51
|
Naiman S, Huynh FK, Gil R, Glick Y, Shahar Y, Touitou N, Nahum L, Avivi MY, Roichman A, Kanfi Y, Gertler AA, Doniger T, Ilkayeva OR, Abramovich I, Yaron O, Lerrer B, Gottlieb E, Harris RA, Gerber D, Hirschey MD, Cohen HY. SIRT6 Promotes Hepatic Beta-Oxidation via Activation of PPARα. Cell Rep 2019; 29:4127-4143.e8. [PMID: 31851938 PMCID: PMC7165364 DOI: 10.1016/j.celrep.2019.11.067] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/11/2019] [Accepted: 11/15/2019] [Indexed: 12/27/2022] Open
Abstract
The pro-longevity enzyme SIRT6 regulates various metabolic pathways. Gene expression analyses in SIRT6 heterozygotic mice identify significant decreases in PPARα signaling, known to regulate multiple metabolic pathways. SIRT6 binds PPARα and its response element within promoter regions and activates gene transcription. Sirt6+/- results in significantly reduced PPARα-induced β-oxidation and its metabolites and reduced alanine and lactate levels, while inducing pyruvate oxidation. Reciprocally, starved SIRT6 transgenic mice show increased pyruvate, acetylcarnitine, and glycerol levels and significantly induce β-oxidation genes in a PPARα-dependent manner. Furthermore, SIRT6 mediates PPARα inhibition of SREBP-dependent cholesterol and triglyceride synthesis. Mechanistically, SIRT6 binds PPARα coactivator NCOA2 and decreases liver NCOA2 K780 acetylation, which stimulates its activation of PPARα in a SIRT6-dependent manner. These coordinated SIRT6 activities lead to regulation of whole-body respiratory exchange ratio and liver fat content, revealing the interactions whereby SIRT6 synchronizes various metabolic pathways, and suggest a mechanism by which SIRT6 maintains healthy liver.
Collapse
Affiliation(s)
- Shoshana Naiman
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Frank K Huynh
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA; Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Reuven Gil
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Yair Glick
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Yael Shahar
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Noga Touitou
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Liat Nahum
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Matan Y Avivi
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Asael Roichman
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Yariv Kanfi
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Asaf A Gertler
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Tirza Doniger
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Ifat Abramovich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron Street, Bat Galim, Haifa, Israel
| | - Orly Yaron
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Batia Lerrer
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron Street, Bat Galim, Haifa, Israel
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Doron Gerber
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel; Bar Ilan Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Matthew D Hirschey
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Haim Y Cohen
- Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
52
|
Gilani N, Haghshenas R, Esmaeili M. Application of multivariate longitudinal models in SIRT6, FBS, and BMI analysis of the elderly. Aging Male 2019; 22:260-265. [PMID: 29901417 DOI: 10.1080/13685538.2018.1477933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Objective: SIRT6 is a main regulator of metabolism and lifespan and its importance has been implicated in the prevention against aging-related diseases. The objective of this study was to examine the application of multivariate longitudinal models in SIRT6, FBS, and BMI analysis in the elderly men after eight weeks concurrent training with supplementation of l-arginine (l-Arg). Methods: Thirty two elderly men with mean age of 63.09 ± 3.71 years were randomly divided into four equal-sized groups (each n = 8); Exercise + supplement (ES) group; exercise + placebo (EP) group; supplement (S) group and control (C) group. The ES and EP groups performed the eight weeks of concurrent training, three sessions per week. Group ES and group S consumed 1000 mg of l-Arg per day at 8:00 pm. Measurements of biochemical variables were done by ELISA Reader method. For analytical purposes, we used the paired sample t-test and multivariate longitudinal modeling with generalized estimating equation (GEE) methodology. All analyses have been implemented in R-3.4.1. p Values less than .05 were considered statistically significant. Results: With respect to significant association between sirt6, FBS, and BMI, this study showed that synergy effect of training and supplementation was greater than the sum of their individual effects on SIRT6 (β = 0.79, p < .001), FBS (β = -5.56, p = .022), and BMI (β = -3.89; p = .041). Also exercise alone had a significantly larger effect than supplementation alone on responses. Conclusions: It can be concluded that the joint usage of concurrent training and supplement of l-Arg for elderly men could improve the metabolism and body composition.
