1
|
Grootaert MOJ. Cell senescence in cardiometabolic diseases. NPJ AGING 2024; 10:46. [PMID: 39433786 PMCID: PMC11493982 DOI: 10.1038/s41514-024-00170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024]
Abstract
Cellular senescence has been implicated in many age-related pathologies including atherosclerosis, heart failure, age-related cardiac remodeling, diabetic cardiomyopathy and the metabolic syndrome. Here, we will review the characteristics of senescent cells and their endogenous regulators, and summarize the metabolic stressors that induce cell senescence. We will discuss the evidence of cell senescence in the onset and progression of several cardiometabolic diseases and the therapeutic potential of anti-senescence therapies.
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Endocrinology, Diabetes and Nutrition, UCLouvain, Brussels, Belgium.
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Frohlich J, Liorni N, Mangoni M, Lochmanová G, Pírek P, Kaštánková N, Pata P, Kucera J, Chaldakov GN, Tonchev AB, Pata I, Gorbunova V, Leire E, Zdráhal Z, Mazza T, Vinciguerra M. Epigenetic and transcriptional control of adipocyte function by centenarian-associated SIRT6 N308K/A313S mutant. Clin Epigenetics 2024; 16:96. [PMID: 39033117 PMCID: PMC11265064 DOI: 10.1186/s13148-024-01710-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Obesity is a major health burden. Preadipocytes proliferate and differentiate in mature adipocytes in the adipogenic process, which could be a potential therapeutic approach for obesity. Deficiency of SIRT6, a stress-responsive protein deacetylase and mono-ADP ribosyltransferase enzyme, blocks adipogenesis. Mutants of SIRT6 (N308K/A313S) were recently linked to the in the long lifespan Ashkenazi Jews. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect adipogenesis at the transcriptional and epigenetic level. METHODS We analyzed the role of SIRT6 wild-type (WT) or SIRT6 centenarian-associated mutant (N308K/A313S) overexpression in adipogenesis, by creating stably transduced preadipocyte cell lines using lentivirus on the 3T3-L1 model. Histone post-translational modifications (PTM: acetylation, methylation) and transcriptomic changes were analyzed by mass spectrometry (LC-MS/MS) and RNA-Seq, respectively, in 3T3-L1 adipocytes. In addition, the adipogenic process and related signaling pathways were investigated by bioinformatics and biochemical approaches. RESULTS Overexpression of centenarian-associated SIRT6 mutant increased adipogenic differentiation to a similar extent compared to the WT form. However, it triggered distinct histone PTM profiles in mature adipocytes, with significantly higher acetylation levels, and activated divergent transcriptional programs, including those dependent on signaling related to the sympathetic innervation and to PI3K pathway. 3T3-L1 mature adipocytes overexpressing SIRT6 N308K/A313S displayed increased insulin sensitivity in a neuropeptide Y (NPY)-dependent manner. CONCLUSIONS SIRT6 N308K/A313S overexpression in mature adipocytes ameliorated glucose sensitivity and impacted sympathetic innervation signaling. These findings highlight the importance of targeting SIRT6 enzymatic activities to regulate the co-morbidities associated with obesity.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Niccolò Liorni
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Manuel Mangoni
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Gabriela Lochmanová
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavlína Pírek
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Nikola Kaštánková
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | | | - Jan Kucera
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| | - George N Chaldakov
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Faculty of Medicine, Varna, Bulgaria
| | - Anton B Tonchev
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Faculty of Medicine, Varna, Bulgaria
| | | | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Eric Leire
- GenFlow Biosciences Srl, Charleroi, Belgium
- Clinique 135, Brussels, Belgium
| | - Zbyněk Zdráhal
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tommaso Mazza
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.
- Faculty of Science, Liverpool John Moores University (LJMU), Liverpool, UK.
| |
Collapse
|
3
|
Yang Z, Li S, Zhao C, Zhao Z, Tan J, Zhang L, Huang Y. X-Box binding protein 1 downregulates SIRT6 to promote injury in pancreatic ductal epithelial cells. Immun Inflamm Dis 2024; 12:e1301. [PMID: 38967361 PMCID: PMC11225082 DOI: 10.1002/iid3.1301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/19/2024] [Accepted: 05/19/2024] [Indexed: 07/06/2024] Open
Abstract
OBJECTIVE Acute pancreatitis (AP) stands as a frequent cause for clinical emergency hospital admissions. The X-box binding protein 1 (XBP1) was found to be implicated in pancreatic acinar cell apoptosis. The objective is to unveil the potential mechanisms governed by XBP1 and SIRT6 in the context of AP. METHODS Caerulein-treated human pancreatic duct epithelial (HPDE) cells to establish an in vitro research model. The levels and regulatory role of SIRT6 in the treated cells were evaluated, including its effects on inflammatory responses, oxidative stress, apoptosis, and endoplasmic reticulum stress. The relationship between XBP1 and SIRT6 was explored by luciferase and ChIP experiments. Furthermore, the effect of XBP1 overexpression on the regulatory function of SIRT6 on cells was evaluated. RESULTS Caerulein promoted the decrease of SIRT6 and the increase of XBP1 in HPDE cells. Overexpression of SIRT6 slowed down the secretion of inflammatory factors, oxidative stress, apoptosis level, and endoplasmic reticulum stress in HPDE cells. However, XBP1 negatively regulated SIRT6, and XBP1 overexpression partially reversed the regulation of SIRT6 on the above aspects. CONCLUSION Our study illuminates the role of XBP1 in downregulating SIRT6 in HPDE cells, thereby promoting cellular injury. Inhibiting XBP1 or augmenting SIRT6 levels holds promise in preserving cell function and represents a potential therapeutic avenue in the management of AP.
Collapse
Affiliation(s)
- Zhuo Yang
- Intensive Care Unit, Bazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Shaojun Li
- Acupuncture and Rehabilitation DepartmentBazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Chuan Zhao
- Intensive Care Unit, Bazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Zongzheng Zhao
- Intensive Care Unit, Bazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Juan Tan
- Intensive Care Unit, Bazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Lu Zhang
- Acupuncture and Rehabilitation DepartmentBazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| | - Yuanqing Huang
- Intensive Care Unit, Bazhong Hospital of Traditional Chinese MedicineBazhongSichuanChina
| |
Collapse
|
4
|
Kumar KK, Aburawi EH, Ljubisavljevic M, Leow MKS, Feng X, Ansari SA, Emerald BS. Exploring histone deacetylases in type 2 diabetes mellitus: pathophysiological insights and therapeutic avenues. Clin Epigenetics 2024; 16:78. [PMID: 38862980 PMCID: PMC11167878 DOI: 10.1186/s13148-024-01692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the β-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting β-cells and reducing T2DM-associated complications, among others.
Collapse
Affiliation(s)
- Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Elhadi Husein Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Milos Ljubisavljevic
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Melvin Khee Shing Leow
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
- Dept of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Xu Feng
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates.
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
5
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
6
|
Morisseau L, Tokito F, Lucas M, Poulain S, Kim SH, Plaisance V, Pawlowski V, Legallais C, Jellali R, Sakai Y, Abderrahmani A, Leclerc E. Transcriptomic profiling analysis of the effect of palmitic acid on 3D spheroids of β-like cells derived from induced pluripotent stem cells. Gene 2024; 917:148441. [PMID: 38608795 DOI: 10.1016/j.gene.2024.148441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
Type 2 diabetes (T2D) is posing a serious public health concern with a considerable impact on human life and health expenditures worldwide. The disease develops when insulin plasma level is insufficient for coping insulin resistance, caused by the decline of pancreatic β-cell function and mass. In β-cells, the lipotoxicity exerted by saturated free fatty acids in particular palmitate (PA), which is chronically elevated in T2D, plays a major role in β-cell dysfunction and mass. However, there is a lack of human relevant in vitro model to identify the underlying mechanism through which palmitate induces β-cell failure. In this frame, we have previously developed a cutting-edge 3D spheroid model of β-like cells derived from human induced pluripotent stem cells. In the present work, we investigated the signaling pathways modified by palmitate in β-like cells derived spheroids. When compared to the 2D monolayer cultures, the transcriptome analysis (FDR set at 0.1) revealed that the 3D spheroids upregulated the pancreatic markers (such as GCG, IAPP genes), lipids metabolism and transporters (CD36, HMGSC2 genes), glucose transporter (SLC2A6). Then, the 3D spheroids are exposed to PA 0.5 mM for 72 h. The differential analysis demonstrated that 32 transcription factors and 135 target genes were mainly modulated (FDR set at 0.1) including the upregulation of lipid and carbohydrates metabolism (HMGSC2, LDHA, GLUT3), fibrin metabolism (FGG, FGB), apoptosis (CASP7). The pathway analysis using the 135 selected targets extracted the fibrin related biological process and wound healing in 3D PA treated conditions. An overall pathway gene set enrichment analysis, performed on the overall gene set (with pathway significance cutoff at 0.2), highlighted that PA perturbs the citrate cycle, FOXO signaling and Hippo signaling as observed in human islets studies. Additional RT-PCR confirmed induction of inflammatory (IGFBP1, IGFBP3) and cell growth (CCND1, Ki67) pathways by PA. All these changes were associated with unaffected glucose-stimulated insulin secretion (GSIS), suggesting that they precede the defect of insulin secretion and death induced by PA. Overall, we believe that our data demonstrate the potential of our spheroid 3D islet-like cells to investigate the pancreatic-like response to diabetogenic environment.
