiliation(s)
- Jonathan Pham
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Kanaga Arul Nambi Rajan
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| | - Ping Li
- Department of PathologyMedical School of Jinan University, Guangzhou, China
| | - Mana M Parast
- Department of PathologyUniversity of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative MedicineUniversity of California San Diego, La Jolla, California, USA
| |
Collapse
|
135
|
Megakaryocyte lineage development is controlled by modulation of protein acetylation. PLoS One 2018; 13:e0196400. [PMID: 29698469 PMCID: PMC5919413 DOI: 10.1371/journal.pone.0196400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/12/2018] [Indexed: 12/11/2022] Open
Abstract
Treatment with lysine deacetylase inhibitors (KDACi) for haematological malignancies, is accompanied by haematological side effects including thrombocytopenia, suggesting that modulation of protein acetylation affects normal myeloid development, and specifically megakaryocyte development. In the current study, utilising ex-vivo differentiation of human CD34+ haematopoietic progenitor cells, we investigated the effects of two functionally distinct KDACi, valproic acid (VPA), and nicotinamide (NAM), on megakaryocyte differentiation, and lineage choice decisions. Treatment with VPA increased the number of megakaryocyte/erythroid progenitors (MEP), accompanied by inhibition of megakaryocyte differentiation, whereas treatment with NAM accelerated megakaryocyte development, and stimulated polyploidisation. Treatment with both KDACi resulted in no significant effects on erythrocyte differentiation, suggesting that the effects of KDACi primarily affect megakaryocyte lineage development. H3K27Ac ChIP-sequencing analysis revealed that genes involved in myeloid development, as well as megakaryocyte/erythroid (ME)-lineage differentiation are uniquely modulated by specific KDACi treatment. Taken together, our data reveal distinct effects of specific KDACi on megakaryocyte development, and ME-lineage decisions, which can be partially explained by direct effects on promoter acetylation of genes involved in myeloid differentiation.
Collapse
|
136
|
Zhou M, Luo J, Zhang H. Role of Sirtuin 1 in the pathogenesis of ocular disease (Review). Int J Mol Med 2018; 42:13-20. [PMID: 29693113 DOI: 10.3892/ijmm.2018.3623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/29/2018] [Indexed: 11/06/2022] Open
Abstract
Sirtuin (SIRT)1, a member of the SIRT family, is a highly conserved NAD+‑dependent histone deacetylase, which has a regulatory role in numerous physiological and pathological processes by removing acetyl groups from various proteins. SIRT1 controls the activity of numerous transcription factors and cofactors, which impacts the downstream gene expression, and eventually alleviates oxidative stress and associated damage. Numerous studies have revealed that dysfunction of SIRT1 is linked with ocular diseases, including cataract, age‑associated macular degeneration, diabetic retinopathy and glaucoma, while ectopic upregulation of SIRT1 protects against various ocular diseases. In the present review, the significant role of SIRT1 and the potential therapeutic value of modulating SIRT1 expression in ocular development and eye diseases is summarized.