Collapse
Affiliation(s)
- Neda Gilani
- Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Rouhollah Haghshenas
- Department of Sport Sciences, Faculty of Humanities, Semnan University , Semnan , Iran
| | - Mahmoud Esmaeili
- Department of Sport Sciences, Faculty of Humanities, Semnan University , Semnan , Iran
| |
Collapse
|
53
|
Zheng S, Wohlfahrt J, Cohen I, Cen Y. Methods for studying human sirtuins with activity-based chemical probes. Methods Enzymol 2019; 633:251-269. [PMID: 32046849 DOI: 10.1016/bs.mie.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sirtuins are unique posttranslational modification enzymes that utilize NAD+ as the co-substrate to remove acyl groups from lysine residues. The deacylation events result in profound biological consequences, from transcription silencing to metabolism regulation. This article focuses on a newly developed technology using activity-based chemical probes to report sirtuin functional state in various settings. These chemical probes, thioacyllysine peptides carrying photo-cross-linker as well as bioorthogonal functionality, target the active site of sirtuins to form stalled reaction intermediate. Subsequently, the probe forms covalent adduct with the protein through photocrosslinking. Ultimately, the active sirtuin can be visualized via "click" chemistry-mediated conjugation to a fluorescent tag. Here, we describe the labeling protocols on recombinant protein, whole cell lysate, and in situ labeling.
Collapse
Affiliation(s)
- Song Zheng
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT, United States
| | - Jessica Wohlfahrt
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT, United States
| | - Ian Cohen
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT, United States
| | - Yana Cen
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT, United States.
| |
Collapse
|
54
|
Kim HG, Huang M, Xin Y, Zhang Y, Zhang X, Wang G, Liu S, Wan J, Ahmadi AR, Sun Z, Liangpunsakul S, Xiong X, Dong XC. The epigenetic regulator SIRT6 protects the liver from alcohol-induced tissue injury by reducing oxidative stress in mice. J Hepatol 2019; 71:960-969. [PMID: 31295533 PMCID: PMC6801027 DOI: 10.1016/j.jhep.2019.06.019] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/29/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS As a nicotinamide adenine dinucleotide-dependent deacetylase and a key epigenetic regulator, sirtuin 6 (SIRT6) has been implicated in the regulation of metabolism, DNA repair, and inflammation. However, the role of SIRT6 in alcohol-related liver disease (ALD) remains unclear. The aim of this study was to investigate the function and mechanism of SIRT6 in ALD pathogenesis. METHODS We developed and characterized Sirt6 knockout (KO) and transgenic mouse models that were treated with either control or ethanol diet. Hepatic steatosis, inflammation, and oxidative stress were analyzed using biochemical and histological methods. Gene regulation was analyzed by luciferase reporter and chromatin immunoprecipitation assays. RESULTS The Sirt6 KO mice developed severe liver injury characterized by a remarkable increase of oxidative stress and inflammation, whereas the Sirt6 transgenic mice were protected from ALD via normalization of hepatic lipids, inflammatory response, and oxidative stress. Our molecular analysis has identified a number of novel Sirt6-regulated genes that are involved in antioxidative stress, including metallothionein 1 and 2 (Mt1 and Mt2). Mt1/2 genes were downregulated in the livers of Sirt6 KO mice and patients with alcoholic hepatitis. Overexpression of Mt1 in the liver of Sirt6 KO mice improved ALD by reducing hepatic oxidative stress and inflammation. We also identified a critical link between SIRT6 and metal regulatory transcription factor 1 (Mtf1) via a physical interaction and functional coactivation. Mt1/2 promoter reporter assays showed a strong synergistic effect of SIRT6 on the transcriptional activity of Mtf1. CONCLUSIONS Our data suggest that SIRT6 plays a critical protective role against ALD and it may serve as a potential therapeutic target for ALD. LAY SUMMARY The liver, the primary organ for ethanol metabolism, can be damaged by the byproducts of ethanol metabolism, including reactive oxygen species. In this study, we have identified a key epigenetic regulator SIRT6 that plays a critical role in protecting the liver from oxidative stress-induced liver injury. Thus, our data suggest that SIRT6 may be a potential therapeutic target for alcohol-related liver disease.