Collapse
Affiliation(s)
- Lisa Morisseau
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu CS 60319, 60203 Compiègne Cedex, France
| | - Fumiya Tokito
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Mathilde Lucas
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Stéphane Poulain
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Soo Hyeon Kim
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Valérie Plaisance
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valérie Pawlowski
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Cécile Legallais
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu CS 60319, 60203 Compiègne Cedex, France
| | - Rachid Jellali
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de recherche Royallieu CS 60319, 60203 Compiègne Cedex, France
| | - Yasuyuki Sakai
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; CNRS/IIS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Eric Leclerc
- CNRS/IIS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan.
| |
Collapse
|
7
|
Pierre TH, Toren E, Kepple J, Hunter CS. Epigenetic Regulation of Pancreas Development and Function. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 239:1-30. [PMID: 39283480 DOI: 10.1007/978-3-031-62232-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
The field of epigenetics broadly seeks to define heritable phenotypic modifications that occur within cells without changes to the underlying DNA sequence. These modifications allow for precise control and specificity of function between cell types-ultimately creating complex organ systems that all contain the same DNA but only have access to the genes and sequences necessary for their cell-type-specific functions. The pancreas is an organ that contains varied cellular compartments with functions ranging from highly regulated glucose-stimulated insulin secretion in the β-cell to the pancreatic ductal cells that form a tight epithelial lining for the delivery of digestive enzymes. With diabetes cases on the rise worldwide, understanding the epigenetic mechanisms driving β-cell identity, function, and even disease is particularly valuable. In this chapter, we will discuss the known epigenetic modifications in pancreatic islet cells, how they are deposited, and the environmental and metabolic contributions to epigenetic mechanisms. We will also explore how a deeper understanding of epigenetic effectors can be used as a tool for diabetes therapeutic strategies.
Collapse
Affiliation(s)
- Tanya Hans Pierre
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eliana Toren
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessica Kepple
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chad S Hunter
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
8
|
Wang Y, Liu T, Cai Y, Liu W, Guo J. SIRT6's function in controlling the metabolism of lipids and glucose in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1244705. [PMID: 37876546 PMCID: PMC10591331 DOI: 10.3389/fendo.2023.1244705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes mellitus (DM) and the main cause of excess mortality in patients with type 2 DM. The pathogenesis and progression of DN are closely associated with disorders of glucose and lipid metabolism. As a member of the sirtuin family, SIRT6 has deacetylation, defatty-acylation, and adenosine diphosphate-ribosylation enzyme activities as well as anti-aging and anticancer activities. SIRT6 plays an important role in glucose and lipid metabolism and signaling, especially in DN. SIRT6 improves glucose and lipid metabolism by controlling glycolysis and gluconeogenesis, affecting insulin secretion and transmission and regulating lipid decomposition, transport, and synthesis. Targeting SIRT6 may provide a new therapeutic strategy for DN by improving glucose and lipid metabolism. This review elaborates on the important role of SIRT6 in glucose and lipid metabolism, discusses the potential of SIRT6 as a therapeutic target to improve glucose and lipid metabolism and alleviate DN occurrence and progression of DN, and describes the prospects for future research.
Collapse
Affiliation(s)
- Ying Wang
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuzi Cai
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Chen H, Zhang X, Liao N, Ji Y, Mi L, Gan Y, Su Y, Wen F. Downregulation of SIRT6 and NMNAT2 is associated with proliferative diabetic retinopathy. Mol Vis 2023; 29:160-168. [PMID: 38222451 PMCID: PMC10784219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/30/2023] [Indexed: 01/16/2024] Open
Abstract
Purpose To determine the expression levels of SIRT6 and NMNAT2 in diabetic retinopathy (DR). Methods We obtained peripheral blood mononuclear cells (PBMCs) and vitreous samples from 77 patients with type 2 diabetes mellitus: 52 with DR and 25 without DR, and 27 healthy control subjects. Western blot analysis and qRT-PCR were performed to evaluate the expression of SIRT6 and NMNAT2 in their PBMCs. The levels of IL-1β, IL-6, and TNF-α in the vitreous fluid were determined by ELISA. Immunohistochemistry was performed to detect the expression of SIRT6 and NMNAT2 in proliferative DR (PDR) and the control subjects. Results The expression of SIRT6 and NMNAT2 was markedly downregulated in DR patients, which was negatively correlated with the increased expression of IL-1β, IL-6 and TNF-α. Additionally, we observed decreased expression of SIRT6 and NMNAT2 in the fibrovascular membranes of PDR patients. Conclusions The downregulated expression of SIRT6 and NMNAT2 in PDR patients reveals a potential pathogenic association; more extended studies could verify them as potential therapeutic targets.
Collapse
Affiliation(s)
- Hui Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiongze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Nanying Liao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuying Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lan Mi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuhong Gan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yongyue Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
10
|
Song N, Guan X, Zhang S, Wang Y, Wang X, Lu Z, Chong D, Wang JY, Yu R, Yu W, Jiang T, Gu Y. Discovery of a pyrrole-pyridinimidazole derivative as novel SIRT6 inhibitor for sensitizing pancreatic cancer to gemcitabine. Cell Death Dis 2023; 14:499. [PMID: 37542062 PMCID: PMC10403574 DOI: 10.1038/s41419-023-06018-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 07/17/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
Pancreatic cancer is a highly aggressive cancer, and is primarily treated with gemcitabine, with increasing resistance. SIRT6 as a member of sirtuin family plays important roles in lifespan and diverse diseases, such as cancer, diabetes, inflammation and neurodegenerative diseases. Considering the role of SIRT6 in the cytoprotective effect, it might be a potential anticancer drug target, and is associated with resistance to anticancer therapy. However, very few SIRT6 inhibitors have been reported. Here, we reported the discovery of a pyrrole-pyridinimidazole derivative, 8a, as a new non-competitive SIRT6 inhibitor, and studied its roles and mechanisms in the antitumor activity and sensitization of pancreatic cancer to gemcitabine. Firstly, we found a potent SIRT6 inhibitor compound 8a by virtual screening and identified by molecular and cellular SIRT6 activity assays. 8a could effectively inhibit SIRT6 deacetylation activity with IC50 values of 7.46 ± 0.79 μM in FLUOR DE LYS assay, and 8a significantly increased the acetylation levels of H3 in cells. Then, we found that 8a could inhibit the cell proliferation and induce cell apoptosis in pancreatic cancer cells. We further demonstrate that 8a sensitize pancreatic cancer cells to gemcitabine via reversing the activation of PI3K/AKT/mTOR and ERK signaling pathways induced by gemcitabine and blocking the DNA damage repair pathway. Moreover, combination of 8a and gemcitabine induces cooperative antitumor activity in pancreatic cancer xenograft model in vivo. Overall, we demonstrate that 8a, a novel SIRT6 inhibitor, could be a promising potential drug candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Nannan Song
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xian Guan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Siqi Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yanqing Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xuekai Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Zhongxia Lu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Daochen Chong
- Department of Pathology, 971 Hospital of PLA Navy, Qingdao, 266071, China
| | - Jennifer Yiyang Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Wengong Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Yuchao Gu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
11
|
Hashemi N, Tabatabaee SH, Shams F, Rahimpour A, Kazemi B, Rajabibazl M, Ranjbari J. Overexpression of SIRT6 alleviates apoptosis and enhances cell viability and monoclonal antibody expression in CHO-K1 cells. Mol Biol Rep 2023:10.1007/s11033-023-08483-5. [PMID: 37286776 DOI: 10.1007/s11033-023-08483-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/24/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Chinese hamster ovary (CHO) cells are the most predominantly utilized host for the production of monoclonal antibodies (mAbs) and other complex glycoproteins. A major challenge in the process of CHO cell culture is the occurrence of cell death following different stressful conditions, which hinders the production yield. Engineering genes involved in pathways related to cell death is a remarkable strategy to delay apoptosis, improve cell viability and enhance productivity. SIRT6 is a stress-responsive protein that regulates DNA repair, maintains genome integrity, and is critical for longevity and cell survival in organisms. METHODS AND RESULTS In this study, SIRT6 was stably overexpressed in CHO-K1 cells and the impact of its expression on apoptosis related gene expression profile, viability, apoptosis, and mAb productivity was investigated. While a significant increase was observed in Bcl-2 mRNA level, caspase-3 and Bax mRNA levels were decreased in the SIRT6 engineered cells compared to the parental CHO-K1 cells. Moreover, improved cell viability and decreased rate of apoptotic progression was observed in a SIRT6-derived clone in comparision to the CHO-K1 cells during 5 days of batch culture. anti-CD52 IgG1 mAb titers were improved up to 1.7- and 2.8-fold in SIRT6-derived clone during transient and stable expression, respectively. CONCLUSIONS This study indicates the positive effects of SIRT6 overexpression on cell viability and anti-CD52 IgG1 mAb expression in CHO-K1 cells. Further studies are needed to examine the potential of SIRT6-engineered host cells for the production of recombinant biotherapeutics in industrial settings.