Collapse
Affiliation(s)
- Mengwen Zhou
- Department of Ophthalmology, Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jing Luo
- Department of Ophthalmology, Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Huiming Zhang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
137
|
Kang X, Yang W, Wang R, Xie T, Li H, Feng D, Jin X, Sun H, Wu S. Sirtuin-1 (SIRT1) stimulates growth-plate chondrogenesis by attenuating the PERK-eIF-2α-CHOP pathway in the unfolded protein response. J Biol Chem 2018; 293:8614-8625. [PMID: 29653943 DOI: 10.1074/jbc.m117.809822] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/21/2018] [Indexed: 12/20/2022] Open
Abstract
The NAD+-dependent deacetylase sirtuin-1 (SIRT1) has emerged as an important regulator of chondrogenesis and cartilage homeostasis, processes that are important for physiological skeletal growth and that are dysregulated in osteoarthritis. However, the functional role and underlying mechanism by which SIRT1 regulates chondrogenesis remain unclear. Using cultured rat metatarsal bones and chondrocytes isolated from rat metatarsal rudiments, here we studied the effects of the SIRT1 inhibitor EX527 or of SIRT1 siRNA on chondrocyte proliferation, hypertrophy, and apoptosis. We show that EX527 or SIRT1 siRNA inhibits chondrocyte proliferation and hypertrophy and induces apoptosis. We also observed that SIRT1 inhibition mainly induces the PERK-eIF-2α-CHOP axis of the endoplasmic reticulum (ER) stress response in growth-plate chondrocytes. Of note, EX527- or SIRT1 siRNA-mediated inhibition of metatarsal growth and growth-plate chondrogenesis were partly neutralized by phenylbutyric acid, a chemical chaperone that attenuates ER stress. Moreover, EX527-mediated impairment of chondrocyte function (i.e. of chondrocyte proliferation, hypertrophy, and apoptosis) was partly reversed in CHOP-/- cells. We also present evidence that SIRT1 physically interacts with and deacetylates PERK. Collectively, our findings indicate that SIRT1 deacetylates PERK and attenuates the PERK-eIF-2α-CHOP axis of the unfolded protein response pathway and thereby promotes growth-plate chondrogenesis and longitudinal bone growth.
Collapse
Affiliation(s)
- Xiaomin Kang
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Wei Yang
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Ruiqi Wang
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Tianping Xie
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Huixia Li
- the Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China, and
| | - Dongxu Feng
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China.,the Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710054, China
| | - Xinxin Jin
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Hongzhi Sun
- the Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China, and
| | - Shufang Wu
- From the Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China,
| |
Collapse
|
138
|
Yanagisawa S, Baker JR, Vuppusetty C, Koga T, Colley T, Fenwick P, Donnelly LE, Barnes PJ, Ito K. The dynamic shuttling of SIRT1 between cytoplasm and nuclei in bronchial epithelial cells by single and repeated cigarette smoke exposure. PLoS One 2018; 13:e0193921. [PMID: 29509781 PMCID: PMC5839577 DOI: 10.1371/journal.pone.0193921] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 02/21/2018] [Indexed: 12/18/2022] Open
Abstract
SIRT1 (silent information regulator 2 homolog 1) is a crucial cellular survival protein especially in oxidative stress environments, and has been thought to locate within the nuclei, but also known to shuttle between cytoplasm and nuclei in some cell types. Here, we show for the first time the dynamics of SIRT1 in the presence of single or concurrent cigarette smoke extract (CSE) exposure in human bronchial epithelial cells (HBEC). In BEAS-2B HBEC or primary HBEC, SIRT1 was localized predominantly in cytoplasm, and the CSE (3%) induced nuclear translocation of SIRT1 from cytoplasm in the presence of L-buthionine sulfoximine (an irreversible inhibitor of γ-glutamylcystein synthetase), mainly through the activation of phosphatidylinositol 3-kinase (PI3K) α subunit. This SIRT1 nuclear shuttling was associated with FOXO3a nuclear translocation and the strong induction of several anti-oxidant genes including superoxide dismutase (SOD) 2 and 3; therefore seemed to be an adaptive response. When BEAS-2B cells were pretreated with repeated exposure to a lower concentration of CSE (0.3%), the CSE-induced SIRT1 shuttling and resultant SOD2/3 mRNA induction were significantly impaired. Thus, this result offers a useful cell model to mimic the impaired anti-oxidant capacity in cigarette smoking-associated lung disease such as chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Satoru Yanagisawa
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jonathan R. Baker
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Chaitanya Vuppusetty
