1
|
Miranda ER, Varshney P, Mazo CE, Shadiow J, Ludlow AT, Haus JM. Loss of NAMPT and SIRT2 but not SIRT1 attenuate GLO1 expression and activity in human skeletal muscle. Redox Biol 2024; 75:103300. [PMID: 39142179 PMCID: PMC11367650 DOI: 10.1016/j.redox.2024.103300] [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: 07/18/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
Glyoxalase I (GLO1) is the primary enzyme for detoxification of the reactive dicarbonyl methylglyoxal (MG). Loss of GLO1 promotes accumulation of MG resulting in a recapitulation of diabetic phenotypes. We previously demonstrated attenuated GLO1 protein in skeletal muscle from individuals with type 2 diabetes (T2D). However, whether GLO1 attenuation occurs prior to T2D and the mechanisms regulating GLO1 abundance in skeletal muscle are unknown. GLO1 expression and activity were determined in skeletal muscle tissue biopsies from 15 lean healthy individuals (LH, BMI: 22.4 ± 0.7) and 5 individuals with obesity (OB, BMI: 32.4 ± 1.3). GLO1 protein was attenuated by 26 ± 0.3 % in OB compared to LH skeletal muscle (p = 0.019). Similar reductions for GLO1 activity were observed (p = 0.102). NRF2 and Keap1 expression were equivocal between groups despite a 2-fold elevation in GLO1 transcripts in OB skeletal muscle (p = 0.008). GLO1 knock-down (KD) in human immortalized myotubes promoted downregulation of muscle contraction and organization proteins indicating the importance of GLO1 expression for skeletal muscle function. SIRT1 KD had no effect on GLO1 protein or activity whereas, SIRT2 KD attenuated GLO1 protein by 28 ± 0.29 % (p < 0.0001) and GLO1 activity by 42 ± 0.12 % (p = 0.0150). KD of NAMPT also resulted in attenuation of GLO1 protein (28 ± 0.069 %, p = 0.003), activity (67 ± 0.09 %, p = 0.011) and transcripts (50 ± 0.13 %, p = 0.049). Neither the provision of the NAD+ precursors NR nor NMN were able to prevent this attenuation in GLO1 protein. However, NR did augment GLO1 specific activity (p = 0.022 vs NAMPT KD). These perturbations did not alter GLO1 acetylation status. SIRT1, SIRT2 and NAMPT protein levels were all equivocal in skeletal muscle tissue biopsies from individuals with obesity and lean individuals. These data implicate NAD+-dependent regulation of GLO1 in skeletal muscle independent of altered GLO1 acetylation and provide rationale for exploring NR supplementation to rescue attenuated GLO1 abundance and activity in conditions such as obesity.
Collapse
Affiliation(s)
- Edwin R Miranda
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Pallavi Varshney
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Corey E Mazo
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Andrew T Ludlow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Jacob M Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Wu LE, Fiveash CE, Bentley NL, Kang M, Govindaraju H, Barbour JA, Wilkins BP, Hancock SE, Madawala R, Das A, Massudi H, Li C, Kim L, Wong ASA, Marinova MB, Sultani G, Das A, Youngson NA, Le Couteur DG, Sinclair DA, Turner N. SIRT2 transgenic over-expression does not impact lifespan in mice. Aging Cell 2023; 22:e14027. [PMID: 38009412 PMCID: PMC10726910 DOI: 10.1111/acel.14027] [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: 04/06/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 11/28/2023] Open
Abstract
The NAD+ -dependent deacylase family of sirtuin enzymes have been implicated in biological ageing, late-life health and overall lifespan, though of these members, a role for sirtuin-2 (SIRT2) is less clear. Transgenic overexpression of SIRT2 in the BubR1 hypomorph model of progeria can rescue many aspects of health and increase overall lifespan, due to a specific interaction between SIRT2 and BubR1 that improves the stability of this protein. It is less clear whether SIRT2 is relevant to biological ageing outside of a model where BubR1 is under-expressed. Here, we sought to test whether SIRT2 over-expression would impact the overall health and lifespan of mice on a nonprogeroid, wild-type background. While we previously found that SIRT2 transgenic overexpression prolonged female fertility, here, we did not observe any additional impact on health or lifespan, which was measured in both male and female mice on standard chow diets, and in males challenged with a high-fat diet. At the biochemical level, NMR studies revealed an increase in total levels of a number of metabolites in the brain of SIRT2-Tg animals, pointing to a potential impact in cell composition; however, this did not translate into functional differences. Overall, we conclude that strategies to enhance SIRT2 protein levels may not lead to increased longevity.
Collapse
Affiliation(s)
- Lindsay E. Wu
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Corrine E. Fiveash
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | | | - Myung‐Jin Kang
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Hemna Govindaraju
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
- Victor Chang Cardiac Research InstituteDarlinghurstNew South WalesAustralia
| | - Jayne A. Barbour
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Brendan P. Wilkins
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Sarah E. Hancock
- Victor Chang Cardiac Research InstituteDarlinghurstNew South WalesAustralia
| | - Romanthi Madawala
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Abhijit Das
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
- School of PsychologyUNSW SydneyKensingtonNew South WalesAustralia
| | - Hassina Massudi
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Catherine Li
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Lynn‐Jee Kim
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Ashley S. A. Wong
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Maria B. Marinova
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Ghazal Sultani
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Abhirup Das
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - Neil A. Youngson
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
| | - David G. Le Couteur
- ANZAC Medical Research InstituteConcordNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
| | - David A. Sinclair
- Department of Genetics, Blavatnik InstitutePaul F. Glenn Center for Biology of Aging Research, Harvard Medical SchoolBostonMassachusettsUnited States
| | - Nigel Turner
- School of Biomedical SciencesUNSW SydneyKensingtonNew South WalesAustralia
- Victor Chang Cardiac Research InstituteDarlinghurstNew South WalesAustralia
| |
Collapse
|
3
|
Molinari S, Imbriano C, Moresi V, Renzini A, Belluti S, Lozanoska-Ochser B, Gigli G, Cedola A. Histone deacetylase functions and therapeutic implications for adult skeletal muscle metabolism. Front Mol Biosci 2023; 10:1130183. [PMID: 37006625 PMCID: PMC10050567 DOI: 10.3389/fmolb.2023.1130183] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Skeletal muscle is a highly adaptive organ that sustains continuous metabolic changes in response to different functional demands. Healthy skeletal muscle can adjust fuel utilization to the intensity of muscle activity, the availability of nutrients and the intrinsic characteristics of muscle fibers. This property is defined as metabolic flexibility. Importantly, impaired metabolic flexibility has been associated with, and likely contributes to the onset and progression of numerous pathologies, including sarcopenia and type 2 diabetes. Numerous studies involving genetic and pharmacological manipulations of histone deacetylases (HDACs) in vitro and in vivo have elucidated their multiple functions in regulating adult skeletal muscle metabolism and adaptation. Here, we briefly review HDAC classification and skeletal muscle metabolism in physiological conditions and upon metabolic stimuli. We then discuss HDAC functions in regulating skeletal muscle metabolism at baseline and following exercise. Finally, we give an overview of the literature regarding the activity of HDACs in skeletal muscle aging and their potential as therapeutic targets for the treatment of insulin resistance.
Collapse
Affiliation(s)
- Susanna Molinari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Viviana Moresi
- Institute of Nanotechnology, Department of Physics, National Research Council (CNR-NANOTEC), Sapienza University of Rome, Rome, Italy
- *Correspondence: Viviana Moresi,
| | - Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Lecce, Italy
| | - Alessia Cedola
- Institute of Nanotechnology, Department of Physics, National Research Council (CNR-NANOTEC), Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Sandonà M, Cavioli G, Renzini A, Cedola A, Gigli G, Coletti D, McKinsey TA, Moresi V, Saccone V. Histone Deacetylases: Molecular Mechanisms and Therapeutic Implications for Muscular Dystrophies. Int J Mol Sci 2023; 24:4306. [PMID: 36901738 PMCID: PMC10002075 DOI: 10.3390/ijms24054306] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Histone deacetylases (HDACs) are enzymes that regulate the deacetylation of numerous histone and non-histone proteins, thereby affecting a wide range of cellular processes. Deregulation of HDAC expression or activity is often associated with several pathologies, suggesting potential for targeting these enzymes for therapeutic purposes. For example, HDAC expression and activity are higher in dystrophic skeletal muscles. General pharmacological blockade of HDACs, by means of pan-HDAC inhibitors (HDACi), ameliorates both muscle histological abnormalities and function in preclinical studies. A phase II clinical trial of the pan-HDACi givinostat revealed partial histological improvement and functional recovery of Duchenne Muscular Dystrophy (DMD) muscles; results of an ongoing phase III clinical trial that is assessing the long-term safety and efficacy of givinostat in DMD patients are pending. Here we review the current knowledge about the HDAC functions in distinct cell types in skeletal muscle, identified by genetic and -omic approaches. We describe the signaling events that are affected by HDACs and contribute to muscular dystrophy pathogenesis by altering muscle regeneration and/or repair processes. Reviewing recent insights into HDAC cellular functions in dystrophic muscles provides new perspectives for the development of more effective therapeutic approaches based on drugs that target these critical enzymes.
Collapse
Affiliation(s)
| | - Giorgia Cavioli
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessia Cedola
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), 73100 Lecce, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
- CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Aging B2A, Sorbonne Université, 75005 Paris, France
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Viviana Moresi
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Valentina Saccone
- IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
5
|
Yadav Y, Dey CS. PP2Cα aggravates neuronal insulin resistance leading to AD-like phenotype in vitro. Biochem Biophys Res Commun 2023; 644:49-54. [PMID: 36630734 DOI: 10.1016/j.bbrc.2023.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/13/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Neuronal insulin resistance is a major risk for development of Alzheimer's Disease (AD). Studies already reported few kinases participating in neuronal insulin signaling connected with progression of AD pathogenesis, yet complete information is missing. α isoform of Protein Phosphatase-2C (PP2C) is a Ser/Thr phosphatase, only known in 3T3-L1 adipocytes as a positive regulator of insulin signaling. However, many aspects of its function in neuronal insulin signaling and insulin resistance are unidentified. Recently, we reported that PP2Cα positively regulates neuronal glucose uptake possibly by a mechanism of dephosphorylation of IRS-1 at Ser522 and by inactivating AMPK, exacerbating hyperinsulinemia mediated neuronal insulin resistance. Since PP2Cα affected neuronal insulin signaling and AD is connected to neuronal insulin resistance, in the present study, we studied the role of PP2Cα in regulating activities of both isoforms of GSK3α and GSK3β (one of the leading kinases for AD progression). The results led us to test the role of PP2Cα on AD hallmarks. Silencing of PP2Cα caused hyperphosphorylation of a potential kinase Tau, leading to NFT formation and increased Aβ deposition. Our study thereby demonstrates escalation of hyperinsulinemia mediated neuronal insulin resistance leading to AD-like pathogenesis by PP2Cα in vitro and hints a novel molecule, PP2Cα, linking AD pathogenesis.