Collapse
Affiliation(s)
- Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayed Hassan Tabatabaee
- Department of Life Science Engineering, Faculty of New Sciences and Technology, University of Tehran, Tehran, Iran
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Yan S, Yao N, Li X, Sun M, Yang Y, Cui W, Li B. The Association between the Differential Expression of lncRNA and Type 2 Diabetes Mellitus in People with Hypertriglyceridemia. Int J Mol Sci 2023; 24:ijms24054279. [PMID: 36901708 PMCID: PMC10002095 DOI: 10.3390/ijms24054279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Compared with diabetic patients with normal blood lipid, diabetic patients with dyslipidemia such as high triglycerides have a higher risk of clinical complications, and the disease is also more serious. For the subjects with hypertriglyceridemia, the lncRNAs affecting type 2 diabetes mellitus (T2DM) and the specific mechanisms remain unclear. Transcriptome sequencing was performed on peripheral blood samples of new-onset T2DM (six subjects) and normal blood control (six subjects) in hypertriglyceridemia patients using gene chip technology, and differentially expressed lncRNA profiles were constructed. Validated by the GEO database and RT-qPCR, lncRNA ENST00000462455.1 was selected. Subsequently, fluorescence in situ hybridization (FISH), real-time quantitative polymerase chain reaction (RT-qPCR), CCK-8 assay, flow cytometry, and enzyme-linked immunosorbent assay (ELISA) were used to observe the effect of ENST00000462455.1 on MIN6. When silencing the ENST00000462455.1 for MIN6 in high glucose and high fat, the relative cell survival rate and insulin secretion decreased, the apoptosis rate increased, and the expression of the transcription factors Ins1, Pdx-1, Glut2, FoxO1, and ETS1 that maintained the function and activity of pancreatic β cells decreased (p < 0.05). In addition, we found that ENST00000462455.1/miR-204-3p/CACNA1C could be the core regulatory axis by using bioinformatics methods. Therefore, ENST00000462455.1 was a potential biomarker for hypertriglyceridemia patients with T2DM.
Collapse
Affiliation(s)
- Shoumeng Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
- School of Nursing, Jilin University, Changchun 130021, China
| | - Nan Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
| | - Xiaotong Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
| | - Mengzi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
| | - Yixue Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: (W.C.); (B.L.); Tel.: +86-431-85619455 (W.C.); +86-43185619451 (B.L.)
| | - Bo Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: (W.C.); (B.L.); Tel.: +86-431-85619455 (W.C.); +86-43185619451 (B.L.)
| |
Collapse
|
13
|
Human centenarian-associated SIRT6 mutants modulate hepatocyte metabolism and collagen deposition in multilineage hepatic 3D spheroids. GeroScience 2022; 45:1177-1196. [PMID: 36534275 PMCID: PMC9886743 DOI: 10.1007/s11357-022-00713-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), encompassing fatty liver and its progression into nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC), is one of the rapidly rising health concerns worldwide. SIRT6 is an essential nuclear sirtuin that regulates numerous pathological processes including insulin resistance and inflammation, and recently it has been implicated in the amelioration of NAFLD progression. SIRT6 overexpression protects from formation of fibrotic lesions. However, the underlying molecular mechanisms are not fully delineated. Moreover, new allelic variants of SIRT6 (N308K/A313S) were recently associated with the longevity in Ashkenazi Jews by improving genome maintenance and DNA repair, suppressing transposons and killing cancer cells. Whether these new SIRT6 variants play different or enhanced roles in liver diseases is currently unknown. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect liver metabolism and associated diseases. We present evidence that overexpression of centenarian-associated SIRT6 variants dramatically altered the metabolomic and secretomic profiles of unchallenged immortalized human hepatocytes (IHH). Most amino acids were increased in the SIRT6 N308K/A313S overexpressing IHH when compared to IHH transfected with the SIRT6 wild-type sequence. Several unsaturated fatty acids and glycerophospholipids were increased, and ceramide tended to be decreased upon SIRT6 N308K/A313S overexpression. Furthermore, we found that overexpression of SIRT6 N308K/A313S in a 3D hepatic spheroid model formed by the co-culture of human immortalized hepatocytes (IHH) and hepatic stellate cells (LX2) inhibited collagen deposition and fibrotic gene expression in absence of metabolic or dietary challenges. Hence, our findings suggest that novel longevity associated SIRT6 N308K/A313S variants could favor the prevention of NASH by altering hepatocyte proteome and lipidome.
Collapse
|
14
|
Guo Z, Li P, Ge J, Li H. SIRT6 in Aging, Metabolism, Inflammation and Cardiovascular Diseases. Aging Dis 2022; 13:1787-1822. [PMID: 36465178 PMCID: PMC9662279 DOI: 10.14336/ad.2022.0413] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/13/2022] [Indexed: 07/28/2023] Open
Abstract
As an important NAD+-dependent enzyme, SIRT6 has received significant attention since its discovery. In view of observations that SIRT6-deficient animals exhibit genomic instability and metabolic disorders and undergo early death, SIRT6 has long been considered a protein of longevity. Recently, growing evidence has demonstrated that SIRT6 functions as a deacetylase, mono-ADP-ribosyltransferase and long fatty deacylase and participates in a variety of cellular signaling pathways from DNA damage repair in the early stage to disease progression. In this review, we elaborate on the specific substrates and molecular mechanisms of SIRT6 in various physiological and pathological processes in detail, emphasizing its links to aging (genomic damage, telomere integrity, DNA repair), metabolism (glycolysis, gluconeogenesis, insulin secretion and lipid synthesis, lipolysis, thermogenesis), inflammation and cardiovascular diseases (atherosclerosis, cardiac hypertrophy, heart failure, ischemia-reperfusion injury). In addition, the most recent advances regarding SIRT6 modulators (agonists and inhibitors) as potential therapeutic agents for SIRT6-mediated diseases are reviewed.