Collapse
Affiliation(s)
- Hyeong Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.,Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Yue Xin
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, 453003, P.R. China,Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang Medical University, Xinxiang, Henan, 453003, P.R. China
| | - Yang Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Xinge Zhang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, 453003, P.R. China,Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang Medical University, Xinxiang, Henan, 453003, P.R. China
| | - Gaihong Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.,Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Ali Reza Ahmadi
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana 46202, USA
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| | - Xiaocheng Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
55
|
Beneficial effects of running and milk protein supplements on Sirtuins and risk factors of metabolic disorders in rats with low aerobic capacity. Metabol Open 2019; 4:100019. [PMID: 32812928 PMCID: PMC7424841 DOI: 10.1016/j.metop.2019.100019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022] Open
Abstract
Background Physical activity and dietary intake of dairy products are associated with improved metabolic health. Dairy products are rich with branched chain amino acids that are essential for energy production. To gain insight into the mechanisms underlying the benefit of the sub-chronic effects of running and intake of milk protein supplements, we studied Low Capacity Runner rats (LCR), a rodent exercise model with risk for metabolic disorders. We especially focused on the role of Sirtuins, energy level dependent proteins that affect many cellular metabolic processes. Methods Forty-seven adult LCR female rats sedentary or running voluntarily in wheels were fed normal chow and given supplements of either whey or milk protein drink (PD)-supplemented water, or water only for 21 weeks. Physiological responses were measured in vivo. Blood lipids were determined from serum. Mitochondrial markers and Sirtuins (Sirt1-7) including downstream targets were measured in plantaris muscle by western blotting. Results For the first 10 weeks whey-drinking rats ran about 50% less compared to other groups; still, in all runners glucose tolerance improved and triglycerides decreased. Generally, running induced a ∼six-fold increase in running capacity and a ∼8% decrease in % body fat. Together with running, protein supplements increased the relative lean mass of the total body weight by ∼11%. In comparison with sedentary controls, running and whey increased HDL (21%) and whey, with or without running, lowered LDL (−34%). Running increased mitochondrial biogenesis and Sirtuins 3 and 4. When combined with exercise, both whey and milk protein drink induced about a 4-fold increase in Sirt3, compared to runners drinking water only, and about a 2-fold increase compared to the respective sedentary group. Protein supplements, with or without running, enhanced the phosphorylation level of the acetyl-coA-carboxylase, suggesting increased fat oxidation. Both supplemented diets increased Sirt5 and Sirt7 without an additional effect from exercise. Running diminished and PD supplement increased Sirt6. Conclusion We demonstrate in rats new sub-chronic effects of milk proteins on metabolism that involve Sirtuins and their downstream targets in skeletal muscle. The results show that running and milk proteins act on reducing the risk factors of metabolic disorders and suggest that the underlying mechanisms may involve Sirtuins. Notably, we found that milk protein supplements have some favorable effects on metabolism even without running. Interactive effects of running and/or milk protein supplements were studied. Milk protein drink enhanced and whey diminished the amount of voluntary running. Despite less running whey-supplementation improved metabolic health. Almost all Sirtuins in muscle adapted to milk protein and running interventions.
Collapse
|
56
|
Yue Z, Zhang L, Li C, Chen Y, Tai Y, Shen Y, Sun Z. Advances and potential of gene therapy for type 2 diabetes mellitus. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1643783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Zonghao Yue
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
- Department of Food Science, Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, PR China
| | - Lijuan Zhang
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Chunyan Li
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yanjuan Chen
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yaping Tai
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yihao Shen
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Zhongke Sun
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
- Department of Food Science, Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, PR China
- Department of Molecular Engineering, College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
57
|
Lin ST, Teng LZ, Lin Y, Miao LH, Ge XP, Hao JY, Huang X, Liu B. Molecular and functional characterization of sirt4 and sirt6 in Megalobrama amblycephala under high glucose metabolism. Comp Biochem Physiol B Biochem Mol Biol 2019; 231:87-97. [DOI: 10.1016/j.cbpb.2019.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/06/2018] [Accepted: 01/14/2019] [Indexed: 12/19/2022]
|
58
|
Hernández-Saavedra D, Moody L, Xu GB, Chen H, Pan YX. Epigenetic Regulation of Metabolism and Inflammation by Calorie Restriction. Adv Nutr 2019; 10:520-536. [PMID: 30915465 PMCID: PMC6520046 DOI: 10.1093/advances/nmy129] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/26/2018] [Accepted: 12/17/2018] [Indexed: 12/16/2022] Open
Abstract
Chronic caloric restriction (CR) without malnutrition is known to affect different cellular processes such as stem cell function, cell senescence, inflammation, and metabolism. Despite the differences in the implementation of CR, the reduction of calories produces a widespread beneficial effect in noncommunicable chronic diseases, which can be explained by improvements in immuno-metabolic adaptation. Cellular adaptation that occurs in response to dietary patterns can be explained by alterations in epigenetic mechanisms such as DNA methylation, histone modifications, and microRNA. In this review, we define these modifications and systematically summarize the current evidence related to CR and the epigenome. We then explain the significance of genome-wide epigenetic modifications in the context of disease development. Although substantial evidence exists for the widespread effect of CR on longevity, there is no consensus regarding the epigenetic regulations of the underlying cellular mechanisms that lead to improved health. We provide compelling evidence that CR produces long-lasting epigenetic effects that mediate expression of genes related to immuno-metabolic processes. Epigenetic reprogramming of the underlying chronic low-grade inflammation by CR can lead to immuno-metabolic adaptations that enhance quality of life, extend lifespan, and delay chronic disease onset.