Collapse
Affiliation(s)
- Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 10016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 10016, India.
| |
Collapse
|
6
|
Sharma M, Dey CS. PHLPP isoforms differentially regulate Akt isoforms and AS160 affecting neuronal insulin signaling and insulin resistance via Scribble. Cell Commun Signal 2022; 20:179. [PMID: 36376971 PMCID: PMC9664818 DOI: 10.1186/s12964-022-00987-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/08/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The aim of the present study was to determine the role of individual PHLPP isoforms in insulin signaling and insulin resistance in neuronal cells. METHODS PHLPP isoforms were either silenced or overexpressed individually, and the effects were observed on individual Akt isoforms, AS160 and on neuronal glucose uptake, under insulin sensitive and resistant conditions. To determine PHLPP regulation itself, we tested effect of scaffold protein, Scribble, on PHLPP isoforms and neuronal glucose uptake. RESULTS We observed elevated expression of both PHLPP1 and PHLPP2 in insulin resistant neuronal cells (Neuro-2A, mouse neuroblastoma; SHSY-5Y, human neuroblastoma) as well as in the whole brain lysates of high-fat-diet mediated diabetic mice. In insulin sensitive condition, PHLPP isoforms differentially affected activation of all Akt isoforms, wherein PHLPP1 regulated serine phosphorylation of Akt2 and Akt3, while PHLPP2 regulated Akt1 and Akt3. This PHLPP mediated Akt isoform specific regulation activated AS160 affecting glucose uptake. Under insulin resistant condition, a similar trend of results were observed in Akt isoforms, AS160 and glucose uptake. Over-expressed PHLPP isoforms combined with elevated endogenous expression under insulin resistant condition drastically affected downstream signaling, reducing neuronal glucose uptake. No compensation was observed amongst PHLPP isoforms under all conditions tested, indicating independent roles and pointing towards possible scaffolding interactions behind isoform specificity. Silencing of Scribble, a scaffolding protein known to interact with PHLPP, affected cellular localization of both PHLPP1 and PHLPP2, and caused increase in glucose uptake. CONCLUSIONS PHLPP isoforms play independent roles via Scribble in regulating Akt isoforms differentially, affecting AS160 and neuronal glucose uptake. Video abstract.
Collapse
Affiliation(s)
- Medha Sharma
- grid.417967.a0000 0004 0558 8755Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016 India
| | - Chinmoy Sankar Dey
- grid.417967.a0000 0004 0558 8755Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016 India
| |
Collapse
|
7
|
Zhou C, Jung CG, Kim MJ, Watanabe A, Abdelhamid M, Taslima F, Michikawa M. Insulin Deficiency Increases Sirt2 Level in Streptozotocin-Treated Alzheimer's Disease-Like Mouse Model: Increased Sirt2 Induces Tau Phosphorylation Through ERK Activation. Mol Neurobiol 2022; 59:5408-5425. [PMID: 35701718 PMCID: PMC9395464 DOI: 10.1007/s12035-022-02918-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/07/2022] [Indexed: 11/11/2022]
Abstract
Accumulating evidence suggests that insulin deficiency is a risk factor for Alzheimer's disease (AD); however, the underlying molecular mechanisms are not completely understood. Here, we investigated the effects of insulin deficiency on AD-like pathologies using an insulin-deficient amyloid-β (Aβ) precursor protein (APP) transgenic mouse model (Tg2576 mice). Female Tg2576 mice were injected intraperitoneally with streptozotocin (STZ) to induce insulin deficiency, and their body weights, serum glucose levels, and serum insulin levels were evaluated. STZ-treated mice showed exacerbated Aβ accumulation, tau hyperphosphorylation, glial activation, neuroinflammation, and increased Sirt2 protein levels in the brain, as determined by two-dimensional gel electrophoresis (2-DE) coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) and Western blotting. Furthermore, our in vitro experiments revealed that insulin depletion or interleukin-6 treatment increased Sirt2 protein levels in both Neuro2a and Neuro2a-P301L cells. The overexpression of Sirt2 in these cells induced tau hyperphosphorylation through extracellular signal-regulated kinase (ERK) activation. Conversely, Sirt2 knockdown reversed tau hyperphosphorylation in these cells. We showed for the first time that Sirt2 is upregulated in the brains of STZ-treated Tg2576 mice and is involved in tau phosphorylation through ERK activation. Our findings suggest that Sirt2 is a promising therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Chunyu Zhou
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601 Japan
| | - Cha-Gyun Jung
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601 Japan
| | - Mi-Jeong Kim
- Department of Food & Biotechnology, Korea University, Sejong, 30019 South Korea
| | - Atsushi Watanabe
- Laboratory of Research Advancement, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511 Japan
| | - Mona Abdelhamid
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601 Japan
| | - Ferdous Taslima
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601 Japan
| | - Makoto Michikawa
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601 Japan
| |
Collapse
|
8
|
Abbotto E, Scarano N, Piacente F, Millo E, Cichero E, Bruzzone S. Virtual Screening in the Identification of Sirtuins’ Activity Modulators. Molecules 2022; 27:molecules27175641. [PMID: 36080416 PMCID: PMC9457788 DOI: 10.3390/molecules27175641] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Sirtuins are NAD+-dependent deac(et)ylases with different subcellular localization. The sirtuins’ family is composed of seven members, named SIRT-1 to SIRT-7. Their substrates include histones and also an increasing number of different proteins. Sirtuins regulate a wide range of different processes, ranging from transcription to metabolism to genome stability. Thus, their dysregulation has been related to the pathogenesis of different diseases. In this review, we discussed the pharmacological approaches based on sirtuins’ modulators (both inhibitors and activators) that have been attempted in in vitro and/or in in vivo experimental settings, to highlight the therapeutic potential of targeting one/more specific sirtuin isoform(s) in cancer, neurodegenerative disorders and type 2 diabetes. Extensive research has already been performed to identify SIRT-1 and -2 modulators, while compounds targeting the other sirtuins have been less studied so far. Beside sections dedicated to each sirtuin, in the present review we also included sections dedicated to pan-sirtuins’ and to parasitic sirtuins’ modulators. A special focus is dedicated to the sirtuins’ modulators identified by the use of virtual screening.
Collapse
Affiliation(s)
- Elena Abbotto
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
- Correspondence:
| |
Collapse
|
9
|
Lee EJ, Lee MM, Park S, Jeong KS. Sirt2 positively regulates muscle regeneration after Notexin-induced muscle injury. Exp Mol Pathol 2022; 127:104798. [DOI: 10.1016/j.yexmp.2022.104798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/01/2022] [Accepted: 05/24/2022] [Indexed: 01/04/2023]
|
10
|
Santos HO, Genario R, Tinsley GM, Ribeiro P, Carteri RB, Coelho-Ravagnani CDF, Mota JF. A scoping review of intermittent fasting, chronobiology, and metabolism. Am J Clin Nutr 2022; 115:991-1004. [PMID: 34978321 DOI: 10.1093/ajcn/nqab433] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/29/2021] [Indexed: 12/19/2022] Open
Abstract
Chronobiology plays a crucial role in modulating many physiologic systems in which there is nutritional synergism with meal timing. Given that intermittent fasting (IF) has grown as a flexible dietary method consisting of delayed or early eating windows, this scoping review addresses the effects of IF protocols on metabolism as they relate to clinical nutrition and the circadian system. Although nocturnal habits are associated with circadian misalignments and impaired cardiometabolic profile-and nutritional physiology is better orchestrated during the day-most findings are based on animal experiments or human studies with observational designs or acute meal tests. Well-controlled randomized clinical trials employing IF protocols of delayed or early eating windows have sometimes demonstrated clinical benefits, such as improved glycemic and lipid profiles, as well as weight loss. However, IF does not appear to be more effective than traditional diets at the group level, and its effects largely depend on energy restriction. Thus, efforts must be made to identify patient biological rhythms, preferences, routines, and medical conditions before individual dietary prescription in clinical practice.
Collapse
Affiliation(s)
- Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Rafael Genario
- School of Medicine, University of São Paulo (USP), São Paulo, Brazil
| | - Grant M Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, TX, USA
| | - Poliana Ribeiro
- Institute of Genetics and Biochemistry, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Randhall B Carteri
- Methodist University Center-Porto Alegre Institute, Porto Alegre, Brazil
| | | | - João F Mota
- Clinical and Sports Nutrition Research Laboratory, Faculty of Nutrition, Goiás Federal University, Goiânia, GO, Brazil
| |
Collapse
|
11
|
Manglani K, Dey CS. CDK5 inhibition improves glucose uptake in insulin-resistant neuronal cells via ERK1/2 pathway. Cell Biol Int 2021; 46:488-497. [PMID: 34865281 DOI: 10.1002/cbin.11735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/30/2021] [Accepted: 11/28/2021] [Indexed: 01/07/2023]
Abstract
Role of CDK5 and its inhibition in various neuronal processes and functions are well established. However, role of CDK5 and its inhibition in neuronal insulin-signaling and-resistance is not yet explored. In the present study, we investigated the effect of CDK5 inhibition in neuronal insulin signaling, specifically insulin-stimulated glucose uptake. CDK5 expression in neuro-2a cells was increased under insulin-resistant state, developed by chronic treatment of insulin, confirming the crucial role of CDK5 in insulin resistance in neuronal cells. However, whether increased expression of CDK5 in hyperinsulinemia-mediated insulin-resistant conditions is a cause or a consequence, is still an unanswered question. We showed that CDK5 inhibition did not affect basal insulin signaling; however, insulin-stimulated glucose uptake enhanced in insulin-resistant cells. Moreover, CDK5 inhibition could improve glucose uptake, the ultimate outcome of insulin signaling, in insulin-resistant neuro-2a cells. We first time showed that CDK5 inhibition by roscovitine could ameliorate insulin resistance and increase glucose uptake in neuronal cells via ERK1/2 pathway. Our study provides intriguing insights about the effect of CDK5 inhibition on neuronal insulin resistance and opens up a new paradigm to develop new therapeutic strategies for neuronal insulin resistance and associated pathophysiological conditions.