Collapse
Affiliation(s)
- Zhenyang Guo
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Peng Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hua Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Oh H, Cho W, Park SY, Abd El-Aty A, Jeong JH, Jung TW. Ginsenoside Rb3 ameliorates podocyte injury under hyperlipidemic conditions via PPARδ- or SIRT6-mediated suppression of inflammation and oxidative stress. J Ginseng Res 2022; 47:400-407. [DOI: 10.1016/j.jgr.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/29/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
|
16
|
Small J, Joblin-Mills A, Carbone K, Kost-Alimova M, Ayukawa K, Khodier C, Dancik V, Clemons PA, Munkacsi AB, Wagner BK. Phenotypic Screening for Small Molecules that Protect β-Cells from Glucolipotoxicity. ACS Chem Biol 2022; 17:1131-1142. [PMID: 35439415 PMCID: PMC9127801 DOI: 10.1021/acschembio.2c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022]
Abstract
Type 2 diabetes is marked by progressive β-cell failure, leading to loss of β-cell mass. Increased levels of circulating glucose and free fatty acids associated with obesity lead to β-cell glucolipotoxicity. There are currently no therapeutic options to address this facet of β-cell loss in obese type 2 diabetes patients. To identify small molecules capable of protecting β-cells, we performed a high-throughput screen of 20,876 compounds in the rat insulinoma cell line INS-1E in the presence of elevated glucose and palmitate. We found 312 glucolipotoxicity-protective small molecules (1.49% hit rate) capable of restoring INS-1E viability, and we focused on 17 with known biological targets. 16 of the 17 compounds were kinase inhibitors with activity against specific families including but not limited to cyclin-dependent kinases (CDK), PI-3 kinase (PI3K), Janus kinase (JAK), and Rho-associated kinase 2 (ROCK2). 7 of the 16 kinase inhibitors were PI3K inhibitors. Validation studies in dissociated human islets identified 10 of the 17 compounds, namely, KD025, ETP-45658, BMS-536924, AT-9283, PF-03814735, torin-2, AZD5438, CP-640186, ETP-46464, and GSK2126458 that reduced glucolipotoxicity-induced β-cell death. These 10 compounds decreased markers of glucolipotoxicity including caspase activation, mitochondrial depolarization, and increased calcium flux. Together, these results provide a path forward toward identifying novel treatments to preserve β-cell viability in the face of glucolipotoxicity.
Collapse
Affiliation(s)
- Jonnell
C. Small
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
- Chemistry
Biology Program, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Aidan Joblin-Mills
- School
of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Kaycee Carbone
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Maria Kost-Alimova
- Center
for the Development of Therapeutics, Broad
Institute, Cambridge, Massachusetts 02142, United States
| | - Kumiko Ayukawa
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
- JT
Pharmaceuticals Inc., Takatsuki 569-1125, Osaka, Japan
| | - Carol Khodier
- Center
for the Development of Therapeutics, Broad
Institute, Cambridge, Massachusetts 02142, United States
| | - Vlado Dancik
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Paul A. Clemons
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Andrew B. Munkacsi
- School
of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Bridget K. Wagner
- Chemical
Biology and Therapeutics Science Program, Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
17
|
Raghu S, Prabhashankar AB, Shivanaiah B, Tripathi E, Sundaresan NR. Sirtuin 6 Is a Critical Epigenetic Regulator of Cancer. Subcell Biochem 2022; 100:337-360. [PMID: 36301499 DOI: 10.1007/978-3-031-07634-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian sirtuin family with deacetylase, deacylase, and mono-ADP-ribosyl-transferase activities. It is a multitasking chromatin-associated protein regulating different cellular and physiological functions in cells. Specifically, SIRT6 dysfunction is implicated in several aging-related human diseases, including cancer. Studies indicate that SIRT6 has a tumor-specific role, and it is considered a tumor suppressor as well as a tumor growth inducer, depending on the type of cancer. In this chapter, we review the role of SIRT6 in metabolism, genomic stability, and cancer. Further, we provide an insight into the interplay of the tumor-suppressing and oncogenic roles of SIRT6 in cancer. Additionally, we discuss the use of small-molecule SIRT6 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Sukanya Raghu
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Arathi Bangalore Prabhashankar
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Bhoomika Shivanaiah
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Ekta Tripathi
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, India.
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| | - Nagalingam Ravi Sundaresan
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| |
Collapse
|
18
|
Paula VG, Sinzato YK, Moraes Souza RQ, Soares TS, Souza FQG, Karki B, Andrade Paes AM, Corrente JE, Damasceno DC, Volpato GT. Metabolic changes in female rats exposed to intrauterine Hyperglycemia and post-weaning consumption of high-fat diet. Biol Reprod 2021; 106:200-212. [PMID: 34668971 DOI: 10.1093/biolre/ioab195] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/01/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
We evaluated the influence of the hyperglycemic intrauterine environment and post-weaning consumption of a high-fat diet on the glycemia, insulin, lipid and immunological profile of rat offspring in adulthood. Female rats received citrate buffer (Control - C) or Streptozotocin (a beta cell-cytotoxic drug to induce diabetes - D) on post-natal day 5. In adulthood, these rats were mated to obtain female offspring, who were fed a standard diet (SD) or high-fat diet (HFD) from weaning to adulthood (n = 10 rats/group). OC/SD and OC/HFD represent female offspring of control mothers and received SD or HFD, respectively; OD/SD and OD/HFD represent female offspring of diabetic mothers and received SD or HFD, respectively. At adulthood, the Oral Glucose Tolerance Test (OGTT) was performed and, next, the rats were anesthetized and euthanized. Pancreas was collected and analyzed, and adipose tissue was weighted. Blood samples were collected to determine biochemical and immunological profiles. The food intake was lower in HFD-fed rats and visceral fat weight was increased in the OD/HFD group. OC/HFD, OD/SD, and OD/HFD groups presented glucose intolerance and lower insulin secretion during OGTT. An impaired pancreatic beta-cell function was shown in the adult offspring of diabetic rats, regardless of diet. Interleukin (IL)-6 and IL-10 concentrations were lower in the OD/HFD group and associated to a low-grade inflammatory condition. The fetal programming was responsible for impaired beta cell function in experimental animals. The association of maternal diabetes and post-weaning high-fat diet is responsible for greater glucose intolerance, impaired insulin secretion and immunological change.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Rafaianne Queiroz Moraes Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Thaigra Souza Soares
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Franciane Quintanilha Gallego Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Barshana Karki
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Antonio Marcus Andrade Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão - UFMA -Maranhão State, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| |
Collapse
|
19
|
Michalak A, Lach T, Cichoż-Lach H. Oxidative Stress-A Key Player in the Course of Alcohol-Related Liver Disease. J Clin Med 2021; 10:jcm10143011. [PMID: 34300175 PMCID: PMC8303854 DOI: 10.3390/jcm10143011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is known to be an inseparable factor involved in the presentation of liver disorders. Free radicals interfere with DNA, proteins, and lipids, which are crucial in liver metabolism, changing their expression and biological functions. Additionally, oxidative stress modifies the function of micro-RNAs, impairing the metabolism of hepatocytes. Free radicals have also been proven to influence the function of certain transcriptional factors and to alter the cell cycle. The pathological appearance of alcohol-related liver disease (ALD) constitutes an ideal example of harmful effects due to the redox state. Finally, ethanol-induced toxicity and overproduction of free radicals provoke irreversible changes within liver parenchyma. Understanding the underlying mechanisms associated with the redox state in the course of ALD creates new possibilities of treatment for patients. The future of hepatology may become directly dependent on the effective action against reactive oxygen species. This review summarizes current data on the redox state in the natural history of ALD, highlighting the newest reports on this topic.
Collapse
Affiliation(s)
- Agata Michalak
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
| | - Tomasz Lach
- Department of Orthopedics and Traumatology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
| | - Halina Cichoż-Lach
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
- Correspondence: ; Tel.: +48-601377656; Fax: +48-814796135
| |
Collapse
|
20
|
Liu G, Chen H, Liu H, Zhang W, Zhou J. Emerging roles of SIRT6 in human diseases and its modulators. Med Res Rev 2021; 41:1089-1137. [PMID: 33325563 PMCID: PMC7906922 DOI: 10.1002/med.21753] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
The biological functions of sirtuin 6 (SIRT6; e.g., deacetylation, defatty-acylation, and mono-ADP-ribosylation) play a pivotal role in regulating lifespan and several fundamental processes controlling aging such as DNA repair, gene expression, and telomeric maintenance. Over the past decades, the aberration of SIRT6 has been extensively observed in diverse life-threatening human diseases. In this comprehensive review, we summarize the critical roles of SIRT6 in the onset and progression of human diseases including cancer, inflammation, diabetes, steatohepatitis, arthritis, cardiovascular diseases, neurodegenerative diseases, viral infections, renal and corneal injuries, as well as the elucidation of the related signaling pathways. Moreover, we discuss the advances in the development of small molecule SIRT6 modulators including activators and inhibitors as well as their pharmacological profiles toward potential therapeutics for SIRT6-mediated diseases.