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Takeshi Koga
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas Colley
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter Fenwick
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Louise E. Donnelly
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J. Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kazuhiro Ito
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
139
|
Premature aging in behavior and immune functions in tyrosine hydroxylase haploinsufficient female mice. A longitudinal study. Brain Behav Immun 2018; 69:440-455. [PMID: 29341892 DOI: 10.1016/j.bbi.2018.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 01/03/2023] Open
Abstract
Aging is accompanied by impairment in the nervous, immune, and endocrine systems as well as in neuroimmunoendocrine communication. In this context, there is an age-related alteration of the physiological response to acute stress, which is modulated by catecholamine (CA), final products of the sympathetic-adreno-medullary axis. The involvement of CA in essential functions of the nervous system is consistent with the neuropsychological deficits found in mice with haploinsufficiency (hemizygous; HZ) of tyrosine hydroxylase (TH) enzyme (TH-HZ). However, other possible alterations in regulatory systems have not been studied in these animals. The aim of the present work was to analyze whether adult TH-HZ female mice presented the impairment of behavioral traits and immunological responses that occurs with aging and whether they had affected their mean lifespan. ICR-CD1 female TH-HZ and wild type (WT) mice were used in a longitudinal study. Behavioral tests were performed on adult and old mice in order to evaluate their sensorimotor abilities and exploratory capacity, as well as anxiety-like behaviors. At the ages of 2 ± 1, 4 ± 1, 9 ± 1, 13 ± 1 and 20 ± 1 months, peritoneal leukocytes were extracted and several immune functions were assessed (phagocytic capacity, Natural Killer (NK) cytotoxicity, and lymphoproliferative response to lipopolysaccharide (LPS) and concanavalin A (ConA)). In addition, several oxidative stress parameters (catalase, glutathione reductase and glutathione peroxidase activities, and reduced glutathione (GSH) concentrations as antioxidant compounds as well as xanthine oxidase activity, oxidized glutathione (GSSG) concentrations, and GSSG/GSH ratio as oxidants) were analyzed. As inflammatory stress parameters TNF-alpha and IL-10 concentrations, and TNF-alpha/IL-10 ratios as inflammatory/anti-inflammatory markers, were measured. Animals were maintained in standard conditions until their natural death. The results indicate that adult TH-HZ mice presented worse sensorimotor abilities and exploratory capacity than their WT littermates as well as greater anxiety-like behaviors. With regards to the immune system, adult TH-HZ animals exhibited lower values of phagocytic capacity, NK cytotoxicity, and lymphoproliferative response to LPS and ConA than WT mice. Moreover, immune cells of TH-HZ mice showed higher oxidative and inflammatory stress than those of WT animals. Although these differences between TH-HZ and WT, in general, decreased with aging, this premature immunosenescence and impairment of behavior of TH-HZ mice was accompanied by a shorter mean lifespan in comparison to WT counterparts. In conclusion, haploinsufficiency of th gene in female mice appears to provoke premature aging of the regulatory systems affecting mean lifespan.
Collapse
|
140
|
Zhou Y, Song T, Peng J, Zhou Z, Wei H, Zhou R, Jiang S, Peng J. SIRT1 suppresses adipogenesis by activating Wnt/β-catenin signaling in vivo and in vitro. Oncotarget 2018; 7:77707-77720. [PMID: 27776347 PMCID: PMC5363615 DOI: 10.18632/oncotarget.12774] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/12/2016] [Indexed: 02/07/2023] Open
Abstract
Sirtuin 1 (SIRT1) regulates adipocyte and osteoblast differentiation. However, the underlying mechanism should be investigated. This study revealed that SIRT1 acts as a crucial repressor of adipogenesis. RNA-interference-mediated SIRT1 knockdown or genetic ablation enhances adipogenic potential, whereas SIRT1 overexpression inhibits adipogenesis in mesenchymal stem cells (MSCs). SIRT1 also deacetylates the histones of sFRP1, sFRP2, and Dact1 promoters; inhibits the mRNA expression of sFRP1, sFRP2, and Dact1; activates Wnt signaling pathways; and suppresses adipogenesis. SIRT1 deacetylates β-catenin to promote its accumulation in the nucleus and thus induces the transcription of genes that block MSC adipogenesis. In mice, the partial absence of SIRT1 promotes the formation of white adipose tissues without affecting the development of the body of mice. Our study described the regulatory role of SIRT1 in Wnt signaling and proposed a regulatory mechanism of adipogenesis.