Collapse
Affiliation(s)
| | | | - Guanying Bianca Xu
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Hong Chen
- Division of Nutritional Sciences,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL,Address correspondence to Y-XP (e-mail: )
| |
Collapse
|
59
|
Xu X, Wang D, Zheng C, Gao B, Fan J, Cheng P, Liu B, Yang L, Luo Z. Progerin accumulation in nucleus pulposus cells impairs mitochondrial function and induces intervertebral disc degeneration and therapeutic effects of sulforaphane. Theranostics 2019; 9:2252-2267. [PMID: 31149042 PMCID: PMC6531300 DOI: 10.7150/thno.30658] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/10/2019] [Indexed: 01/18/2023] Open
Abstract
Progerin, a truncated unprocessed lamin A protein, causes tissue aging and degeneration. In this study we explored the role of progerin in the pathogenesis of intervertebral disc degeneration (IDD). We also examined the effect of sulforaphane (SFN) on progerin accumulation and mitochondrial dysfunction in IDD. Methods: The role of progerin in IDD was explored using human nucleus pulposus (NP) tissues, rat NP cells, and Lmna G609G knock-in mice. Immunostaining, X-ray imaging, and Western blotting were performed to assess the phenotypes of intervertebral discs. Alterations in senescence and apoptosis were evaluated by SA-β-galactosidase, immunofluorescence, flow cytometry, and TUNEL assays. Mitochondrial function was investigated by JC-1 staining, transmission electron microscopy, and determination of the level of ATP and the activities of mitochondrial enzymes. Results: The progerin level was elevated in degenerated human NP tissues. Lmna G609G/G609G mice displayed IDD, as evidenced by increased matrix metalloproteinase-13 expression and decreased collagen II and aggrecan expression and disc height. Furthermore, progerin overexpression in rat NP cells induced mitochondrial dysfunction (decreased ATP synthesis, mitochondrial membrane potential, and activities of mitochondrial complex enzymes), morphologic abnormalities, and disrupted mitochondrial dynamic (abnormal expression of proteins involved in fission and fusion), resulting in apoptosis and senescence. SFN ameliorated the progerin-induced aging defects and mitochondrial dysfunction in NP cells and IDD in Lmna G609G/G609G mice. Conclusions: Progerin is involved in the pathogenesis of IDD. Also, SFN alleviates progerin‑induced IDD, which is associated with amelioration of aging defects and mitochondrial dysfunction. Thus, SFN shows promise for the treatment of IDD.
Collapse
|
60
|
Samant SA, Pillai VB, Gupta MP. Cellular mechanisms promoting cachexia and how they are opposed by sirtuins 1. Can J Physiol Pharmacol 2019; 97:235-245. [PMID: 30407871 DOI: 10.1139/cjpp-2018-0479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many chronic diseases are associated with unintentional loss of body weight, which is termed "cachexia". Cachexia is a complex multifactorial syndrome associated with the underlying primary disease, and characterized by loss of skeletal muscle with or without loss of fat tissue. Patients with cachexia face dire symptoms like dyspnea, fatigue, edema, exercise intolerance, and low responsiveness to medical therapy, which worsen quality of life. Because cachexia is not a stand-alone disorder, treating primary disease - such as cancer - takes precedence for the physician, and it remains mostly a neglected illness. Existing clinical trials have demonstrated limited success mostly because of their monotherapeutic approach and late detection of the syndrome. To conquer cachexia, it is essential to identify as many molecular targets as possible using the latest technologies we have at our disposal. In this review, we have discussed different aspects of cachexia, which include various disease settings, active molecular pathways, and recent novel advances made in this field to understand consequences of this illness. We also discuss roles of the sirtuins, the NAD+-dependent lysine deacetylases, microRNAs, certain dietary options, and epigenetic drugs as potential approaches, which can be used to tackle cachexia as early as possible in its course.