Collapse
Affiliation(s)
- Kapil Manglani
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Chinmoy S Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| |
Collapse
|
12
|
Sharma M, Dey CS. Role of Akt isoforms in neuronal insulin signaling and resistance. Cell Mol Life Sci 2021; 78:7873-7898. [PMID: 34724097 PMCID: PMC11073101 DOI: 10.1007/s00018-021-03993-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/04/2023]
Abstract
The aim of the present study was to determine the role of Akt isoforms in insulin signaling and resistance in neuronal cells. By silencing Akt isoforms individually and in pairs, in Neuro-2a and HT22 cells we observed that, in insulin-sensitive condition, Akt isoforms differentially reduced activation of AS160 and glucose uptake with Akt2 playing the major role. Under insulin-resistant condition, phosphorylation of all isoforms and glucose uptake were severely affected. Over-expression of individual isoforms in insulin-sensitive and resistant cells differentially reversed AS160 phosphorylation with concomitant reversal in glucose uptake indicating a compensatory role of Akt isoforms in controlling neuronal insulin signaling. Post-insulin stimulation Akt2 translocated to the membrane the most followed by Akt3 and Akt1, decreasing glucose uptake in the similar order in insulin-sensitive cells. None of the Akt isoforms translocated in insulin-resistant cells or high-fat-diet mediated diabetic mice brain cells. Based on our data, insulin-dependent differential translocation of Akt isoforms to the plasma membrane turns out to be the key factor in determining Akt isoform specificity. Thus, isoforms play parallel with predominant role by Akt2, and compensatory yet novel role by Akt1 and Akt3 to regulate neuronal insulin signaling, glucose uptake, and insulin-resistance.
Collapse
Affiliation(s)
- Medha Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
13
|
Abstract
The Akt isoforms-AS160-GLUT4 axis is the primary axis that governs glucose homeostasis in the body. The first step on the path to insulin resistance is deregulated Akt isoforms. This could be Akt isoform expression, its phosphorylation, or improper isoform-specific redistribution to the plasma membrane in a specific tissue system. The second step is deregulated AS160 expression, its phosphorylation, improper dissociation from glucose transporter storage vesicles (GSVs), or its inability to bind to 14-3-3 proteins, thus not allowing it to execute its function. The final step is improper GLUT4 translocation and aberrant glucose uptake. These processes lead to insulin resistance in a tissue-specific way affecting the whole-body glucose homeostasis, eventually progressing to an overt diabetic phenotype. Thus, the relationship between these three key proteins and their proper regulation comes out as the defining axis of insulin signaling and -resistance. This review summarizes the role of this central axis in insulin resistance and disease in a new light.
Collapse
Affiliation(s)
- Medha Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
14
|
Martins VF, LaBarge SA, Stanley A, Svensson K, Hung CW, Keinan O, Ciaraldi TP, Banoian D, Park JE, Ha C, Hetrick B, Meyer GA, Philp A, David LL, Henry RR, Aslan JE, Saltiel AR, McCurdy CE, Schenk S. p300 or CBP is required for insulin-stimulated glucose uptake in skeletal muscle and adipocytes. JCI Insight 2021; 7:141344. [PMID: 34813504 PMCID: PMC8765050 DOI: 10.1172/jci.insight.141344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
While current thinking posits that insulin signaling to GLUT4 exocytic translocation and glucose uptake in skeletal muscle and adipocytes is controlled by phosphorylation-based signaling, many proteins in this pathway are acetylated on lysine residues. However, the importance of acetylation and lysine acetyltransferases to insulin-stimulated glucose uptake is incompletely defined. Here, we demonstrate that combined loss of the acetyltransferases E1A binding protein p300 (p300) and cAMP response element binding protein binding protein (CBP) in mouse skeletal muscle causes a complete loss of insulin-stimulated glucose uptake. Similarly, brief (i.e. 1 h) pharmacological inhibition of p300/CBP acetyltransferase activity recapitulates this phenotype in human and rodent myotubes, 3T3-L1 adipocytes, and mouse muscle. Mechanistically, these effects are due to p300/CBP-mediated regulation of GLUT4 exocytic translocation and occurs downstream of Akt signaling. Taken together, we highlight a fundamental role for acetylation and p300/CBP in the direct regulation of insulin-stimulated glucose transport in skeletal muscle and adipocytes.
Collapse
Affiliation(s)
- Vitor F Martins
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Samuel A LaBarge
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Alexandra Stanley
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Kristoffer Svensson
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Chao-Wei Hung
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Omer Keinan
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Theodore P Ciaraldi
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Dion Banoian
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Ji E Park
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Christina Ha
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, United States of America
| | - Gretchen A Meyer
- Program in Physical Therapy, Washington University in St. Louis, St. Louis, United States of America
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Larry L David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States of America
| | - Robert R Henry
- Division of Endocrinology & Metabolism, VA San Diego Healthcare System, San Diego, United States of America
| | - Joseph E Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, United States of America
| | - Alan R Saltiel
- University of California, San Diego, La Jolla, United States of America
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, United States of America
| | - Simon Schenk
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| |
Collapse
|
15
|
Programmed cell death 5 improves skeletal muscle insulin resistance by inhibiting IRS-1 ubiquitination through stabilization of MDM2. Life Sci 2021; 285:119918. [PMID: 34480939 DOI: 10.1016/j.lfs.2021.119918] [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: 04/19/2021] [Revised: 07/18/2021] [Accepted: 08/24/2021] [Indexed: 11/20/2022]
Abstract
AIMS Insulin resistance is defined as the decreased sensitivity of tissues and organs to insulin and it is the main pathological basis of metabolic syndrome. PDCD5 is widely expressed in tissues including skeletal muscle and liver, but its exact function and the role in insulin resistance has not been studied. The present study is to explore the effect of PDCD5 on insulin resistance in skeletal muscle, the largest target organ of insulin, and its mechanism. MATERIALS AND METHODS Mice were fed with high-fat diet to establish obesity model. C2C12 myoblasts differentiated into myotubes and then were treated with palmitate to induce insulin resistance. Gain-of-function and loss-of-function experiments were performed by infecting C2C12 with adenovirus containing PDCD5 cDNA or PDCD5 shRNA. KEY FINDINGS PDCD5 protein was first increased and then decreased in the skeletal muscle from high-fat diet induced obese mice and consistently in palmitate induced insulin resistance C2C12 myotubes. Overexpression of PDCD5 in C2C12 cells did not affect the sensitivity to insulin but inhibited the palmitate induced insulin resistance, while knockdown of PDCD5 aggravated the insulin resistance. Mechanistically, PDCD5 interacted with ubiquitin ligase MDM2; overexpression of PDCD5 decreased MDM2 protein level, inhibited the increased interaction of MDM2 with IRS-1 and the degradation of IRS-1 by palmitate stimulation. SIGNIFICANCE PDCD5 is upregulated during the early stage of insulin resistance in skeletal muscle. The increased PDCD5 inhibits IRS-1 ubiquitination, increases the stability of IRS-1 by interacting with and degrading MDM2, thus providing a protective effect on insulin resistance in skeletal muscle.
Collapse
|
16
|
Wei R, Han C, He F, Xiong X, Ye F, Liu H, Li L, Xu H, Wei S, Zeng X. Role of forkhead box protein O1 and insulin on cell proliferation mediated by sirtuin 1 in goose primary hepatocytes. J APPL POULTRY RES 2021. [DOI: 10.1016/j.japr.2021.100144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
17
|
Dewanjee S, Vallamkondu J, Kalra RS, Chakraborty P, Gangopadhyay M, Sahu R, Medala V, John A, Reddy PH, De Feo V, Kandimalla R. The Emerging Role of HDACs: Pathology and Therapeutic Targets in Diabetes Mellitus. Cells 2021; 10:1340. [PMID: 34071497 PMCID: PMC8228721 DOI: 10.3390/cells10061340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus (DM) is one of the principal manifestations of metabolic syndrome and its prevalence with modern lifestyle is increasing incessantly. Chronic hyperglycemia can induce several vascular complications that were referred to be the major cause of morbidity and mortality in DM. Although several therapeutic targets have been identified and accessed clinically, the imminent risk of DM and its prevalence are still ascending. Substantial pieces of evidence revealed that histone deacetylase (HDAC) isoforms can regulate various molecular activities in DM via epigenetic and post-translational regulation of several transcription factors. To date, 18 HDAC isoforms have been identified in mammals that were categorized into four different classes. Classes I, II, and IV are regarded as classical HDACs, which operate through a Zn-based mechanism. In contrast, class III HDACs or Sirtuins depend on nicotinamide adenine dinucleotide (NAD+) for their molecular activity. Functionally, most of the HDAC isoforms can regulate β cell fate, insulin release, insulin expression and signaling, and glucose metabolism. Moreover, the roles of HDAC members have been implicated in the regulation of oxidative stress, inflammation, apoptosis, fibrosis, and other pathological events, which substantially contribute to diabetes-related vascular dysfunctions. Therefore, HDACs could serve as the potential therapeutic target in DM towards developing novel intervention strategies. This review sheds light on the emerging role of HDACs/isoforms in diabetic pathophysiology and emphasized the scope of their targeting in DM for constituting novel interventional strategies for metabolic disorders/complications.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | | | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba 305 8565, Japan;
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India;
| | - Moumita Gangopadhyay
- School of Life Science and Biotechnology, ADAMAS University, Barasat, Kolkata 700126, West Bengal, India;
| | - Ranabir Sahu
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling 734013, West Bengal, India;
| | - Vijaykrishna Medala
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
| | - Albin John
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
| | - P. Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.J.); (P.H.R.)