Collapse
Affiliation(s)
- Yuanfei Zhou
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Disease, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China.,Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Jie Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Zheng Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Disease, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, P. R. China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Siwen Jiang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, P. R. China
| |
Collapse
|
141
|
Wang H, Dong X, Liu Z, Zhu S, Liu H, Fan W, Hu Y, Hu T, Yu Y, Li Y, Liu T, Xie C, Gao Q, Li G, Zhang J, Ding Z, Sun J. Resveratrol Suppresses Rotenone-induced Neurotoxicity Through Activation of SIRT1/Akt1 Signaling Pathway. Anat Rec (Hoboken) 2018; 301:1115-1125. [PMID: 29350822 DOI: 10.1002/ar.23781] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/20/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Hui Wang
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Xiaoguang Dong
- Department of Orthopedic; Osteological Hospital of Yishengjian; Qingdao Shandong 266100 China
| | - Zengxun Liu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Shaowei Zhu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Haili Liu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Wenchuang Fan
- Department of Traumatic Orthopaedics; Yantaishan Hospital; Yantai Shandong 264025 China
| | - Yanlai Hu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Tao Hu
- Department of Orthopedic; Osteological Hospital of Yishengjian; Qingdao Shandong 266100 China
| | - Yonghui Yu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Yizhao Li
- Department of Rehabilitation; Laiwu Rehabilitation Hospital; Laiwu Shandong 271100 China
| | - Tianwei Liu
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Chengjia Xie
- Department of Periodontics; Stomatological Hospital of Shandong University; Shandong 250012 China
| | - Qing Gao
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Guibao Li
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Jing Zhang
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Zhaoxi Ding
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Sciences; Shandong University; Jinan Shandong 250012 China
| |
Collapse
|
142
|
Tatone C, Di Emidio G, Barbonetti A, Carta G, Luciano AM, Falone S, Amicarelli F. Sirtuins in gamete biology and reproductive physiology: emerging roles and therapeutic potential in female and male infertility. Hum Reprod Update 2018; 24:267-289. [DOI: 10.1093/humupd/dmy003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | - Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | | | - Gaspare Carta
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Gynecology Unit, Reproductive Service, San Salvatore Hospital, Via Vetoio, 67100 L’Aquila, Italy
| | - Alberto M Luciano
- Department of Health, Animal Science and Food Safety, Reproductive and Developmental Biology Laboratory, University of Milan, 20133 Milan, Italy
| | - Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
- Institute of Translational Pharmacology (IFT), CNR, 67100 L’Aquila, Italy
| |
Collapse
|
143
|
Yang XW, He WB, Gong F, Li W, Li XR, Zhong CG, Lu GX, Lin G, Du J, Tan YQ. Novel FOXL2 mutations cause blepharophimosis-ptosis-epicanthus inversus syndrome with premature ovarian insufficiency. Mol Genet Genomic Med 2018; 6:261-267. [PMID: 29378385 PMCID: PMC5902393 DOI: 10.1002/mgg3.366] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/29/2017] [Accepted: 12/27/2017] [Indexed: 01/02/2023] Open
Abstract
Background Blepharophimosis‐ptosis‐epicanthus inversus syndrome (BPES) is a malformation of the eyelids. Forkhead Box L2 (FOXL2) is the only gene known to be associated with BPES. Methods We identified two Han Chinese BPES families with premature ovarian insufficiency (POI). Sanger sequencing and in vitro functional analysis were performed to identify the genetic cause. Results Sanger sequencing identified two novel mutations (c.462_468del, c.988_989insG) in FOXL2, one in each family. The in vitro functional analysis confirmed that both novel mutations were associated with impaired transactivation of downstream genes. Specifically, the single‐base insertion, c.988_989insG, led to subcellular mislocalization and aggregation of the encoded protein, which validated the hypothesis that the two novel FOXL2 mutations are deleterious and associated with POI in the two BPES families. Conclusion The novel mutations identified in the present study will enhance the present knowledge of the mutation spectrum of FOXL2. The in vitro experiments provide further insights into the molecular mechanism by which the two new variants mediate disease pathogenesis and may contribute to elucidating the genotype‐phenotype correlation between the two novel FOXL2 mutations and POI.