Collapse
Affiliation(s)
- Sadhana A Samant
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Vinodkumar B Pillai
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Mahesh P Gupta
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
- Department of Surgery, Committee on Molecular and Cellular Physiology, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
61
|
Tang C, Liu P, Zhou Y, Jiang B, Song Y, Sheng L. Sirt6 deletion in hepatocytes increases insulin sensitivity of female mice by enhancing ERα expression. J Cell Physiol 2019; 234:18615-18625. [PMID: 30912134 DOI: 10.1002/jcp.28500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 01/14/2023]
Abstract
Sirtuin 6 (Sirt6), a NAD+ -dependent protein deacetylase, is involved in hepatic glucose metabolism and insulin sensitivity, which impact metabolic homeostasis. In this paper, we discover that Sirt6 affects the insulin sensitivity of mice in a gender-dependent manner; few studies have been conducted on this issue. Based on reports revealing the influences of sex hormones on insulin signaling, this investigation explores the mechanism by which Sirt6 regulates the estrogen pathway and disrupts insulin signal transduction. Hepatocyte-specific Sirt6 knockout (Sirt6HKO) mice were generated to investigate the function of Sirt6 in hepatocytes. Mice were castrated or spayed to eliminate sex hormones. Insulin sensitivity was assessed via an insulin tolerance test (ITT) in vivo. The interaction of Sirt6 with the estrogen pathway and their impacts on insulin signal transduction were revealed by immunoblot and immunoprecipitation. Sirt6 deletion in hepatocytes significantly enhanced insulin sensitivity and signal transduction in female mice but not in male or spayed female mice as demonstrated by ITT and the phosphorylation level of Akt in the liver. We also identified upregulation of p300, ERα, and interaction of ERα with p85 in the liver of female Sirt6HKO mice. Additionally, Sirt6 was found to inhibit ERα protein stability in a p300-dependent manner without interacting directly with ERα. Our findings show that hepatic Sirt6 downregulates the ERα protein level in a p300-dependent manner and thus disturbs estrogen-induced improvement in insulin sensitivity in the liver, which may partially explain the gender difference in insulin sensitivity.
Collapse
Affiliation(s)
- Chuanfeng Tang
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peiyu Liu
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Zhou
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bijie Jiang
- Pharmacy College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yu Song
- Pharmacy College, Xinxiang Medical University, Xinxiang, Henan, China
| | - Liang Sheng
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu, China.,Pharmacy College, Xinxiang Medical University, Xinxiang, Henan, China.,Key Laboratory of Rare Metabolic Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
62
|
Fritzen AM, Lundsgaard A, Jeppesen JF, Sjøberg KA, Høeg LD, Deleuran HH, Wojtaszewski JFP, Richter EA, Kiens B. Fatty acid type–specific regulation of SIRT1 does not affect insulin sensitivity in human skeletal muscle. FASEB J 2019; 33:5510-5519. [DOI: 10.1096/fj.201801950r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Andreas Mæchel Fritzen
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Anne‐Marie Lundsgaard
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Jacob Fuglsbjerg Jeppesen
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Kim Anker Sjøberg
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Louise Dalgas Høeg
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Henrik Hall Deleuran
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Jørgen F. P. Wojtaszewski
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Erik A. Richter
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Bente Kiens
- Section of Molecular PhysiologyDepartment of Nutrition, Exercise, and Sports, Faculty of ScienceUniversity of Copenhagen Copenhagen Denmark
| |
Collapse
|
63
|
Fan Y, Yang Q, Yang Y, Gao Z, Ma Y, Zhang L, Liang W, Ding G. Sirt6 Suppresses High Glucose-Induced Mitochondrial Dysfunction and Apoptosis in Podocytes through AMPK Activation. Int J Biol Sci 2019; 15:701-713. [PMID: 30745856 PMCID: PMC6367578 DOI: 10.7150/ijbs.29323] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Previous studies have shown that mitochondrial dysfunction plays an important role in high- glucose(HG)-induced podocyte injury and thus contributes to the progression of diabetic nephropathy(DN). The histone deacetylase Sirtuin6 (Sirt6) has been revealed to have an essential role in the regulation of mitochondrial function in skeletal muscle and cardiomyocytes. However, its specific role in mitochondrial homeostasis in podocytes is undetermined. Here, we aimeds to explore the physiological function of Sirt6 in podocyte mitochondria and apoptosis under HG conditions and explore the possible mechanism. Herein, we observed that Sirt6-WT-1 colocalization was suppressed in the glomeruli of patients with DN. In addition, diabetic mice exhibited reduced Sirt6 expression and AMP kinase (AMPK) dephosphorylation accompanied by mitochondrial morphological abnormalities. In vitro, podocytes exposed to HG presented with mitochondrial morphological alterations and podocyte apoptosis accompanied by Sirt6 and p-AMPK downregulation. In addition, HG promoted a decrease in mitochondrial number and an increase in mitochondrial superoxide production as well as a decreased mitochondrial membrane potential. ROS production was also increased in HG-treated podocytes. Conversely, all these mitochondrial defects induced by HG were significantly alleviated by Sirt6 plasmid transfection. Sirt6 overexpression simultaneously alleviated HG-induced podocyte apoptosis and oxidative stress, as well as increased AMPK phosphorylation. Increased levels of H3K9ac and H3K56ac induced by HG were attenuated in podocytes transfected with Sirt6 plasmids. Therefore, these results elucidated that Sirt6 protects mitochondria of podocytes and exerts anti-apoptotic effects via activating AMPK pathway. The present findings provide key insights into the pivotal role of mitochondria regulation by SIRT6 in its protective effects on podocytes.