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana, India
| |
Collapse
|
18
|
Type-2 diabetes, a co-morbidity in Covid-19: does insulin signaling matter? Biochem Soc Trans 2021; 49:987-995. [PMID: 33666220 DOI: 10.1042/bst20201062] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/18/2021] [Accepted: 02/05/2021] [Indexed: 12/16/2022]
Abstract
Type-2 Diabetes is associated with one of the co-morbidities due to SARS-Coronavirus 2 (SARS-Cov2) infection. Clinical studies show out of control glucose levels in SARS-Cov2 infected patients with type-2 diabetes. There is no experimental evidence suggesting aberrant molecular pathway(s) that explains why SARS-Cov2 infected patients with type-2 diabetes have uncontrolled glucose homeostasis and are co-morbid. In this article, we have highlighted major proteins involved in SARS-Cov2 infection, like, ACE 2, proteases like, TMPRSS2, Furin and their connectivity to insulin signaling molecules like, PI3K, Akt, AMPK, MAPK, mTOR, those regulate glucose homeostasis and the possible outcome of that cross-talk. We also raised concerns about the effect of anti-SARS-Cov2 drugs on patients with type-2 diabetes with reference to insulin signaling and the outcome of their possible cross-talk. There are no studies to decipher the possibilities of these obvious cross-talks. The major objective of this article is to urge the scientific community to explore the possibility of determining whether derangement of insulin signaling could be one of the possible causes of the patients with type-2 diabetes being co-morbid due to SARS-Cov2 infection.
Collapse
|
19
|
Palomer X, Aguilar-Recarte D, García R, Nistal JF, Vázquez-Carrera M. Sirtuins: To Be or Not To Be in Diabetic Cardiomyopathy. Trends Mol Med 2021; 27:554-571. [PMID: 33839024 DOI: 10.1016/j.molmed.2021.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy is the leading cause of death among people with diabetes. Despite its severity and poor prognosis, there are currently no approved specific drugs to prevent or even treat diabetic cardiomyopathy. There is a need to understand the pathogenic mechanisms underlying the development of diabetic cardiomyopathy to design new therapeutic strategies. These mechanisms are complex and intricate and include metabolic dysregulation, inflammation, oxidative stress, fibrosis, and apoptosis. Sirtuins, a group of deacetylase enzymes, play an important role in all these processes and are, therefore, potential molecular targets for treating this disease. In this review, we discuss the role of sirtuins in the heart, focusing on their contribution to the pathogenesis of diabetic cardiomyopathy and how their modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - David Aguilar-Recarte
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Raquel García
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - J Francisco Nistal
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Departamento de Ciencias Médicas y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL); Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Santander, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
20
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
21
|
Zhou F, Zhang L, Zhu K, Bai M, Zhang Y, Zhu Q, Wang S, Sheng C, Yuan M, Liu Y, Lu J, Shao L, Wang X, Zhou L. SIRT2 ablation inhibits glucose-stimulated insulin secretion through decreasing glycolytic flux. Am J Cancer Res 2021; 11:4825-4838. [PMID: 33754030 PMCID: PMC7978320 DOI: 10.7150/thno.55330] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/06/2021] [Indexed: 11/30/2022] Open
Abstract
Rationale: Sirtuins are NAD+-dependent protein deacylases known to have protective effects against age-related diseases such as diabetes, cancer, and neurodegenerative disease. SIRT2 is the only primarily cytoplasmic isoform and its overall role in glucose homeostasis remains uncertain. Methods: SIRT2-knockout (KO) rats were constructed to evaluate the role of SIRT2 in glucose homeostasis. The effect of SIRT2 on β-cell function was detected by investigating the morphology, insulin secretion, and metabolomic state of islets. The deacetylation and stabilization of GKRP in β-cells by SIRT2 were determined by western blot, adenoviral infection, and immunoprecipitation. Results: SIRT2-KO rats exhibited impaired glucose tolerance and glucose-stimulated insulin secretion (GSIS), without change in insulin sensitivity. SIRT2 deficiency or inhibition by AGK2 decreased GSIS in isolated rat islets, with lowered oxygen consumption rate. Adenovirus-mediated overexpression of SIRT2 enhanced insulin secretion from rat islets. Metabolomics analysis revealed a decrease in metabolites of glycolysis and tricarboxylic acid cycle in SIRT2-KO islets compared with control islets. Our study further demonstrated that glucokinase regulatory protein (GKRP), an endogenous inhibitor of glucokinase (GCK), was expressed in rat islets. SIRT2 overexpression deacetylated GKRP in INS-1 β-cells. SIRT2 knockout or inhibition elevated GKRP protein stability in islet β-cells, leading to an increase in the interaction of GKRP and GCK. On the contrary, SIRT2 inhibition promoted the protein degradation of ALDOA, a glycolytic enzyme. Conclusions: SIRT2 ablation inhibits GSIS through blocking GKRP protein degradation and promoting ALDOA protein degradation, resulting in a decrease in glycolytic flux.
Collapse
|
22
|
Parikh HM, Elgzyri T, Alibegovic A, Hiscock N, Ekström O, Eriksson KF, Vaag A, Groop LC, Ström K, Hansson O. Relationship between insulin sensitivity and gene expression in human skeletal muscle. BMC Endocr Disord 2021; 21:32. [PMID: 33639916 PMCID: PMC7912896 DOI: 10.1186/s12902-021-00687-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Insulin resistance (IR) in skeletal muscle is a key feature of the pre-diabetic state, hypertension, dyslipidemia, cardiovascular diseases and also predicts type 2 diabetes. However, the underlying molecular mechanisms are still poorly understood. METHODS To explore these mechanisms, we related global skeletal muscle gene expression profiling of 38 non-diabetic men to a surrogate measure of insulin sensitivity, i.e. homeostatic model assessment of insulin resistance (HOMA-IR). RESULTS We identified 70 genes positively and 110 genes inversely correlated with insulin sensitivity in human skeletal muscle, identifying autophagy-related genes as positively correlated with insulin sensitivity. Replication in an independent study of 9 non-diabetic men resulted in 10 overlapping genes that strongly correlated with insulin sensitivity, including SIRT2, involved in lipid metabolism, and FBXW5 that regulates mammalian target-of-rapamycin (mTOR) and autophagy. The expressions of SIRT2 and FBXW5 were also positively correlated with the expression of key genes promoting the phenotype of an insulin sensitive myocyte e.g. PPARGC1A. CONCLUSIONS The muscle expression of 180 genes were correlated with insulin sensitivity. These data suggest that activation of genes involved in lipid metabolism, e.g. SIRT2, and genes regulating autophagy and mTOR signaling, e.g. FBXW5, are associated with increased insulin sensitivity in human skeletal muscle, reflecting a highly flexible nutrient sensing.
Collapse
Affiliation(s)
- Hemang M Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, 3650 Spectrum Blvd, Tampa, FL, 33612, USA.
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden.
| | - Targ Elgzyri
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
| | | | - Natalie Hiscock
- Unilever Discover R & D, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Ola Ekström
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
| | - Karl-Fredrik Eriksson
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
| | - Allan Vaag
- Steno Diabetes Center, DK-2820, Gentofte, Denmark
| | - Leif C Groop
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
- Finnish Institute of Molecular Medicine, FI-00014, University of Helsinki, Helsinki, Finland
| | - Kristoffer Ström
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
- Swedish Winter Sports Research Centre, Mid Sweden University, SE-83125, Östersund, Sweden
| | - Ola Hansson
- Department of Clinical Sciences, Diabetes & Endocrinology, Lund University, University Hospital Malmö, SE-20502, Malmö, Sweden
- Finnish Institute of Molecular Medicine, FI-00014, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Han Z, Chang C, Zhu W, Zhang Y, Zheng J, Kang X, Jin G, Gong Z. Role of SIRT2 in regulating the dexamethasone-activated autophagy pathway in skeletal muscle atrophy. Biochem Cell Biol 2021; 99:562-569. [PMID: 33481678 DOI: 10.1139/bcb-2020-0445] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The proteolytic autophagy system is involved in a major regulatory pathway in dexamethasone (Dex)-induced muscle atrophy. Sirtuin 2 (SIRT2) is known to modulate autophagy signaling, exerting effects in skeletal muscle atrophy. We examined the effects of SIRT2 on autophagy in Dex-induced myoatrophy. Tostudy this, mice were randomly distributed among the normal, Dex, and sirtinol groups. C2C12 cells were differentiated into myotubes and transduced with lentivirus carrying Sirt2-green fluorescent protein (GFP) or Sirt2 short hairpin RNA (Sirt2-shRNA)-GFP. To evaluate the mass and function of skeletal muscles, we measured myofiber cross-sectional area, myotube size, gastrocnemius (GA) muscle wet mass:body mass ratio (%), and time to exhaustion. The expression levels of SIRT2, myosin heavy chain, microtubule-associated protein 1 light chain 3 (LC3), and Beclin-1 were measured using Western blotting and quantitative reverse transcription - polymerase chain reaction. Inhibition of SIRT2 markedly attenuated GA muscle mass and endurance capacity. The same phenotype was observed in Sirt2-shRNA-treated myotubes, as evidenced by their decreased size. Conversely, overexpression of SIRT2 alleviated Dex-induced myoatrophy in vitro. Moreover, SIRT2 negatively regulated the expression of LC3b and Beclin-1 in skeletal muscles. These findings suggest that SIRT2 activation protects myotubes against Dex-induced atrophy through inhibition of the autophagy system; this phenomenon may serve as a target for treating glucocorticoid-induced myopathy.