Collapse
Affiliation(s)
- Gang Liu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| |
Collapse
|
21
|
Yu B, Liu J, Cheng J, Zhang L, Song C, Tian X, Fan Y, Lv Y, Zhang X. A Static Magnetic Field Improves Iron Metabolism and Prevents High-Fat-Diet/Streptozocin-Induced Diabetes. ACTA ACUST UNITED AC 2021; 2:100077. [PMID: 34557734 PMCID: PMC8454665 DOI: 10.1016/j.xinn.2021.100077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/03/2021] [Indexed: 11/16/2022]
Abstract
Type 2 diabetes (T2D) is a metabolic disorder with high prevalence and severe complications that has recently been indicated to be treatable by a combined static magnetic field (SMF) and electric field. We systematically compared four types of SMFs and found that a downward SMF of ∼100 mT could effectively reduce the development of hyperglycemia, fatty liver, weight gain, and tissue injury in high-fat-diet (HFD)/streptozocin-induced T2D mice, but not the upward SMF. The downward SMF markedly restored the Bacteroidetes population and reversed the iron complex outer membrane receptor gene reduction in the mice gut microbiota, and reduced iron deposition in the pancreas. SMF also reduced the labile iron and reactive oxygen species level in pancreatic Min6 cells in vitro and prevented palmitate-induced Min6 cell number reduction. Therefore, this simple SMF setting could partially prevent HFD-induced T2D development and ameliorate related symptoms, which could provide a low-cost and non-invasive physical method to prevent and/or treat T2D in the future.
Collapse
Affiliation(s)
- Biao Yu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui 230036, P.R. China
| | - Juanjuan Liu
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Jing Cheng
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Lei Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Chao Song
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui 230036, P.R. China
| | - Xiaofei Tian
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, P.R. China
| | - Yixiang Fan
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui 230036, P.R. China
| | - Yue Lv
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui 230036, P.R. China
| | - Xin Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China.,Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui 230036, P.R. China.,Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
22
|
Raj S, Dsouza LA, Singh SP, Kanwal A. Sirt6 Deacetylase: A Potential Key Regulator in the Prevention of Obesity, Diabetes and Neurodegenerative Disease. Front Pharmacol 2020; 11:598326. [PMID: 33442387 PMCID: PMC7797778 DOI: 10.3389/fphar.2020.598326] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sirtuins, NAD + dependent proteins belonging to class III histone deacetylases, are involved in regulating numerous cellular processes including cellular stress, insulin resistance, inflammation, mitochondrial biogenesis, chromatin silencing, cell cycle regulation, transcription, and apoptosis. Of the seven mammalian sirtuins present in humans, Sirt6 is an essential nuclear sirtuin. Until recently, Sirt6 was thought to regulate chromatin silencing, but new research indicates its role in aging, diabetes, cardiovascular disease, lipid metabolism, neurodegenerative diseases, and cancer. Various murine models demonstrate that Sirt6 activation is beneficial in alleviating many disease conditions and increasing lifespan, showing that Sirt6 is a critical therapeutic target in the treatment of various disease conditions in humans. Sirt6 also regulates the pathogenesis of multiple diseases by acting on histone proteins and non-histone proteins. Endogenous and non-endogenous modulators regulate both activation and inhibition of Sirt6. Few Sirt6 specific non-endogenous modulators have been identified. Hence the identification of Sirt6 specific modulators may have potential therapeutic roles in the diseases described above. In this review, we describe the development of Sirt6, the role it plays in the human condition, the functional role and therapeutic importance in disease processes, and specific modulators and molecular mechanism of Sirt6 in the regulation of metabolic homeostasis, cardiovascular disease, aging, and neurodegenerative disease.
Collapse
Affiliation(s)
- Swapnil Raj
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Liston Augustine Dsouza
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Shailendra Pratap Singh
- Department of Biomedical Engineering, School of Engineering and Technology, Central University of Rajasthan, Kishangarh, India
| | - Abhinav Kanwal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| |
Collapse
|
23
|
Chen W, Wei J, Zhang L, Chen J, Li Y, Pei D, Wang N, Liu Y, Di D. Fish Roe Polypeptide Exerts Hypoglycemia Activity via Regulating Insulin Secretion Mediated by Nrf2/ERK Signaling. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10106-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Li W, Li P, Li R, Yu Z, Sun X, Ji G, Yang X, Zhu L, Zhu S. GLP1R Single-Nucleotide Polymorphisms rs3765467 and rs10305492 Affect β Cell Insulin Secretory Capacity and Apoptosis Through GLP-1. DNA Cell Biol 2020; 39:1700-1710. [PMID: 32721233 DOI: 10.1089/dna.2020.5424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The increased secretion of glucagon-like peptide-1 (GLP-1) after Roux-en-Y gastric bypass (RYGB) is regarded as the main reason for the improvement of blood glucose. However, the single-nucleotide polymorphisms (SNPs) of GLP-1 Receptor (GLP1R) impair receptor function, subsequently affecting β cell insulin secretion function, ultimately affecting the efficacy of RYGB. In this study, we revealed that two SNPs in GLP1R gene, rs3765467 and rs10305492, could significantly reduce the insulin secreted by β cells and the cyclic AMP concentration, whereas promote β cell apoptosis. Under high glucose exposure, rs3765467 and rs10305492 impaired β cell secretion of insulin and β cell viability in the same way; in other words, GLP1R rs3765467 and rs10305492 exert an effect on pancreatic β cell glucose-stimulated insulin secretion. Moreover, GLP-1 antagonist Exendin (9-39) further enhanced, whereas GLP-1 agonist Exendin-4 partially attenuated the effects of SNPs on the functions and apoptosis of β cells. In conclusion, the rs3765467 and rs10305492 SNPs in GLP1R show to exert a critical effect on regulating insulin secretory capacity of β cells and β cell mass. Through leading to the dysfunction and apoptosis of β cells, GLP1R rs3765467 and rs10305492 might also impair GLP-1 interaction with GLP1R, therefore attenuating the therapeutic effect of RYGB.
Collapse
Affiliation(s)
- Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rao Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhaomei Yu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangnian Ji
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiangwu Yang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Zhong X, Huang M, Kim HG, Zhang Y, Chowdhury K, Cai W, Saxena R, Schwabe RF, Liangpunsakul S, Dong XC. SIRT6 Protects Against Liver Fibrosis by Deacetylation and Suppression of SMAD3 in Hepatic Stellate Cells. Cell Mol Gastroenterol Hepatol 2020; 10:341-364. [PMID: 32305562 PMCID: PMC7327931 DOI: 10.1016/j.jcmgh.2020.04.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is a chronic liver disease that is manifested clinically by an increase in hepatic triglycerides, inflammation, and fibrosis. The pathogenesis of NASH remains incompletely understood. Sirtuin 6 (Sirt6), a nicotinamide adenine dinucleotide-dependent deacetylase, has been implicated in fatty liver disease; however, the underlying molecular mechanisms in the NASH pathogenesis are elusive. The aims of this study were to elucidate the role of hepatic Sirt6 in NASH. METHODS Wild-type, liver-specific Sirt6 knockout (KO), hepatic stellate cell (HSC)-specific Sirt6 knockout (HSC-KO), and Sirt6 transgenic mice were subjected to a Western diet for 4 weeks. Hepatic phenotypes were characterized and underlying mechanisms were investigated. RESULTS Remarkably, both the liver-KO and HSC-KO mice developed much worse NASH than the wild-type mice, whereas the transgenic mice were protected from the diet-induced NASH. Our cell signaling analysis showed that Sirt6 negatively regulates the transforming growth factor β-Smad family member 3 (Smad3) pathway. Biochemical analysis showed a physical interaction between Sirt6 and Smad3 in hepatic stellate cells. Moreover, our molecular data further showed that Sirt6 deacetylated Smad3 at key lysine residues K333 and K378, and attenuated its transcriptional activity induced by transforming growth factor β in hepatic stellate cells. CONCLUSIONS Our data suggest that SIRT6 plays a critical role in the protection against NASH development and it may serve as a potential therapeutic target for NASH.