Collapse
Affiliation(s)
- Xiao-Wen Yang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China
| | - Wen-Bin He
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Wen Li
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Xiu-Rong Li
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Chang-Gao Zhong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Guang-Xiu Lu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, Hunan, China
| |
Collapse
|
144
|
Shin AN, Han L, Dasgupta C, Huang L, Yang S, Zhang L. SIRT1 increases cardiomyocyte binucleation in the heart development. Oncotarget 2018; 9:7996-8010. [PMID: 29487709 PMCID: PMC5814276 DOI: 10.18632/oncotarget.23847] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/05/2017] [Indexed: 12/17/2022] Open
Abstract
SIRT1 regulates cell senescence. We investigated a novel role of SIRT1 in the regulation of cardiomyocyte terminal differentiation in the developing heart. Retinoic acid (RA)-induced binucleation of H9c2 cells was associated with increased SIRT1 expression. Inhibition of SIRT1 activity or expression significantly decreased RA-induced binucleation. SIRT1 expression was minimal in the fetal heart and significantly upregulated in the hearts of postnatal day 7 (P7) rat pups. In contrast, heart-specific miR-133a expression was high in the fetal heart but significantly reduced in P7 pup hearts. The miR-133a promoter contains a canonical HRE element and hypoxia upregulated miR-133a gene expression in the heart. SIRT1 mRNA 3′UTR has miR-133a binding sequences and miR-133a and hypoxia suppressed SIRT1 expression in cardiomyocytes. Of importance, inhibition of SIRT1 significantly reduced binucleated cardiomyocytes in the hearts of P7 pups. Taken together, the present study reveals a novel role of SIRT1 and its regulation by miR-133a in cardiomyocyte terminal differentiation of the developing heart, and suggests a potential therapeutic strategy that may impact cardiac function later in life.
Collapse
Affiliation(s)
- Alexandra N Shin
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA.,Department of Biological Sciences, California Baptist University, Riverside, California, USA
| | - Limin Han
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Chiranjib Dasgupta
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lei Huang
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Shumei Yang
- Department of Chemistry and Biochemistry, California State University, San Bernardino, California, USA
| | - Lubo Zhang
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
145
|
Sirtuin1 is required for proper trophoblast differentiation and placental development in mice. Placenta 2017; 62:1-8. [PMID: 29405961 DOI: 10.1016/j.placenta.2017.12.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/21/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Placental insufficiency, arising from abnormal trophoblast differentiation and function, is a major cause of fetal growth restriction. Sirtuin-1 (Sirt1) is a ubiquitously-expressed NAD-dependent protein deacetylase which plays a key role in numerous cellular processes, including cellular differentiation and metabolism. Though Sirt1 has been widely studied, its role in placentation and trophoblast differentiation is unclear. METHOD Sirt1-heterozygous mice were mated and evaluated at various points during embryogenesis. In situ hybridization and immunohistochemistry were used to further characterize the placental phenotype of Sirt1-null mice. Wild-type (WT) and Sirt1-null mouse trophoblast stem cell (TSC) lines were derived from e3.5 littermate blastocysts. These cells were then evaluated at various points following differentiation. Differentiation was evaluated by expression of lineage specific markers using qPCR and flow cytometry, as well as Matrigel invasion assays. Global gene expression changes were evaluated using microarray-based RNA profiling; changes in specific pathways were validated using qPCR and western blot. RESULTS In the absence of Sirt1, both embryos and placentas were small, with placentas showing abnormalities in both the labyrinthine layer and junctional zone. Sirt1-null TSCs exhibited an altered phenotype in both undifferentiated and differentiated states, phenotypes which corresponded to changes in pathways relevant to both TSC maintenance and differentiation. Specifically, Sirt1-null TSC showed blunted differentiation, and appeared to be suspended in an Epcamhigh trophoblast progenitor state. DISCUSSION Our results suggest that Sirt1 is required for proper TSC differentiation and placental development.