Collapse
Affiliation(s)
- Ziqiu Han
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Cen Chang
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Weiyi Zhu
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yanlei Zhang
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jing Zheng
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xiangping Kang
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Guoqin Jin
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Zhangbin Gong
- Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Department of Biochemistry, College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
24
|
Gui LS, Raza SHA, Zhou L, Garcia M, Abd El-Aziz AH, Wei D, Hou S, Jia J, Wang Z. Association between Single Nucleotide Polymorphisms in SIRT1 and SIRT2 Loci and Growth in Tibetan Sheep. Animals (Basel) 2020; 10:ani10081362. [PMID: 32781630 PMCID: PMC7459998 DOI: 10.3390/ani10081362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary In summary, three single nucleotide polymorphisms (SNPs) were observed including two SNPs (g.3148 C > T and g.3570 G > A) in SIRT1, and one SNP (g.8074 T > A) in SIRT2 through sequence analysis. Association analyses suggested that all three SNPs were associated growth-related traits in Tibetan sheep. These findings imply that both SIRT1 and SIRT2 may play an important role in growth traits and are potential biomarkers for Marker-assisted selection (MAS). Abstract Silent information regulator 1 and 2 (SIRT1, 2) were NAD+-dependent histone or non-histone deacetylase, which emerged as key metabolic sensors in several tissues of mammals. In the present study, the search for polymorphisms within the ovine SIRT1 and SIRT2 loci as well as association analyses between SNPs and growth-related traits were performed in Tibetan sheep. To determine the expression pattern of SIRT1 and SIRT2 genes in Tibetan sheep, the quantitative real-time polymerase chain reaction (qPCR) analysis revealed that those two genes were widely expressed in diverse tissues. Expression of SIRT1 was less in abomasum of lamb, whereas it was greater in duodenum within adult stage. In the case of SIRT2, the greatest expression was observed in reticulum (lamb) and in muscle (adult), whereas the least expression was in liver for lamb and in kidney for adult animals. The association analysis demonstrated that g.3148 C > T polymorphism of SIRT1 affected heart girth (p = 0.002). The g.8074 T > A SNP of SIRT2 had a significant correlation with body weight (p = 0.011) and body length (p = 0.008). These findings suggested that the SIRT1 and SIRT2 polymorphism was involved in growth-related traits in Tibetan sheep, which may be considered to be genetic markers for improving the growth traits of Tibetan sheep.
Collapse
Affiliation(s)
- Lin-sheng Gui
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China; (L.-s.G.); (L.Z.); (S.H.); (J.J.)
| | - Sayed Haidar Abbas Raza
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Li Zhou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China; (L.-s.G.); (L.Z.); (S.H.); (J.J.)
| | - Matthew Garcia
- School of Animal Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA;
| | - Ayman Hassan Abd El-Aziz
- Animal Husbandry and Animal Wealth Development Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Dawei Wei
- School of Agriculture, Ningxia University, Yinchuan 750021, China;
| | - Shengzhen Hou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China; (L.-s.G.); (L.Z.); (S.H.); (J.J.)
| | - Jianlei Jia
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China; (L.-s.G.); (L.Z.); (S.H.); (J.J.)
| | - Zhiyou Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, Qinghai, China; (L.-s.G.); (L.Z.); (S.H.); (J.J.)
- Correspondence:
| |
Collapse
|
25
|
Haplotypes of the Mutated SIRT2 Promoter Contributing to Transcription Factor Binding and Type 2 Diabetes Susceptibility. Genes (Basel) 2020; 11:genes11050569. [PMID: 32438712 PMCID: PMC7288287 DOI: 10.3390/genes11050569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/26/2022] Open
Abstract
Genetic variability is an important causative factor for susceptibility and pathogenesis of type 2 diabetes (T2D). Histone deacetylase, sirtuin 2 (SIRT2), plays regulatory roles in glucose metabolism and insulin sensitivity. However, whether the SIRT2 variants or haplotypes contribute to T2D risk remain to be elucidated. In this study, we first detected three novel polymorphisms (P-MU1, P-MU2, and P-MU3) in the promoter of SIRT2 in the Chinese population. All pairwise sets of the three loci were strongly in linkage disequilibrium. Next, we constructed the haplotype block structure, and found H1-GGC and H2-CCA accounted for the most (total 91.8%) in T2D. The haplotype combination H1-H1-GGGGCC displayed a high risk for T2D (OR = 2.03, 95% CI = 1.12-3.72). By association analysis, we found the individuals carrying H1-H1-GGGGCC had significantly higher fasting plasma glucose and glycated hemoglobin. The haplotype H1-GGC presented a 6.74-fold higher promoter activity than H2-CCA, which was consistent with the correlation results. Furthermore, we clarified the mechanism whereby the C allele of both the P-MU1 and P-MU2 loci disrupted the signal transducer and activator of transcription 1 (STAT1) binding sites, leading to the attenuation of the SIRT2 transcription. Together, these data suggest that the linked haplotype GGC could be considered as a promising marker for T2D diagnosis and therapy assessment.
Collapse
|
26
|
Emamgholipour S, Ebrahimi R, Bahiraee A, Niazpour F, Meshkani R. Acetylation and insulin resistance: a focus on metabolic and mitogenic cascades of insulin signaling. Crit Rev Clin Lab Sci 2020:1-19. [DOI: 10.1080/10408363.2019.1699498] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farshad Niazpour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Wang Y, Yang J, Hong T, Chen X, Cui L. SIRT2: Controversy and multiple roles in disease and physiology. Ageing Res Rev 2019; 55:100961. [PMID: 31505260 DOI: 10.1016/j.arr.2019.100961] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/11/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022]
Abstract
Sirtuin 2 (SIRT2) is an NAD+-dependent deacetylase that was under studied compared to other sirtuin family members. SIRT2 is the only sirtuin protein which is predominantly found in the cytoplasm but is also found in the mitochondria and in the nucleus. Recently, accumulating evidence has uncovered a growing number of substrates and additional detailed functions of SIRT2 in a wide range of biological processes, marking its crucial role. Here, we give a comprehensive profile of the crucial physiological functions of SIRT2 and its role in neurological diseases, cancers, and other diseases. This review summarizes the functions of SIRT2 in the nervous system, mitosis regulation, genome integrity, cell differentiation, cell homeostasis, aging, infection, inflammation, oxidative stress, and autophagy. SIRT2 inhibition rescues neurodegenerative disease symptoms and hence SIRT2 is a potential therapeutic target for neurodegenerative disease. SIRT2 is undoubtedly dysfunctional in cancers and plays a dual-faced role in different types of cancers, and although its mechanism is unresolved, SIRT2 remains a promising therapeutic target for certain cancers. In future, the continued rapid growth in SIRT2 research will help clarify its role in human health and disease, and promote the progress of this target in clinical practice.
Collapse
Affiliation(s)
- Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jingqi Yang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tingting Hong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiongjin Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
28
|
Chen M, Yao YL, Yang Y, Zhu M, Tang Y, Liu S, Li K, Tang Z. Comprehensive Profiles of mRNAs and miRNAs Reveal Molecular Characteristics of Multiple Organ Physiologies and Development in Pigs. Front Genet 2019; 10:756. [PMID: 31552085 PMCID: PMC6737989 DOI: 10.3389/fgene.2019.00756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
The pig (Sus scrofa) is not only an important livestock animal but also widely used as a biomedical model. However, the understanding of the molecular characteristics of organs and of the developmental skeletal muscle of the pig is severely limited. Here, we performed a comprehensive transcriptome profiling of mRNAs and miRNAs across nine tissues and three skeletal muscle developmental stages in the Guizhou miniature pig. The reproductive organs (ovary and testis) had greater transcriptome complexity and activity than other tissues, and the highest transcriptome similarity was between skeletal muscle and heart (R = 0.79). We identified 1,819 mRNAs and 96 miRNAs to be tissue-specific in nine organs. Testis had the largest number of tissue-specific mRNAs (992) and miRNAs (40). Only 15 genes and two miRNAs were specifically expressed in skeletal muscle and fat, respectively. During postnatal skeletal muscle development, the mRNAs associated with focal adhesion, Notch signaling, protein digestion, and absorption pathways were up-regulated from D0 to D30 and then down-regulated from D30 and D240, while genes with opposing expression patterns were significantly enriched in the oxidative phosphorylation and proteasome pathways. The miRNAs mainly regulated genes associated with insulin, Wnt, fatty acid biosynthesis, Notch, MAPK, TGF-beta, insulin secretion, ECM-receptor interaction, focal adhesion, and calcium signaling pathways. We also identified 37 new miRNA-mRNA interaction pairs involved in skeletal muscle development. Overall, our data not only provide a rich resource for understanding pig organ physiology and development but also aid the study of the molecular functions of mRNA and miRNA in mammals.
Collapse
Affiliation(s)
- Muya Chen
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yi Long Yao
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yalan Yang
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Min Zhu
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yijie Tang
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Siyuan Liu
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Kui Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhonglin Tang
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.,Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
29
|
Martins VF, Begur M, Lakkaraju S, Svensson K, Park J, Hetrick B, McCurdy CE, Schenk S. Acute inhibition of protein deacetylases does not impact skeletal muscle insulin action. Am J Physiol Cell Physiol 2019; 317:C964-C968. [PMID: 31461343 DOI: 10.1152/ajpcell.00159.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Whether the histone deacetylase (HDAC) and sirtuin families of protein deacetylases regulate insulin-stimulated glucose uptake, independent of their transcriptional effects, has not been studied. Our objective was to determine the nontranscriptional role of HDACs and sirtuins in regulation of skeletal muscle insulin action. Basal and insulin-stimulated glucose uptake and signaling and acetylation were assessed in L6 myotubes and skeletal muscle from C57BL/6J mice that were treated acutely (1 h) with HDAC (trichostatin A, panobinostat, TMP195) and sirtuin inhibitors (nicotinamide). Treatment of L6 myotubes with HDAC inhibitors or skeletal muscle with a combination of HDAC and sirtuin inhibitors increased tubulin and pan-protein acetylation, demonstrating effective impairment of HDAC and sirtuin deacetylase activities. Despite this, neither basal nor insulin-stimulated glucose uptake or insulin signaling was impacted. Acute reduction of the deacetylase activity of HDACs and/or sirtuins does not impact insulin action in skeletal muscle.