Collapse
Affiliation(s)
- Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China,Department of Biochemistry and Molecular Biology
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology,Division of Gastroenterology and Hepatology, Department of Medicine
| | | | - Yang Zhang
- Department of Biochemistry and Molecular Biology
| | | | - Wenjie Cai
- Department of Biochemistry and Molecular Biology
| | | | - Robert F. Schwabe
- Institute of Human Nutrition, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Suthat Liangpunsakul
- Department of Biochemistry and Molecular Biology,Division of Gastroenterology and Hepatology, Department of Medicine,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - X. Charlie Dong
- Department of Biochemistry and Molecular Biology,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana,Correspondence Address correspondence to: X. Charlie Dong, PhD, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS-1021D, Indianapolis, Indiana 46202; fax: (317) 274-4686.
| |
Collapse
|
26
|
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
|
27
|
He Y, Yang G, Yao F, Xian Y, Wang G, Chen L, Lv X, Gao H, Zheng Z, Sun L, Wang W, Lin R. Sitagliptin inhibits vascular inflammation via the SIRT6-dependent signaling pathway. Int Immunopharmacol 2019; 75:105805. [DOI: 10.1016/j.intimp.2019.105805] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
|
28
|
Yang Y, Tian T, Wang Y, Li Z, Xing K, Tian G. SIRT6 protects vascular endothelial cells from angiotensin II-induced apoptosis and oxidative stress by promoting the activation of Nrf2/ARE signaling. Eur J Pharmacol 2019; 859:172516. [DOI: 10.1016/j.ejphar.2019.172516] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/23/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
|
29
|
Son DO, Liu W, Li X, Prud'homme GJ, Wang Q. Combined effect of GABA and glucagon-like peptide-1 receptor agonist on cytokine-induced apoptosis in pancreatic β-cell line and isolated human islets. J Diabetes 2019; 11:563-572. [PMID: 30520247 DOI: 10.1111/1753-0407.12881] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment with GABA or glucagon-like peptide-1 (GLP-1) can preserve pancreatic β-cell mass and prevent diabetes. Recently, we reported that the combination of GABA and sitagliptin (a dipeptidyl peptidase-4 inhibitor that increases endogenous GLP-1) was more effective than either agent alone in reducing drug-induced β-cell damage and promoting β-cell regeneration in mice. However, in human islets, it remains unclear whether GABA and GLP-1 exert similar effects. METHODS To investigate GABA and GLP-1 interactions, human islets or INS-1 cells were treated with GABA and/or exendin-4, a GLP-1 receptor agonist (GLP-1RA) in clinical use, and incubated with a cytokine mixture for 24 hours. Cleaved caspase-3 and annexin V binding were measured by western blot and flow cytometry analysis, respectively, to investigate effects on cytokine-induced apoptosis. RESULTS Cytokine-induced apoptosis was reduced by either GABA or exendin-4 alone. This was markedly improved by combining GABA and exendin-4, resulting in a reversal of apoptosis. The combination notably increased Akt pathway signaling. Furthermore, sirtuin-1 (SIRT1) and α-Klotho, both reported to have protective effects on β-cells, were increased. Importantly, the combination ameliorated insulin secretion by human β-cells. CONCLUSIONS The combination of GABA and a GLP-1RA exerted additive effects on β-cell survival and function, suggesting that this combination may be superior to either drug alone in the treatment of diabetes.
Collapse
Affiliation(s)
- Dong Ok Son
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Wenjuan Liu
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
- Department of Endocrinology, Huashan Hospital, Medical School, Fudan University, Shanghai, China
| | - Xiaoming Li
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Gerald J Prud'homme
- Department of Laboratory Medicine, St. Michael's Hospital, Toronto
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
- Department of Endocrinology, Huashan Hospital, Medical School, Fudan University, Shanghai, China
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
30
|
Guo T, Liu T, Sun Y, Liu X, Xiong R, Li H, Li Z, Zhang Z, Tian Z, Tian Y. Sonodynamic therapy inhibits palmitate-induced beta cell dysfunction via PINK1/Parkin-dependent mitophagy. Cell Death Dis 2019; 10:457. [PMID: 31186419 PMCID: PMC6560035 DOI: 10.1038/s41419-019-1695-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022]
Abstract
In type 2 diabetes mellitus (T2DM), the overload of glucose and lipids can promote oxidative stress and inflammatory responses and contribute to the failure of beta cells. However, therapies that can modulate the function of beta cells and thus prevent their failure have not been well explored. In this study, beta cell injury model was established with palmitic acid (PA) to simulate the lipotoxicity (high-fat diet) found in T2DM. Sonodynamic therapy (SDT), a novel physicochemical treatment, was applied to treat injured beta cells. We found that SDT had specific effects on mitochondria and induced transient large amount of mitochondrial reactive oxygen species (ROS) production in beta cells. SDT also improved the morphology and function of abnormal mitochondria, inhibited inflammatory response and reduced beta cell dysfunction. The improvement of mitochondria was mediated by PINK1/Parkin-dependent mitophagy. Additionally, SDT rescued the transcription of PINK1 mRNA which was blocked by PA treatment, thus providing abundant PINK1 for mitophagy. Moreover, SDT also increased insulin secretion from beta cells. The protective effects of SDT were abrogated when mitophagy was inhibited by cyclosporin A (CsA). In summary, SDT potently inhibits lipotoxicity-induced beta cell failure via PINK1/Parkin-dependent mitophagy, providing theoretical guidance for T2DM treatment in aspects of islet protection.
Collapse
Affiliation(s)
- Tian Guo
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Tianyang Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Yun Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Xianna Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Rongguo Xiong
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - He Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Zhitao Li
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China
| | - Zhiguo Zhang
- Laboratory of Photo- and Sono-theranostic Technologies and Condensed Matter Science and Technology Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhen Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China. .,Key Laboratory of Acoustic Photoelectric Magnetic Diagnosis and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, 150081, China.
| | - Ye Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, 150081, China. .,Key Laboratory of Acoustic Photoelectric Magnetic Diagnosis and Treatment of Cardiovascular Diseases in Heilongjiang Province, Harbin, 150081, China. .,Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
31
|
Wang C, Liu Z, Zhang P, Ma X, Che K, Wang Y. The differences in homeostasis model assessment values in type 2 diabetic patients with different lengths of history of diabetes. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:222-227. [PMID: 31066759 PMCID: PMC10522207 DOI: 10.20945/2359-3997000000134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/17/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Type 2 diabetes (T2DM) is characterized by the progressive deterioration of pancreatic islet β-cell function over time and insulin resistance. Knowing more about the differences in pancreatic islet function in T2DM patients who have had diabetes for different lengths of time can help improve therapy for T2DM. SUBJECTS AND METHODS We conducted a cross-sectional study to compare islet β-cell function and insulin resistance in T2DM patients (n = 3,254) who had had diabetes for different lengths of time and those in normal controls (n = 794) using ANOVA and LSD analysis. RESULTS We found that compared with that in normal controls, HOMA-β in T2DM patients with a history of diabetes of less than 1 year was lower (approximately 52% of that of normal controls, p = 0.003), while HOMA-IR in these patients was higher (approximately 50% of that of normal controls, p = 0.007). Compared with that in other diabetic patients, HOMA-β in patients with a history of diabetes of more than 30 years was the lowest. HOMA-IR in patients with a history of diabetes of between 20 and 30 years was lower than that in other diabetic patients (p < 0.05). CONCLUSIONS There were obvious decreases in HOMA-β and increases in HOMA-IR in T2DM patients with a history of diabetes of less than 1 year compared with those in normal controls. Therefore, early screening and intervention for T2DM might help improve islet function and delay the progression of diabetes.