Collapse
Affiliation(s)
- Yanqin Fan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yingjie Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhao Gao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiqiong Ma
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
64
|
Hsu YC, Wu YT, Tsai CL, Wei YH. Current understanding and future perspectives of the roles of sirtuins in the reprogramming and differentiation of pluripotent stem cells. Exp Biol Med (Maywood) 2019; 243:563-575. [PMID: 29557214 DOI: 10.1177/1535370218759636] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In mammalian cells, there are seven members of the sirtuin protein family (SIRT1-7). SIRT1, SIRT6, and SIRT7 catalyze posttranslational modification of proteins in the nucleus, SIRT3, SIRT4, and SIRT5 are in the mitochondria and SIRT2 is in the cytosol. SIRT1 can deacetylate the transcription factor SOX2 and regulate induced pluripotent stem cells (iPSCs) reprogramming through the miR-34a-SIRT1-p53 axis. SIRT2 can regulate the function of pluripotent stem cells through GSK3β. SIRT3 can positively regulate PPAR gamma coactivator 1-alpha (PGC-1α) expression during the differentiation of stem cells. SIRT4 has no direct role in regulating reprogramming but may have the potential to prevent senescence of somatic cells and to facilitate the reprogramming of iPSCs. SIRT5 can deacetylate STAT3, which is an important transcription factor in regulating pluripotency and differentiation of stem cells. SIRT6 can enhance the reprogramming efficiency of iPSCs from aged skin fibroblasts through miR-766 and increase the expression levels of the reprogramming genes including Sox2, Oct4, and Nanog through acetylation of histone H3 lysine 56. SIRT7 plays a regulatory role in the process of mesenchymal-to-epithelial transition (MET), which has been suggested to be a crucial process in the generation of iPSCs from fibroblasts. In this review, we summarize recent findings of the roles of sirtuins in the metabolic reprogramming and differentiation of stem cells and discuss the bidirectional changes in the gene expression and activities of sirtuins in the commitment of differentiation of mesenchymal stem cells (MSCs) and reprogramming of somatic cells to iPSCs, respectively. Thus, understanding the molecular basis of the interplay between different sirtuins and mitochondrial function will provide new insights into the regulation of differentiation of stem cells and iPSCs formation, respectively, and may help design effective stem cell therapies for regenerative medicine. Impact statement This is an extensive review of the recent advances in our understanding of the roles of some members of the sirtuins family, such as SIRT1, SIRT2, SIRT3, and SIRT6, in the regulation of intermediary metabolism during stem cell differentiation and in the reprogramming of somatic cells to form induced pluripotent stem cells (iPSCs). This article provides an updated integrated view on the mechanisms by which sirtuins-mediated posttranslational protein modifications regulate mitochondrial biogenesis, bioenergetics, and antioxidant defense in the maintenance and differentiation of stem cells and in iPSCs formation, respectively.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan.,*These two authors made equal contributions
| | - Yu-Ting Wu
- 2 Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan.,*These two authors made equal contributions
| | - Chia-Ling Tsai
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan
| | - Yau-Huei Wei
- 1 Institute of Biomedical Sciences, 145474 Mackay Medical College , New Taipei City 252, Taiwan.,2 Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
65
|
Kitada M, Ogura Y, Monno I, Koya D. Sirtuins and Type 2 Diabetes: Role in Inflammation, Oxidative Stress, and Mitochondrial Function. Front Endocrinol (Lausanne) 2019; 10:187. [PMID: 30972029 PMCID: PMC6445872 DOI: 10.3389/fendo.2019.00187] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 03/06/2019] [Indexed: 01/05/2023] Open