Collapse
Affiliation(s)
- Vitor F Martins
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California.,Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California
| | - Maedha Begur
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California
| | - Shivani Lakkaraju
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California
| | - Kristoffer Svensson
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California
| | - Ji Park
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, California.,Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California
| |
Collapse
|
30
|
Shi P, Zhou M, Yang Y. Upregulated tumor sirtuin 2 expression correlates with reduced TNM stage and better overall survival in surgical breast cancer patients. Ir J Med Sci 2019; 189:83-89. [PMID: 31418154 DOI: 10.1007/s11845-019-02071-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND This study is aimed at exploring the correlation of sirtuin 2 (SIRT2) with clinical characteristics as well as overall survival (OS) in breast cancer patients. METHODS Totally, 296 primary breast cancer patients who underwent surgical resection were retrospectively reviewed in this study, and SIRT2 expression in tumor and adjacent tissues was determined by immunohistochemistry (IHC) and scored by semiquantitative scoring (0-12). Clinicopathological features were retrieved, and OS was calculated. RESULTS Both SIRT2 IHC semiquantitative score and percentage of SIRT2 high expression by IHC score > 3 were lower in tumor tissues compared with adjacent tissues. Additionally, tumor SIRT2 high expression was associated with lower T stage, decreased N stage, and reduced TNM stage. Kaplan-Meier curves displayed that tumor SIRT2 high expression predicted longer OS. Univariate Cox's regression analysis showed that tumor SIRT2 high expression was associated with prolonged OS, while multivariate Cox's regression analysis displayed that tumor SIRT2 high expression was not an independent predictive factor for OS, which implied that tumor SIRT2 might predict OS indirectly through the interaction of tumor features (such as TNM stage) in breast cancer patients. CONCLUSION SIRT2 expression is lower in tumor tissues compared with adjacent tissues, and tumor SIRT2 high expression correlates with lower T stage, decreased N stage, reduced TNM stage, and longer OS in breast cancer patients.
Collapse
Affiliation(s)
- Pengfei Shi
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street Jiangan District, Wuhan, 430014, China
| | - Min Zhou
- Department of Out-patient, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggang Yang
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street Jiangan District, Wuhan, 430014, China.
| |
Collapse
|
31
|
Chromone-3-aldehyde derivatives – sirtuin 2 inhibitors for correction of muscular dysfunction. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2019. [DOI: 10.2478/cipms-2019-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The aim of the study was to evaluate experimentally, the myoprotective effect of new chromone-3-aldehyde derivatives in conditions of muscular dysfunction and to establish a potential mechanism of myoprotective activity – the blockade of the function of sirutin 2. Materials and methods. The effect of new chromone-3-aldehyde derivatives on the development of muscular dysfunction under the conditions of an electromiostimulation test, was studied. The degree of muscle fatigue was evaluated in the «grip-strength» and through test biochemical assays (determination of the activity of lactate dehydrogenase, creatine kinase, concentration of lactic and pyruvic acids, creatinine, myoglobin, and total protein) to determine the possible mechanism of action of the test compounds (5 new derivatives of chromone-3-aldehyde) and their effect on the function of sirtuin 2 was evaluated.
Results. The study showed that chromone-3-aldehyde derivatives have a pronounced myoprotective effect associated with low toxicity (class 5 toxicity according to the GHS classification), which was confirmed by the results of the «grip-strength» test and biochemical tests data. Test compounds under the X3AC1, X3AOAC and X3AN codes evince sirtuin 2 inhibitory activity, which was reflected in a decrease in its concentration by 63.6% (p <0.05); 130.2% (p <0.05) and 218.8% (p <0.05).
Conclusion. The study showed that chromone-3-aldehyde derivatives are promising subjects for further study with the goal of creating a drug with a high myoprotective effect and an optimal safety profile.
Collapse
|
32
|
High expression of CPT1b in skeletal muscle in metabolically healthy older subjects. DIABETES & METABOLISM 2019; 45:152-159. [DOI: 10.1016/j.diabet.2018.01.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 12/21/2017] [Accepted: 01/14/2018] [Indexed: 12/25/2022]
|
33
|
Ko C, Lin R, Zeng Y, Chang W, Huang D, Wu JS, Chang Y, Shen S. Ameliorative effect of Ruellia tuberosa L. on hyperglycemia in type 2 diabetes mellitus and glucose uptake in mouse C2C12 myoblasts. Food Sci Nutr 2018; 6:2414-2422. [PMID: 30510742 PMCID: PMC6261179 DOI: 10.1002/fsn3.840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 01/07/2023] Open
Abstract
Ruellia tuberosa L. (RTL) exhibits a wide range of phytochemical activities, for example, on treatment of diabetes mellitus (DM), in Orient. There is, however, few study regarding the effect of RTL on glycemic-related homeostasis in type 2 DM (T2DM). We investigated the effect of RTL aqueous and ethanolic extracts on hypoglycemia in high-fat diet (HFD)-fed plus streptozotocin (STZ)-induced T2DM rats, and examined the effect of RTL on glucose uptake in tumor necrosis factor-α-induced insulin-resistant mouse C2C12 myoblasts, a mouse skeletal muscle cell line. The administration of 100 or 400 mg kg BW-1 day-1 of RTL aqueous or ethanolic extracts once a day for 4 weeks significantly ameliorated hyperglycemia, hyperinsulinemia, and the insulin resistance (IR) index in diabetic rats. RTL either aqueous or ethanolic extract at a concentration of 25-800 μg/ml significantly improved glucose uptake in insulin-resistant mouse C2C12 myoblasts, indicating inhibiting the IR in skeletal muscles. These evidences suggest that RTL ameliorates hyperglycemia in HFD/STZ-induced T2DM rats may be attributed to the alleviation of IR in skeletal muscles.
Collapse
Affiliation(s)
- Chih‐Yuan Ko
- Department of Respiratory and Critical Care Medicinethe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- Respiratory Medicine Center of Fujian ProvinceQuanzhouChina
- Key Laboratory of Fujian Medical UniversityFujian Province UniversityQuanzhouChina
- Department of Endocrinology and Metabolismthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Ru‐Hai Lin
- Department of Endocrinology and Metabolismthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Yi‐Ming Zeng
- Department of Respiratory and Critical Care Medicinethe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- Respiratory Medicine Center of Fujian ProvinceQuanzhouChina
- Key Laboratory of Fujian Medical UniversityFujian Province UniversityQuanzhouChina
| | - Wen‐Chang Chang
- Department of Food ScienceNational Chiayi UniversityChiayi CityTaiwan
| | - Da‐Wei Huang
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainan CityTaiwan
| | - James Swi‐Bea Wu
- Graduate Institute of Food Science and TechnologyNational Taiwan UniversityTaipeiTaiwan
| | - Yu‐Fang Chang
- Department of BiotechnologySouthern Taiwan University of Science and TechnologyTainan CityTaiwan
| | - Szu‐Chuan Shen
- Department of Human Development and Family StudiesNational Taiwan Normal UniversityTaipeiTaiwan
- Grauduate Program of Nutrition ScienceNational Taiwan Normal UniversityTaipeiTaiwan
| |
Collapse
|
34
|
Parodi-Rullán RM, Chapa-Dubocq XR, Javadov S. Acetylation of Mitochondrial Proteins in the Heart: The Role of SIRT3. Front Physiol 2018; 9:1094. [PMID: 30131726 PMCID: PMC6090200 DOI: 10.3389/fphys.2018.01094] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
Abstract
A growing number of studies have demonstrated the role of post-translational modifications of proteins, particularly acetylation, in human diseases including neurodegenerative and cardiovascular diseases, diabetes, cancer, and in aging. Acetylation of mitochondrial proteins has been shown to be involved in the pathogenesis of cardiac diseases such as myocardial infarction (ischemia-reperfusion) and heart failure. Indeed, over 60% of mitochondrial proteins contain acetylation sites, and most of these proteins are involved in mitochondrial bioenergetics. Mitochondrial non-enzymatic acetylation is enabled by acetyl-coenzyme A abundance and serves as the primary pathway of acetylation in mitochondria. Hence, regulation of enzymatic deacetylation becomes the most important mechanism to control acetylation/deacetylation of mitochondrial proteins. Acetylation/deacetylation of mitochondrial proteins has been regarded as a key regulator of mitochondrial metabolism and function. Proteins are deacetylated by NAD+-dependent deacetylases known as sirtuins (SIRTs). Among seven sirtuin isoforms, only SIRT3, SIRT4, and SIRT5 are localized in the mitochondria. SIRT3 is the main mitochondrial sirtuin which plays a key role in maintaining metabolic and redox balance in the mitochondria under physiological and pathological conditions. SIRT3 regulates the enzymatic activity of proteins involved in fatty acid oxidation, tricarboxylic acid cycle, electron transport chain, and oxidative phosphorylation. Although many enzymes have been identified as targets for SIRT3, cardiac-specific SIRT3 effects and regulations could differ from those in non-cardiac tissues. Therefore, it is important to elucidate the contribution of SIRT3 and mitochondrial protein acetylation/deacetylation in mitochondrial metabolism and cardiac dysfunction. Here, we summarize previous studies and provide a comprehensive analysis of the role of SIRT3 in mitochondria metabolism and bioenergetics under physiological conditions and in cardiac diseases. In addition, the review discusses mitochondrial protein acetylation as a potential target for cardioprotection.
Collapse
Affiliation(s)
- Rebecca M Parodi-Rullán
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| | - Xavier R Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, United States
| |
Collapse
|
35
|
SIRT2 and Akt mediate NAD+-induced and NADH-induced increases in the intracellular ATP levels of BV2 microglia under basal conditions. Neuroreport 2018; 29:65-70. [PMID: 29189472 DOI: 10.1097/wnr.0000000000000876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
NAD replenishment can restore ATP levels and rescue premature aging in Cockayne syndrome mice. However, there has been no mechanistic study regarding the effects of NAD and NADH on intracellular ATP levels under basal conditions. In our current study, we used BV2 microglia to test our hypothesis that NAD and NADH can increase intracellular ATP levels under basal conditions. We found that both NAD and NADH significantly increased the intracellular ATP levels of BV2 microglia, which were attenuated by SIRT2 siRNA, the SIRT2 inhibitor AGK2, and the phosphatidylinositol 3-kinase/Akt inhibitor LY294002. Our study has also suggested that SIRT2 mediates the NAD-induced and NADH-induced increase in Akt phosphorylation in BV2 microglia. Collectively, our study has suggested that SIRT2 mediates both NAD-induced and NADH-induced increases in the intracellular ATP levels of BV2 microglia by modulating Akt phosphorylation.