Collapse
|
146
|
Williams EO, Taylor AK, Bell EL, Lim R, Kim DM, Guarente L. Sirtuin 1 Promotes Deacetylation of Oct4 and Maintenance of Naive Pluripotency. Cell Rep 2017; 17:809-820. [PMID: 27732856 DOI: 10.1016/j.celrep.2016.09.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/14/2016] [Accepted: 09/15/2016] [Indexed: 01/07/2023] Open
Abstract
The enhancer landscape is dramatically restructured as naive preimplantation epiblasts transition to the post-implantation state of primed pluripotency. A key factor in this process is Otx2, which is upregulated during the early stages of this transition and ultimately recruits Oct4 to a different set of enhancers. In this study, we discover that the acetylation status of Oct4 regulates the induction of the primed pluripotency gene network. Maintenance of the naive state requires the NAD-dependent deacetylase, SirT1, which deacetylates Oct4. The activity of SirT1 is reduced during the naive-to-primed transition; Oct4 becomes hyper-acetylated and binds to an Otx2 enhancer to induce Otx2 expression. Induction of Otx2 causes the reorganization of acetylated Oct4 and results in the induction of the primed pluripotency gene network. Regulation of Oct4 by SirT1 may link stem cell development to environmental conditions, and it may provide strategies to manipulate epiblast cell state.
Collapse
Affiliation(s)
- Eric O Williams
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amy K Taylor
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric L Bell
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rachelle Lim
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel M Kim
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leonard Guarente
- Glenn Laboratory for the Science of Aging and the Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
147
|
Regulation of Sirtuin-Mediated Protein Deacetylation by Cardioprotective Phytochemicals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1750306. [PMID: 29234485 PMCID: PMC5695026 DOI: 10.1155/2017/1750306] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/26/2017] [Accepted: 09/25/2017] [Indexed: 01/06/2023]
Abstract
Modulation of posttranslational modifications (PTMs), such as protein acetylation, is considered a novel therapeutic strategy to combat the development and progression of cardiovascular diseases. Protein hyperacetylation is associated with the development of numerous cardiovascular diseases, including atherosclerosis, hypertension, cardiac hypertrophy, and heart failure. In addition, decreased expression and activity of the deacetylases Sirt1, Sirt3, and Sirt6 have been linked to the development and progression of cardiac dysfunction. Several phytochemicals exert cardioprotective effects by regulating protein acetylation levels. These effects are mainly exerted via activation of Sirt1 and Sirt3 and inhibition of acetyltransferases. Numerous studies support a cardioprotective role for sirtuin activators (e.g., resveratrol), as well as other emerging modulators of protein acetylation, including curcumin, honokiol, oroxilyn A, quercetin, epigallocatechin-3-gallate, bakuchiol, tyrosol, and berberine. Studies also point to a cardioprotective role for various nonaromatic molecules, such as docosahexaenoic acid, alpha-lipoic acid, sulforaphane, and caffeic acid ethanolamide. Here, we review the vast evidence from the bench to the clinical setting for the potential cardioprotective roles of various phytochemicals in the modulation of sirtuin-mediated deacetylation.
Collapse
|
148
|
Tang S, Fang Y, Huang G, Xu X, Padilla-Banks E, Fan W, Xu Q, Sanderson SM, Foley JF, Dowdy S, McBurney MW, Fargo DC, Williams CJ, Locasale JW, Guan Z, Li X. Methionine metabolism is essential for SIRT1-regulated mouse embryonic stem cell maintenance and embryonic development. EMBO J 2017; 36:3175-3193. [PMID: 29021282 DOI: 10.15252/embj.201796708] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 12/19/2022] Open
Abstract
Methionine metabolism is critical for epigenetic maintenance, redox homeostasis, and animal development. However, the regulation of methionine metabolism remains unclear. Here, we provide evidence that SIRT1, the most conserved mammalian NAD+-dependent protein deacetylase, is critically involved in modulating methionine metabolism, thereby impacting maintenance of mouse embryonic stem cells (mESCs) and subsequent embryogenesis. We demonstrate that SIRT1-deficient mESCs are hypersensitive to methionine restriction/depletion-induced differentiation and apoptosis, primarily due to a reduced conversion of methionine to S-adenosylmethionine. This reduction markedly decreases methylation levels of histones, resulting in dramatic alterations in gene expression profiles. Mechanistically, we discover that the enzyme converting methionine to S-adenosylmethionine in mESCs, methionine adenosyltransferase 2a (MAT2a), is under control of Myc and SIRT1. Consistently, SIRT1 KO embryos display reduced Mat2a expression and histone methylation and are sensitive to maternal methionine restriction-induced lethality, whereas maternal methionine supplementation increases the survival of SIRT1 KO newborn mice. Our findings uncover a novel regulatory mechanism for methionine metabolism and highlight the importance of methionine metabolism in SIRT1-mediated mESC maintenance and embryonic development.