Abstract
The rising incidence of type 2 diabetes mellitus (T2DM) is a major public health concern, and novel therapeutic strategies to prevent T2DM are urgently needed worldwide. Aging is recognized as one of the risk factors for metabolic impairments, including insulin resistance and T2DM. Inflammation, oxidative stress, and mitochondrial dysfunction are closely related to both aging and metabolic disease. Calorie restriction (CR) can retard the aging process in organisms ranging from yeast to rodents and delay the onset of numerous age-related disorders, such as insulin resistance and diabetes. Therefore, metabolic CR mimetics may represent new therapeutic targets for insulin resistance and T2DM. Sirtuin 1 (SIRT1), the mammalian homolog of Sir2, was originally identified as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase. The activation of SIRT1 is closely associated with longevity under CR, and it is recognized as a CR mimetic. Currently, seven sirtuins have been identified in mammals. Among these sirtuins, SIRT1 and SIRT2 are located in the nucleus and cytoplasm, SIRT3 exists predominantly in mitochondria, and SIRT6 is located in the nucleus. These sirtuins regulate metabolism through their regulation of inflammation, oxidative stress and mitochondrial function via multiple mechanisms, resulting in the improvement of insulin resistance and T2DM. In this review, we describe the current understanding of the biological functions of sirtuins, especially SIRT1, SIRT2, SIRT3, and SIRT6, focusing on oxidative stress, inflammation, and mitochondrial function, which are closely associated with aging.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
- *Correspondence: Munehiro Kitada
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Itaru Monno
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
66
|
Gao T, Li M, Mu G, Hou T, Zhu WG, Yang Y. PKCζ Phosphorylates SIRT6 to Mediate Fatty Acid β-Oxidation in Colon Cancer Cells. Neoplasia 2018; 21:61-73. [PMID: 30504065 PMCID: PMC6277223 DOI: 10.1016/j.neo.2018.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/12/2018] [Accepted: 11/12/2018] [Indexed: 01/10/2023] Open
Abstract
Protein kinase C (PKC) has critical roles in regulating lipid anabolism and catabolism. PKCζ, a member of atypical PKC family, has been reported to mediate glucose metabolism. However, whether and how PKCζ regulates tumor cells fatty acid β-oxidation are unknown. Here, we report that the phosphorylation of SIRT6 is significantly increased after palmitic acid (PA) treatment in colon cancer cells. PKCζ can physically interact with SIRT6 in vitro and in vivo, and this interaction enhances following PA treatment. Further experiments show that PKCζ is the phosphorylase of SIRT6 and phosphorylates SIRT6 at threonine 294 residue to promote SIRT6 enrichment on chromatin. In the functional study, we find that the expression of ACSL1, CPT1, CACT, and HADHB, the genes related to fatty acid β-oxidation, increases after PA stimulation. We further confirm that PKCζ mediates the binding of SIRT6 specifically to the promoters of fatty acid β-oxidation–related genes and elicits the expression of these genes through SIRT6 phosphorylation. Our findings demonstrate the mechanism of PKCζ as a new phosphorylase of SIRT6 on maintaining tumor fatty acid β-oxidation and define the new role of PKCζ in lipid homeostasis.
Collapse
Affiliation(s)
- Tian Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Meiting Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Guanqun Mu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Tianyun Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China; Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 516080, China
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China.