Collapse
|
36
|
Zullo A, Simone E, Grimaldi M, Musto V, Mancini FP. Sirtuins as Mediator of the Anti-Ageing Effects of Calorie Restriction in Skeletal and Cardiac Muscle. Int J Mol Sci 2018; 19:E928. [PMID: 29561771 PMCID: PMC5979282 DOI: 10.3390/ijms19040928] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/14/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Fighting diseases and controlling the signs of ageing are the major goals of biomedicine. Sirtuins, enzymes with mainly deacetylating activity, could be pivotal targets of novel preventive and therapeutic strategies to reach such aims. Scientific proofs are accumulating in experimental models, but, to a minor extent, also in humans, that the ancient practice of calorie restriction could prove an effective way to prevent several degenerative diseases and to postpone the detrimental signs of ageing. In the present review, we summarize the evidence about the central role of sirtuins in mediating the beneficial effects of calorie restriction in skeletal and cardiac muscle since these tissues are greatly damaged by diseases and advancing years. Moreover, we entertain the possibility that the identification of sirtuin activators that mimic calorie restriction could provide the benefits without the inconvenience of this dietary style.
Collapse
Affiliation(s)
- Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
- CEINGE Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Emanuela Simone
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | - Maddalena Grimaldi
- Department of Pediatric Oncology and Hematology, Charité University Hospital, 13353 Berlin, Germany.
| | - Vincenzina Musto
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | | |
Collapse
|
37
|
Abstract
SIGNIFICANCE Extranuclear sirtuins in cytosol (SIRT2) and mitochondria (SIRT3, SIRT4, and SIRT5) are key regulators of metabolic enzymes and the antioxidative defense mechanisms. They play an important role in the adjustment of metabolic pathways in alterations of the nutritional status. Recent Advances: Recent studies have shown that in addition to lysine deacetylation, sirtuins catalyze several different lysine deacylation reactions, removal of lipid modifications, and adenosine diphosphate-ribosylation. Large-scale studies have revealed hundreds of target proteins regulated by different sirtuin modifications. CRITICAL ISSUES Sensing of the metabolic state and regulation of the sirtuin function and expression are critical components of the machinery, optimizing cellular functions in the switch from fed to fasting condition. Overfeeding, obesity, and metabolic diseases cause metabolic stress that dysregulates the sirtuins, which may play a role in the pathogenesis and complications of metabolic diseases such as type 2 diabetes, fatty liver disease, and cardiac diseases. In the current review, we will discuss the significance of the extranuclear sirtuins as metabolic regulators and in protection against the reactive oxygen species, and also how these sirtuins are regulated by metabolic status and their putative role in metabolic diseases. FUTURE DIRECTIONS To efficiently utilize sirtuins as drug targets for treatment of the metabolic diseases, better understanding of the sirtuin functions, targets, regulation, and cross talk is needed. Furthermore, more studies in humans are needed to confirm the many observations mainly made in animal and cell models so far. Antioxid. Redox Signal. 28, 662-676.
Collapse
Affiliation(s)
- Mahmoud-Sobhy Elkhwanky
- 1 Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu , Oulu, Finland .,2 Medical Research Center Oulu, Oulu University Hospital and University of Oulu , Oulu, Finland
| | - Jukka Hakkola
- 1 Research Unit of Biomedicine, Pharmacology and Toxicology, University of Oulu , Oulu, Finland .,2 Medical Research Center Oulu, Oulu University Hospital and University of Oulu , Oulu, Finland
| |
Collapse
|
38
|
Liu T, Yang W, Pang S, Yu S, Yan B. Functional genetic variants within the SIRT2 gene promoter in type 2 diabetes mellitus. Diabetes Res Clin Pract 2018; 137:200-207. [PMID: 29371109 DOI: 10.1016/j.diabres.2018.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
AIMS Type 2 diabetes mellitus (T2D) is a common and complex metabolic diseases caused by interactions between environmental and genetic factors. Genome-wide association studies have identified more than 80 common genetic variants for T2D, which account for only ∼10% of the heritability of T2D cases. SIRT2, a member of NAD(+)-dependent class III deacetylases, is involved in genomic stability, metabolism, inflammation, oxidative stress and autophagy. In maintaining metabolic homeostasis, SIRT2 regulates adipocyte differentiation, fatty acid oxidation, gluconeogenesis, and insulin sensitivity. Thus, we hypothesized that DNA sequence variants (DSVs) in SIRT2 gene promoter may change SIRT2 levels, contributing to T2D. METHODS SIRT2 gene promoter was genetically and functionally analyzed in large cohorts of T2D patients (n = 365) and ethnic-matched controls (n = 358). RESULTS A total of 18 DSVs, including 5 SNPs, were identified in this study. Four novel heterozygous DSVs (g.38900912G > T, g.38900561C > T, g.38900359C > T and g.38900237G > A) were identified in four T2D patients, three of which (g.38900912G > T, g.38900359C > T and g.38900237G > A) significantly increased the transcriptional activity of the SIRT2 gene promoter in cultured pancreatic beta cells (P < .01). Seven novel heterozygous DSVs were only found in controls, and one heterozygous deletion DSV and five SNPs were found in both T2D patients and controls, which did not significantly affect SIRT2 gene promoter activity (P > .05). CONCLUSIONS Our findings suggested that the DSVs may increase SIRT2 gene promoter activity and SIRT2 levels, contributing to T2D development as a risk factor.
Collapse
Affiliation(s)
- Tingting Liu
- College of Clinical Medicine, Jining Medical University, Jining, Shandong 272100, China
| | - Wentao Yang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Shipeng Yu
- Division of Endocrinology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
39
|
Ye X, Li M, Hou T, Gao T, Zhu WG, Yang Y. Sirtuins in glucose and lipid metabolism. Oncotarget 2018; 8:1845-1859. [PMID: 27659520 PMCID: PMC5352102 DOI: 10.18632/oncotarget.12157] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/13/2016] [Indexed: 01/02/2023] Open
Abstract
Sirtuins are evolutionarily conserved protein, serving as nicotinamide adenine dinucleotide-dependent deacetylases or adenosine diphosphate-ribosyltransferases. The mammalian sirtuins family, including SIRT1~7, is involved in many biological processes such as cell survival, proliferation, senescence, stress response, genome stability and metabolism. Evidence accumulated over the past two decades has indicated that sirtuins not only serve as important energy status sensors but also protect cells against metabolic stresses. In this review, we summarize the background of glucose and lipid metabolism concerning sirtuins and discuss the functions of sirtuins in glucose and lipid metabolism. We also seek to highlight the biological roles of certain sirtuins members in cancer metabolism.
Collapse
Affiliation(s)
- Xin Ye
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Meiting Li
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Tianyun Hou
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Tian Gao
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Schiedel M, Robaa D, Rumpf T, Sippl W, Jung M. The Current State of NAD + -Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets. Med Res Rev 2017; 38:147-200. [PMID: 28094444 DOI: 10.1002/med.21436] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 12/19/2022]
Abstract
Sirtuins are NAD+ -dependent protein deacylases that cleave off acetyl, as well as other acyl groups, from the ε-amino group of lysines in histones and other substrate proteins. Seven sirtuin isotypes (Sirt1-7) have been identified in mammalian cells. As sirtuins are involved in the regulation of various physiological processes such as cell survival, cell cycle progression, apoptosis, DNA repair, cell metabolism, and caloric restriction, a dysregulation of their enzymatic activity has been associated with the pathogenesis of neoplastic, metabolic, infectious, and neurodegenerative diseases. Thus, sirtuins are promising targets for pharmaceutical intervention. Growing interest in a modulation of sirtuin activity has prompted the discovery of several small molecules, able to inhibit or activate certain sirtuin isotypes. Herein, we give an update to our previous review on the topic in this journal (Schemies, 2010), focusing on recent developments in sirtuin biology, sirtuin modulators, and their potential as novel therapeutic agents.
Collapse
Affiliation(s)
- Matthias Schiedel
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Dina Robaa
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Tobias Rumpf
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Wolfgang Sippl
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
43
|
Choudhary AK, Dey CS. Nuclear co-repressor (NCoR) is required to maintain insulin sensitivity in C 2 C 12 myotubes. Cell Biol Int 2016; 41:204-212. [PMID: 27935220 DOI: 10.1002/cbin.10711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/26/2016] [Indexed: 12/30/2022]
Abstract
Nuclear co-repressor (NCoR) regulates peripheral insulin sensitivity; however, its role in modulating insulin sensitivity in skeletal muscle remains elusive. Present study investigated protein expression and effect of NCoR on insulin sensitivity in murine skeletal muscle cell line C2 C12 . Myotubes as compared to myoblasts of C2 C12 cells were found to be more sensitive in response to insulin as increase in insulin-stimulated phosphorylation of AKT at serine 473 residue (pAKTS473 ) was significantly higher in myotubes. Incidentally, reduced protein level of NCoR coincided with differentiation of myoblasts into myotubes of C2 C12 cells. However, insulin stimulation per se failed to affect protein level of NCoR either in myoblasts or myotubes of C2 C12 cells. To assess the role of NCoR on insulin sensitivity, NCoR was transiently knocked down using siRNA in myotubes of C2 C12 . In fact, transient silencing of NCoR led to significant reduction in insulin-stimulated pAKTS473 and impaired glucose uptake. This observation is in contrast to published studies where NCoR has been reported to negatively regulate insulin signaling cascade. Furthermore, transient silencing of NCoR failed to improve insulin sensitivity in chronic hyperinsulinemia-induced insulin-resistant model of C2 C12 cells. Importantly, inhibition of lysosomal protein degradation pathway using ammonium chloride restored protein level of NCoR but failed to increase glucose uptake in serum-starved C2 C12 myotubes. Collectively, data from present study show differential protein level of NCoR under different cell state (myoblast and myotubes) of C2 C12 cells and NCoR proves to be vital for maintaining insulin sensitivity in C2 C12 myotubes.