Collapse
Affiliation(s)
- Chen Wang
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zaibo Liu
- Department of General SurgeryPeople’s Hospital of HaiyangYantaiShandongChinaDepartment of General Surgery, People’s Hospital of Haiyang, Yantai, Shandong, China
| | - Peng Zhang
- Department of Gastroenterology,The Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaDepartment of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaolong Ma
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kui Che
- Laboratory of Thyroid DiseaseThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaLaboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yangang Wang
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
32
|
Yuan EF, Yang Y, Cheng L, Deng X, Chen SM, Zhou X, Liu SM. Hyperglycemia affects global 5-methylcytosine and 5-hydroxymethylcytosine in blood genomic DNA through upregulation of SIRT6 and TETs. Clin Epigenetics 2019; 11:63. [PMID: 30987683 PMCID: PMC6466651 DOI: 10.1186/s13148-019-0660-y] [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: 10/14/2018] [Accepted: 03/31/2019] [Indexed: 01/20/2023] Open
Abstract
Background Accumulating evidence suggests that epigenetic changes play key roles in the pathogenesis of type 2 diabetes mellitus (T2DM). However, the dynamic regulation of 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) in diabetic peripheral blood DNA remains to be elucidated. Results We collected fasting blood samples (104 patients and 108 healthy controls) and glucose-stimulated blood samples at different time points (11 patients and 5 healthy controls underwent oral glucose tolerance test (OGTT)), as well as blood samples from six couples of diabetic and control rats. A HPLC-MS/MS system was used for quantifying global 5mC and 5hmC in genomic DNA from white blood cells (WBCs), and qPCR was performed for detecting mRNA expression of SIRT6 and TETs. We found that global 5mC decreased, while global 5hmC increased in both patients and diabetic rats, with lower 5mC being a risk factor of T2DM (OR = 0.524, 95%CI 0.402–0.683, p = 1.64 × 10−6). The OGTT data from patients showed that 5mC declined within 1 h and then returned to the fasting status at 2 h, while 5hmC rose from 0.5 h to 3 h with increasing glucose. However, the similar patterns were not found in the controls. The mRNA expression of TET2, TET3, and SIRT6 was upregulated in patients (p = 0.012, p = 0.026, and p = 0.035, respectively). The similar results were observed in diabetic OGTT and rats. Correlation analysis indicated that SIRT6 was positively correlated with TET2 in humans (r = 0.277, p < 0.001) and rats (r = 0.942, p < 0.001), in addition to a correlation between glucose and SIRT6 (r = 0.162, p = 0.045) and TET2 (r = 0.174, p = 0.036). Conclusions Hyperglycemia appeared to promote the mRNA expression of SIRT6 and TETs, which in turn might cause the dynamic changes of 5mC and 5hmC in WBCs from T2DM patients. Electronic supplementary material The online version of this article (10.1186/s13148-019-0660-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Er-Feng Yuan
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China.,Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Ying Yang
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Lin Cheng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, Hubei Province, China
| | - Xujing Deng
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Shao-Min Chen
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Xin Zhou
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Song-Mei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
33
|
Yu J, Sun W, Song Y, Liu J, Xue F, Gong K, Yang X, Kang Q. SIRT6 protects retinal ganglion cells against hydrogen peroxide-induced apoptosis and oxidative stress by promoting Nrf2/ARE signaling via inhibition of Bach1. Chem Biol Interact 2019; 300:151-158. [DOI: 10.1016/j.cbi.2019.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/12/2019] [Accepted: 01/16/2019] [Indexed: 01/19/2023]
|
34
|
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
|
35
|
Bae JS, Noh SJ, Kim KM, Park SH, Hussein UK, Park HS, Park BH, Ha SH, Lee H, Chung MJ, Moon WS, Cho DH, Jang KY. SIRT6 Is Involved in the Progression of Ovarian Carcinomas via β-Catenin-Mediated Epithelial to Mesenchymal Transition. Front Oncol 2018; 8:538. [PMID: 30524965 PMCID: PMC6256124 DOI: 10.3389/fonc.2018.00538] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
SIRT6 is involved in various cellular signaling pathways including those involved in tumorigenesis in association with β-catenin. However, the role of SIRT6 in tumorigenesis has been controversially reported and the studies on the role of SIRT6 in ovarian cancers is limited. In this study, we evaluated the expression and roles of SIRT6 in conjunction with the expression of active β-catenin in 104 human ovarian carcinomas and ovarian cancer cells. In human ovarian carcinomas, the expressions of SIRT6 and active β-catenin were associated with higher tumor stage, higher histologic grade, and platinum-resistance. Moreover, nuclear expression of SIRT6 (104 ovarian carcinomas; P = 0.010, 63 high-grade serous carcinomas; P = 0.040), and activated β-catenin (104 ovarian carcinomas; P = 0.013, 63 high-grade serous carcinomas; P = 0.005) were independent indicators of shorter overall survival of ovarian carcinoma patients in multivariate analysis. In OVCAR3 and OVCAR5 ovarian cancer cells, knock-down of SIRT6 significantly inhibited the migration and invasion of cells, but did not inhibit the proliferation of cells. SIRT6-mediated invasiveness of ovarian cancer cells was associated with the expression of epithelial-to-mesenchymal transition-related signaling molecules such as snail, vimentin, N-cadherin, E-cadherin, and activated β-catenin. Especially, SIRT6-mediated increase of invasiveness and activation of epithelial-to-mesenchymal transition signaling was attenuated by knock-down of β-catenin. In conclusion, this study suggests that SIRT6-β-catenin signaling is involved in the epithelial-to-mesenchymal transition of ovarian cancer cells, and the expression of SIRT6 and active β-catenin might be used as indicators of poor prognosis of ovarian carcinoma patients. In addition, our results suggest that SIRT6-β-catenin signaling might be a new therapeutic target of ovarian carcinomas.
Collapse
Affiliation(s)
- Jun Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea
| | - Sang Jae Noh
- Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea.,Department of Forensic Medicine, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea
| | - Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Usama Khamis Hussein
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Ho Sung Park
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea
| | - Byung-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea
| | - Sang Hoon Ha
- Division of Biotechnology, Chonbuk National University, Iksan, South Korea
| | - Ho Lee
- Department of Forensic Medicine, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea
| | - Dong Hyu Cho
- Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea.,Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Chonbuk National University, Jeonju, South Korea.,Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, South Korea.,Research Institute for Clinical Medicine, Chonbuk National University, Jeonju, South Korea.,Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
36
|
Abstract
Sirt6 is one of the sirtuin family members, a kind of NAD+-dependent histone deacetylase and ADP-ribose transferase enzyme. It has an important role in physiological and pathological processes, regulating aging, cancer, obesity, insulin resistance, inflammation, and energy metabolism. Recent studies have suggested that reduced Sirt6 action is related to obesity and diabetes. Aging and overnutrition, two major risk factors for obesity and diabetes, lead to decreased Sirt6 level and function, which results in abnormal glucose and lipid metabolism. Whole-body ablation of Sirt6 in mice results in severe hypoglycemia. Sirt6 deficiency leads to liver steatosis and promotes diet-induced obesity and insulin resistance. Sirt6 has a protective effect on obesity and diabetes. This review surveys evidence for an emerging role of Sirt6 as a regulator of metabolism in mammals and summarizes its major functions in obesity and diabetes.
Collapse
Affiliation(s)
- Jiangying Kuang
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Lei Chen
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Tang
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Zhang
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Li
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhan He
- State Key Laboratory of Biotherapy, Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Song J, Yang B, Jia X, Li M, Tan W, Ma S, Shi X, Feng L. Distinctive Roles of Sirtuins on Diabetes, Protective or Detrimental? Front Endocrinol (Lausanne) 2018; 9:724. [PMID: 30559718 PMCID: PMC6284472 DOI: 10.3389/fendo.2018.00724] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022] Open
Abstract
Dysregulation of metabolic pathways leads to type 2 diabetes, characteristic of high glucose concentration caused by insulin resistance. The histone deacetylases sirtuins exhibit remarkable enzymatic activities. Accumulating evidence indicates that sirtuins can be pharmacologically activated to ameliorate diabetes. Here, we evaluated different roles of sirtuins (SIRT1-SIRT7) in diabetes progression and described their involvement in metabolic pathways of skeletal muscle, adipose tissue and liver. The nuclear sirtuins, SIRT1, SIRT6, and SIRT7, regulate the activity of key transcription factors and cofactors in almost all tissues with the cellular responses to energy demands. The mitochondrial sirtuins, SIRT3, SIRT4, and SIRT5, regulate the activity of mitochondrial enzymes in response to fasting and calorie restriction. Moreover, genetic polymorphisms of SIRT1 and SIRT2 have been reported to associate with diabetes development. It's worth noting that SIRT1, SIRT2, SIRT3, and SIRT6 are positive regulators of insulin resistance in most cases. In the opposite, SIRT4 and SIRT7 inhibit insulin secretion and fatty acid oxidation. Identification of SIRT1 activators for diabetes has gained wide attention, such as metformin, resveratrol, and resveratrol derivatives. Randomized, prospective, and large-scale clinical trials are warrant to uncover the responsibilities of SIRTs modulators on diabetes progress.