Collapse
Affiliation(s)
- Shuang Tang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Yi Fang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Gang Huang
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xiaojiang Xu
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Elizabeth Padilla-Banks
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Wei Fan
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Qing Xu
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sydney M Sanderson
- Department of Pharmacology and Cancer Biology, Duke Cancer Institute, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Julie F Foley
- Cellular and Molecular Pathology Branch and Comparative Medicine Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Scotty Dowdy
- Cellular and Molecular Pathology Branch and Comparative Medicine Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Michael W McBurney
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - David C Fargo
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke Cancer Institute, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
149
|
Deschênes M, Chabot B. The emerging role of alternative splicing in senescence and aging. Aging Cell 2017; 16:918-933. [PMID: 28703423 PMCID: PMC5595669 DOI: 10.1111/acel.12646] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Deregulation of precursor mRNA splicing is associated with many illnesses and has been linked to age-related chronic diseases. Here we review recent progress documenting how defects in the machinery that performs intron removal and controls splice site selection contribute to cellular senescence and organismal aging. We discuss the functional association linking p53, IGF-1, SIRT1, and ING-1 splice variants with senescence and aging, and review a selection of splicing defects occurring in accelerated aging (progeria), vascular aging, and Alzheimer's disease. Overall, it is becoming increasingly clear that changes in the activity of splicing factors and in the production of key splice variants can impact cellular senescence and the aging phenotype.
Collapse
Affiliation(s)
- Mathieu Deschênes
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| |
Collapse
|
150
|
Abstract
Objective Lung cancer, which is the leading cause of cancer death worldwide, is influenced by a wide variety of environmental and genetic risk factors. The silent information regulator 1 (SIRT1) gene is located on the long arm of chromosome 10 (10q21.3) and has been shown to play crucial roles in lung cancer development in previous studies. In this study, we determined whether variation in the SIRT1 gene is associated with lung cancer in Chinese population. Methods The case–control study comprised 246 controls and 257 non-small cell lung cancer patients, comprising 79 squamous cell carcinoma patients and 124 adenocarcinoma patients. All subjects were from Zhejiang, China. Four single-nucleotide polymorphisms of SIRT1 gene were analyzed: rs12778366 (C/T, lies in the 5′ upstream), rs3758391 (C/T, lies in the 5′ upstream), rs2273773 (C/T, lies in the coding) and rs4746720 (C/T, lies in the 3′ untranslated region). Results No significant difference of allele and genotype frequencies was observed between the different groups. Haplotype association analysis carried out on the four single-nucleotide polymorphisms within the case–control cohort also did not reveal a significant association with lung cancer (P>0.05). Conclusion The results suggest the tested SIRT1 gene polymorphisms may not contribute to lung cancer. Further studies are warranted to demonstrate the functional roles of the SIRT1 polymorphism in lung cancer.
Collapse
Affiliation(s)
- Yanbo Lv
- Department of Pathology, Zhejiang Hospital, Hangzhou City, Zhejiang, China
| | - Shuangyan Lin
- Department of Pathology, Zhejiang Hospital, Hangzhou City, Zhejiang, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, Hangzhou City, Zhejiang, China
| |
Collapse
|