Collapse
Affiliation(s)
- Abhijeet K Choudhary
- Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology (IIT)-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy S Dey
- Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology (IIT)-Delhi, Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
44
|
Varshney P, Dey CS. P21-activated kinase 2 (PAK2) regulates glucose uptake and insulin sensitivity in neuronal cells. Mol Cell Endocrinol 2016; 429:50-61. [PMID: 27040307 DOI: 10.1016/j.mce.2016.03.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/15/2022]
Abstract
P21-activated kinases (PAKs) are recently reported as important players of insulin signaling and glucose homeostasis in tissues like muscle, pancreas and liver. However, their role in neuronal insulin signaling is still unknown. Present study reports the involvement of PAK2 in neuronal insulin signaling, glucose uptake and insulin resistance. Irrespective of insulin sensitivity, insulin stimulation decreased PAK2 activity. PAK2 downregulation displayed marked enhancement of GLUT4 translocation with increase in glucose uptake whereas PAK2 over-expression showed its reduction. Treatment with Akti-1/2 and wortmannin suggested that Akt and PI3K are mediators of insulin effect on PAK2 and glucose uptake. Rac1 inhibition demonstrated decreased PAK2 activity while inhibition of PP2A resulted in increased PAK2 activity, with corresponding changes in glucose uptake. Taken together, present study demonstrates an inhibitory role of insulin signaling (via PI3K-Akt) and PP2A on PAK2 activity and establishes PAK2 as a Rac1-dependent negative regulator of neuronal glucose uptake and insulin sensitivity.
Collapse
Affiliation(s)
- Pallavi Varshney
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
45
|
Gao W, Wang H, Zhang L, Cao Y, Bao JZ, Liu ZX, Wang LS, Yang Q, Lu X. Retinol-Binding Protein 4 Induces Cardiomyocyte Hypertrophy by Activating TLR4/MyD88 Pathway. Endocrinology 2016; 157:2282-93. [PMID: 27100622 PMCID: PMC4891784 DOI: 10.1210/en.2015-2022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin resistance plays a major role in the development and progression of cardiac hypertrophy and heart failure. Heart failure in turn promotes insulin resistance and increases the risk for diabetes. The vicious cycle determines significant mortality in patients with heart failure and diabetes. However, the underlying mechanisms for the vicious cycle are not fully elucidated. Here we show that circulating levels and adipose expression of retinol-binding protein 4 (RBP4), an adipokine that contributes to systemic insulin resistance, were elevated in cardiac hypertrophy induced by transverse aortic constriction and angiotensin-II (Ang-II) infusion. Ang-II increased RBP4 expression in adipocytes, which was abolished by losartan, an Ang-II receptor blocker. The elevated RBP4 in cardiac hypertrophy may have pathophysiological consequences because RBP4 increased cell size, enhanced protein synthesis, and elevated the expression of hypertrophic markers including Anp, Bnp, and Myh7 in primary cardiomyocytes. Mechanistically, RBP4 induced the expression and activity of toll-like receptor 4 (TLR4) and myeloid differentiation primary response gene 88 (MyD88) in cardiomyocytes, resulting in enhanced inflammation and reactive oxygen species production. Inhibition or knockdown of the TLR4/MyD88 pathway attenuated inflammatory and hypertrophic responses to RBP4 stimulation. Importantly, RBP4 also reduced the expression of glucose transporter-4 and impaired insulin-stimulated glucose uptake in cardiomyocytes. This impairment was ameliorated in cardiomyocytes from TLR4 knockout mice. Therefore, RBP4 may be a critical modulator promoting the vicious cycle of insulin resistance and heart failure by activating TLR4/MyD88-mediated inflammatory pathways. Potentially, lowering RBP4 might break the vicious cycle and improve both insulin resistance and cardiac hypertrophy.
Collapse
Affiliation(s)
- Wei Gao
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wang
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lin Zhang
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yang Cao
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ji-Zhang Bao
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zheng-Xia Liu
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lian-Sheng Wang
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qin Yang
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiang Lu
- Department of Geriatrics (W.G., Z.-X.L., X.L.), the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China; Department of Medicine, Physiology, and Biophysics (W.G., L.Z., Y.C., J.-Z.B., Q.Y.), Center for Diabetes Research and Treatment, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, California 92697; and Department of Cardiology (H.W., L.-S.W.), the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
46
|
Gong H, Liu L, Ni CX, Zhang Y, Su WJ, Lian YJ, Peng W, Zhang JP, Jiang CL. Dexamethasone rapidly inhibits glucose uptake via non-genomic mechanisms in contracting myotubes. Arch Biochem Biophys 2016; 603:102-9. [PMID: 27246478 DOI: 10.1016/j.abb.2016.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 12/31/2022]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that regulate multiple aspects of glucose homeostasis. In skeletal muscle, it is well established that prolonged GC excess inhibits glucose uptake and utilization through glucocorticoid receptor (GR)-mediated transcriptional changes. However, it remains obscure that whether the rapid non-genomic effects of GC on glucose uptake are involved in acute exercise stress. Therefore, we used electric pulse stimulation (EPS)-evoked contracting myotubes to determine whether the non-genomic actions of GC were involved and its underlying mechanism(s). Pretreatment with dexamethasone (Dex, 10 μM) significantly prevented contraction-stimulated glucose uptake and glucose transporter 4 (Glut4) translocation within 20 min in C2C12 myotubes. Neither GC nuclear receptor antagonist (RU486) nor protein synthesis inhibitor (cycloheximide, Chx) affected the rapid inhibition effects of Dex. AMPK and CaMKII-dependent signaling pathways were associated with the non-genomic effects of Dex. These results provide evidence that GC rapidly suppresses glucose uptake in contracting myotubes via GR-independent non-genomic mechanisms. AMPK and CaMKII-mediated Glut4 translocation may play a critical role in GC-induced rapid inhibition of glucose uptake.
Collapse
Affiliation(s)
- Hong Gong
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Lei Liu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Chen-Xu Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, People's Republics of China
| | - Yi Zhang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Wen-Jun Su
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yong-Jie Lian
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Wei Peng
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jun-Ping Zhang
- Department of Pharmacy, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Chun-Lei Jiang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
47
|
SIRT2 regulates insulin sensitivity in insulin resistant neuronal cells. Biochem Biophys Res Commun 2016; 474:747-752. [PMID: 27163642 DOI: 10.1016/j.bbrc.2016.05.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
Insulin resistance in brain is well-associated with pathophysiology of deficits in whole-body energy metabolism, neurodegenerative diseases etc. Among the seven sirtuins, SIRT2 is the major deacetylase expressed in brain. Inhibition of SIRT2 confers neuroprotection in case of Parkinson's disease (PD) and Huntington's disease (HD). However, the role of this sirtuin in neuronal insulin resistance is not known. In this study, we report the role of SIRT2 in regulating insulin-sensitivity in neuronal cells in vitro. Using approaches like pharmacological inhibition of SIRT2, siRNA mediated SIRT2 knockdown and over-expression of wild-type and catalytically-mutated SIRT2, we observed that downregulation of SIRT2 ameliorated the reduced activity of AKT and increased insulin-stimulated glucose uptake in insulin resistant neuro-2a cells. The data was supported by over expression of catalytically-inactive SIRT2 in insulin-resistant human SH-SY5Y neuronal cells. Data highlights a crucial role of SIRT2 in regulation of neuronal insulin sensitivity under insulin resistant condition.
Collapse
|
48
|
Xu W, Jiang K, Shen M, Qian Y, Peng Y. SIRT2 suppresses non-small cell lung cancer growth by targeting JMJD2A. Biol Chem 2016; 396:929-36. [PMID: 25719312 DOI: 10.1515/hsz-2014-0284] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/12/2015] [Indexed: 11/15/2022]
Abstract
Lung cancer has been the most prolific cancer in China - as in the rest of the world - with a high death rate and low 5-year survival rate. Previous evidence showed that JMJD2A is over-expressed in human non-small cell lung cancer (NSCLC) tissues compared to adjacent normal tissues, and that high level of JMJD2A predicts poor overall and disease-free survival. However, the mechanism by which JMJD2A is regulated in human NSCLC is not fully understood. In the present study, we identified that the SIRT2 as an anti-oncogenic protein in NSCLC was down-regulated. JMJD2A as a target of SIRT2 was negatively correlated with SIRT2 level in NSCLC. SIRT2 bound to the promoter region of JMJD2A and negatively regulated JMJD2A expression. In addition, we found that SIRT2 inhibited NSCLC cells proliferation, colony formation and tumor growth in vitro and in vivo in a JMJD2A-dependent manner. In summary, our findings implicate that SIRT2 suppresses non-small cell lung cancer growth through targeting JMJD2A and SIRT2 activator may serve as candidate drug for NSCLC therapy.
Collapse
|
49
|
Yue CY, Yang CZ, Liu CY, Yang Y. Meta-analysis of relationship between Helicobacter pylori infection and level of insulin resistance. Shijie Huaren Xiaohua Zazhi 2015; 23:5238-5249. [DOI: 10.11569/wcjd.v23.i32.5238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the association between Helicobacter pylori (H. pylori) infection and level of insulin resistance (IR).
METHODS: The articles on the association of H. pylori with IR level were retrieved by searching databases from the inception of each database to July 2015. Data extraction and quality assessment were completed by two authors. Meta-analysis was performed using RevMan 5.3 software.
RESULTS: Seven papers with nine studies were included for meta-analysis. A total of 3097 patients, including 1718 H. pylori (+) subjects, were involved. The results of Meta-analysis showed that there was a significant difference in insulin resistance levels between patients with H. pylori infection and those without in the overall population (WMD = 0.38, 95%CI: 0.13-0.64, P < 0.05). Subgroup analysis indicated that the significant difference in insulin resistance levels between patients with H. pylori infection and those without existed in patients aged < 45 years (WMD = 0.63, 95%CI: 0.40-0.86, P < 0.05), those who underwent endoscopic biopsy (WMD = 0.70, 95%CI: 0.46-0.94, P < 0.05), those who had a body mass index (BMI) < 25 kg/m2 (WMD = 0.72, 95%CI:0.49-0.96, P < 0.05), and Asian patients (WMD = 0.66, 95%CI: 0.44-0.87, P < 0.05), but not in patients aged ≥ 45 years, those who underwent serum H. pylori-IgG antibody testing, those who had a BMI ≥ 25 kg/m2, or American patients. The results of publication bias and sensitivity analysis confirmed the reliability and stability of this Meta-analysis.
CONCLUSION: H. pylori infection is associated with elevated insulin resistance.
Collapse
|
50
|
Gomes P, Fleming Outeiro T, Cavadas C. Emerging Role of Sirtuin 2 in the Regulation of Mammalian Metabolism. Trends Pharmacol Sci 2015; 36:756-768. [DOI: 10.1016/j.tips.2015.08.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 12/23/2022]
|