Collapse
Affiliation(s)
- Jie Song
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mingyu Li
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Tan
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shitang Ma
- Life and Health college, Anhui Science and Technology University, Fengyang, China
| | - Xinhong Shi
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- *Correspondence: Liang Feng
| |
Collapse
|
38
|
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
|
39
|
Jing T, Ya-Shu K, Xue-Jun W, Han-Jing H, Yan L, Yi-An Y, Fei C, Xue-Bo L. Sirt6 mRNA-incorporated endothelial microparticles (EMPs) attenuates DM patient-derived EMP-induced endothelial dysfunction. Oncotarget 2017; 8:114300-114313. [PMID: 29371988 PMCID: PMC5768405 DOI: 10.18632/oncotarget.23259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/03/2017] [Indexed: 11/25/2022] Open
Abstract
Background Endothelial microparticles (EMPs) are small vesicles released by endothelial cells (ECs); they are considered biomarkers for endothelial dysfunction and therapeutic targets in diabetes-related vascular disease. Sirtuins have also been shown to play important roles in diabetes by regulating endothelial dysfunction. However, the effect of sirtuin-incorporated EMPs on their parental ECs remains unknown. Aim The present study aims to investigate the diagnostic value of EMPs in diabetes and detect the protective effects of sirtuin 6 (Sirt6) mRNA -incorporated EMPs on endothelial dysfunction. Methods EMPs were prepared from cultured HUVECs and venous blood from patients with diabetes (n=10) and from healthy volunteers (n=6) after sequential centrifugation. Adv-Sirt6 or Sirt6 siRNA was used to alter Sirt6 expression. EC angiogenesis, inflammatory phenotypes, nitric oxide (NO) formation and eNOS phosphorylation were used to evaluate endothelial dysfunction. Results The levels of EMPs in diabetic patients and high glucose-cultured HUVECs are high, whereas Sirt6 expression in plasma and EMPs is low. EMPs generated from diabetic patients or high glucose-cultured HUVECs increase inflammatory chemokine release and blunt EC angiogenesis. Furthermore, EMPs enriched with Sirt6 mRNA induces EC angiogenesis, increases eNOS phosphorylation and impedes inflammatory chemokine release. Inhibition of Sirt6 mRNA expression in EMPs by siRNA hinders angiogenesis and eNOS phosphorylation but increases cellular inflammation. Conclusion The Sirt6 mRNA-carrying EMPs may ameliorate endothelial dysfunction in diabetic patients.
Collapse
Affiliation(s)
- Tong Jing
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Kuang Ya-Shu
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Wang Xue-Jun
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Hou Han-Jing
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Lai Yan
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Yao Yi-An
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Chen Fei
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Liu Xue-Bo
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
40
|
Zorrilla-Zubilete MA, Yeste A, Quintana FJ, Toiber D, Mostoslavsky R, Silberman DM. Epigenetic control of early neurodegenerative events in diabetic retinopathy by the histone deacetylase SIRT6. J Neurochem 2017; 144:128-138. [DOI: 10.1111/jnc.14243] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 01/03/2023]
Affiliation(s)
- María A. Zorrilla-Zubilete
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO-CONICET); 1ª Cátedra de Farmacología; Facultad de Medicina; UBA; Buenos Aires Argentina
| | - Ada Yeste
- Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts USA
| | - Francisco J. Quintana
- Brigham and Women's Hospital; Harvard Medical School; Boston Massachusetts USA
- Broad Institute of MIT and Harvard; Cambridge Massachusetts USA
| | - Debra Toiber
- Department of Life Sciences; The Zlotowski Center for Neuroscience; Ben-Gurion University of the Negev; Beer-Sheva Israel
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center; Harvard Medical School; Boston Massachusetts USA
- Broad Institute of MIT and Harvard; Cambridge Massachusetts USA
| | - Dafne M. Silberman
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO-CONICET); 1ª Cátedra de Farmacología; Facultad de Medicina; UBA; Buenos Aires Argentina
| |
Collapse
|
41
|
Lin N, Niu Y, Zhang W, Li X, Yang Z, Su Q. microRNA-802 is involved in palmitate-induced damage to pancreatic β cells through repression of sirtuin 6. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:11300-11307. [PMID: 31966484 PMCID: PMC6965834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/30/2017] [Indexed: 06/10/2023]
Abstract
Free fatty acid (FFA)-induced pancreatic β-cell loss is implicated in the pathogenesis of type 2 diabetes mellitus (T2DM). It has been documented that circulating microRNA (miR)-802 levels are significantly greater in T2DM patients than in healthy subjects. However, the role of miR-802 in FFA-induced damage to β cells is still unclear. In the present study, we measured the expression of miR-802 in the INS-1 rat insulinoma cell line after palmitate treatment for 48 h. Gain- and loss-of-function studies were conducted to determine the function of miR-802 in palmitate-induced apoptosis and reactive oxygen species (ROS) production. The target gene(s) of miR-802 was functionally characterized. Compared to control cells, palmitate treatment caused a time- and concentration-dependent induction of miR-802 in INS-1 cells. Knockdown of miR-802 significantly blocked palmitate-induced apoptosis and attenuated ROS formation. Moreover, miR-802 downregulation prevented the reduction of prosurvival proteins Mcl-1 and Bcl-xL by palmitate. In contrast, ectopic expression of miR-802 stimulated apoptosis and ROS generation in INS-1 cells. Sirtuin 6 (SIRT6) was identified to be a direct target gene of miR-802. Overexpression of miR-802 suppressed the expression of SIRT6. Enforced expression of SIRT6 abolished the induction of apoptosis and ROS production by miR-802. Taken together, miR-802 is required for palmitate-induced damage to β cells by targeting SIRT6 and represents a potential therapeutic target for T2DM.
Collapse
Affiliation(s)
- Ning Lin
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yixin Niu
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Weiwei Zhang
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Xiaoyong Li
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Zhen Yang
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
42
|
Feng XT, Duan HM, Li SL. Protective role of Pollen Typhae total flavone against the palmitic acid-induced impairment of glucose-stimulated insulin secretion involving GPR40 signaling in INS-1 cells. Int J Mol Med 2017; 40:922-930. [DOI: 10.3892/ijmm.2017.3070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/05/2017] [Indexed: 11/05/2022] Open
|
43
|
Xiong X, Zhang C, Zhang Y, Fan R, Qian X, Dong XC. Fabp4-Cre-mediated Sirt6 deletion impairs adipose tissue function and metabolic homeostasis in mice. J Endocrinol 2017; 233:307-314. [PMID: 28385723 PMCID: PMC5502685 DOI: 10.1530/joe-17-0033] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/06/2017] [Indexed: 12/24/2022]
Abstract
SIRT6 is a member of sirtuin family of deacetylases involved in diverse processes including genome stability, metabolic homeostasis and anti-inflammation. However, its function in the adipose tissue is not well understood. To examine the metabolic function of SIRT6 in the adipose tissue, we generated two mouse models that are deficient in Sirt6 using the Cre-lox approach. Two commonly used Cre lines that are driven by either the mouse Fabp4 or Adipoq gene promoter were chosen for this study. The Sirt6-knockout mice generated by the Fabp4-Cre line (Sirt6f/f:Fabp4-Cre) had a significant increase in both body weight and fat mass and exhibited glucose intolerance and insulin resistance as compared with the control wild-type mice. At the molecular levels, the Sirt6f/f :Fabp4-Cre-knockout mice had increased expression of inflammatory genes including F4/80, TNFα, IL-6 and MCP-1 in both white and brown adipose tissues. Moreover, the knockout mice showed decreased expression of the adiponectin gene in the white adipose tissue and UCP1 in the brown adipose tissue, respectively. In contrast, the Sirt6 knockout mice generated by the Adipoq-Cre line (Sirt6f/f :Adipoq-Cre) only had modest insulin resistance. In conclusion, our data suggest that the function of SIRT6 in the Fabp4-Cre-expressing cells in addition to mature adipocytes plays a critical role in body weight maintenance and metabolic homeostasis.
Collapse
Affiliation(s)
- Xiwen Xiong
- Department of Forensic MedicineXinxiang Medical University, Xinxiang, Henan, China
| | - Cuicui Zhang
- School of Basic Medical SciencesXinxiang Medical University, Xinxiang, Henan, China
| | - Yang Zhang
- Department of Biochemistry and Molecular BiologyIndiana University School of Medicine, Indianapolis, USA
| | - Rui Fan
- School of Basic Medical SciencesXinxiang Medical University, Xinxiang, Henan, China
| | - Xinlai Qian
- School of Basic Medical SciencesXinxiang Medical University, Xinxiang, Henan, China
| | - X Charlie Dong
- Department of Biochemistry and Molecular BiologyIndiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
44
|
Deota S, Shukla N, Kolthur-Seetharam U. Spatio-Temporal Control of Cellular and Organismal Physiology by Sirtuins. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|