| |
Collapse
|
67
|
Huang L, Sun H, Song F, Cao Z, Jiang X, Zhang L, Li Z, Huang C. SIRT6 overexpression inhibits cementogenesis by suppressing glucose transporter 1. J Cell Physiol 2018; 234:4005-4014. [PMID: 30256390 DOI: 10.1002/jcp.27213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/13/2018] [Indexed: 01/13/2023]
Abstract
Cementum, which shares common features with bone in terms of biochemical composition, is important for the homeostasis of periodontium during periodontitis and orthodontic treatment. Sirtuin 6 (SIRT6), as a member of the sirtuin family, plays key roles in the osteogenic differentiation of bone marrow mesenchymal stem cells. However, the involvement of SIRT6 in cementoblast differentiation and mineralization and the underlying mechanisms remain unknown. In this study, we observed that the expression of SIRT6 increased during cementoblast differentiation initially. Analysis of the gain- and loss-of-function indicated that overexpressing SIRT6 in OCCM-30 cells suppresses cementoblast differentiation and mineralization and downregulating SIRT6 promotes cementogenesis. GLUT1, a glucose transporter necessary in cementogenesis, was inhibited by SIRT6. Overexpressing GLUT1 in SIRT6-overexpressed OCCM-30 cells rescued the inhibitory effect of SIRT6 on cementoblast differentiation and mineralization. Moreover, AMPK was activated after overexpressing SIRT6 and inhibited cementoblast differentiation and mineralization. Downregulating the expression of SIRT6 inhibited AMPK activity. Meanwhile, GLUT1 overexpression significantly decreased AMPK activity. Overall, on one hand, SIRT6 inhibited cementoblast differentiation and mineralization by suppressing GLUT1. On the other hand, SIRT6 inhibited cementoblast differentiation and mineralization by activating the AMPK pathway. GLUT1 overexpression also rescued the increased AMPK pathway activated by SIRT6.
Collapse
Affiliation(s)
- Liyuan Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Hualing Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China.,Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Fangfang Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Zhengguo Cao
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xinquan Jiang
- Department of Prosthodontics, School of Medicine, Ninth People's Hospital affiliated to Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Zhelun Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
68
|
Abstract
PURPOSE OF REVIEW The role of mitochondria in the development of skeletal muscle insulin resistance has been an area of intense investigation and debate for over 20 years. The mitochondria is a multifaceted organelle that plays an integral part in substrate metabolism and cellular signalling. This article aims to summarize the current findings and thought regarding the relationship between mitochondria and insulin resistance in skeletal muscle. RECENT FINDINGS Skeletal muscle insulin resistance was earlier thought to result from deficiency in mitochondrial oxidative capacity and ectopic lipid accumulation. Recent evidence suggests that skeletal muscle insulin resistance in high-energy intake models (i.e. obesity) results primarily from disrupted mitochondrial bioenergetics and alterations in mitochondrial-associated cell signalling. These signalling pathways include reactive oxygen species and redox balance, fatty acid β-oxidation intermediates, mitochondrial derived peptides, sirtuins, microRNAs and novel nuclear-encoded, mitochondria-acting peptides. SUMMARY The pathophysiology of skeletal muscle insulin resistance is likely multifactorial involving many coordinated physiological processes. However, it is apparent that the mitochondria play an essential role in skeletal muscle insulin sensitivity in health, ageing and in numerous metabolic diseases. Deciphering the manifold functions of the mitochondria will allow us to understand the complex relationship between mitochondria and skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- William Todd Cade
- Program in Physical Therapy & Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
69
|
Abstract
The mammalian Sirtuins (SIRT1-7) are an evolutionarily conserved family of NAD+-dependent deacylase and mono-ADP-ribosyltransferase. Sirtuins display distinct subcellular localizations and functions and are involved in cell survival, senescence, metabolism and genome stability. Among the mammalian Sirtuins, SIRT1 and SIRT6 have been thoroughly investigated and have prominent metabolic regulatory roles. Moreover, SIRT1 and SIRT6 have been implicated in obesity, insulin resistance, type 2 diabetes mellitus (T2DM), fatty liver disease and cardiovascular diseases. However, the roles of other Sirtuins are not fully understood. Recent studies have shown that these Sirtuins also play important roles in inflammation, mitochondrial dysfunction, and energy metabolism. Insulin resistance is the critical pathological trait of obesity and metabolic syndrome as well as the core defect in T2DM. Accumulating clinical and experimental animal evidence suggests the potential roles of the remaining Sirtuins in the regulation of insulin resistance through diverse biological mechanisms. In this review, we summarize recent advances in the understanding of the functions of Sirtuins in various insulin resistance-associated physiological processes, including inflammation, mitochondrial dysfunction, the insulin signaling pathway, glucose, and lipid metabolism. In addition, we highlight the important gaps that must be addressed in this field.
Collapse
Affiliation(s)
- Shuang Zhou
- Internal Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaoqiang Tang
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Hou-Zao Chen ;
| |
Collapse
|
70
|
Bae EJ. Sirtuin 6, a possible therapeutic target for type 2 diabetes. Arch Pharm Res 2017; 40:1380-1389. [DOI: 10.1007/s12272-017-0989-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/16/2017] [Indexed: 12/27/2022]
|