1
|
Richter EA, Bilan PJ, Klip A. A comprehensive view of muscle glucose uptake: regulation by insulin, contractile activity, and exercise. Physiol Rev 2025; 105:1867-1945. [PMID: 40173020 DOI: 10.1152/physrev.00033.2024] [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: 08/27/2024] [Revised: 11/07/2024] [Accepted: 03/08/2025] [Indexed: 04/04/2025] Open
Abstract
Skeletal muscle is the main site of glucose deposition in the body during meals and the major glucose utilizer during physical activity. Although in both instances the supply of glucose from the circulation to the muscle is of paramount importance, in most conditions the rate-limiting step in glucose uptake, storage, and utilization is the transport of glucose across the muscle cell membrane. This step is dependent upon the translocation of the insulin- and contraction-responsive glucose transporter GLUT4 from intracellular storage sites to the sarcolemma and T tubules. Here, we first analyze how glucose can traverse the capillary wall into the muscle interstitial space. We then review the molecular processes that regulate GLUT4 translocation in response to insulin and muscle contractions and the methodologies utilized to unravel them. We further discuss how physical activity and inactivity, respectively, lead to increased and decreased insulin action in muscle and touch upon sex differences in glucose metabolism. Although many key processes regulating glucose uptake in muscle are known, the advent of newer and bioinformatics tools has revealed further molecular signaling processes reaching a staggering level of complexity. Much of this molecular mapping has emerged from cellular and animal studies and more recently from application of a variety of -omics in human tissues. In the future, it will be imperative to validate the translatability of results drawn from experimental systems to human physiology.
Collapse
Affiliation(s)
- Erik A Richter
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Lissek T. The universal role of adaptive transcription in health and disease. FEBS J 2025; 292:2479-2505. [PMID: 39609264 PMCID: PMC12103072 DOI: 10.1111/febs.17324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 07/25/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
In animals, adaptive transcription is a crucial mechanism to connect environmental stimulation to changes in gene expression and subsequent organism remodeling. Adaptive transcriptional programs involving molecules such as CREB, SRF, MEF2, FOS, and EGR1 are central to a wide variety of organism functions, including learning and memory, immune system plasticity, and muscle hypertrophy, and their activation increases cellular resilience and prevents various diseases. Yet, they also form the basis for many maladaptive processes and are involved in the progression of addiction, depression, cancer, cardiovascular disorders, autoimmune conditions, and metabolic dysfunction among others and are thus prime examples for mediating the adaptation-maladaptation dilemma. They are implicated in the therapeutic effects of major treatment modalities such as antidepressants and can have negative effects on treatment, for example, contributing to therapy resistance in cancer. This review examines the universal role of adaptive transcription as a mechanism for the induction of adaptive cell state transitions in health and disease and explores how many medical disorders can be conceptualized as caused by errors in cellular adaptation goals. It also considers the underlying principles in the basic structure of adaptive gene programs such as their division into a core and a directional program. Finally, it analyses how one might best reprogram cells via targeting of adaptive transcription in combination with complex stimulation patterns to leverage endogenous cellular reprogramming dynamics and achieve optimal health of the whole organism.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for NeurosciencesHeidelberg UniversityGermany
| |
Collapse
|
3
|
McGee SL, Hargreaves M. Exercise performance and health: Role of GLUT4. Free Radic Biol Med 2024; 224:479-483. [PMID: 39243828 DOI: 10.1016/j.freeradbiomed.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The glucose transporter GLUT4 is integral for optimal skeletal muscle performance during exercise, as well as for metabolic health. Physiological regulation of GLUT4 translocation during exercise and increased GLUT4 expression following exercise involves multiple, redundant signalling pathways. These include effects of reactive oxygen species (ROS). ROS contribute to GLUT4 translocation that increases skeletal muscle glucose uptake during exercise and stimulate signalling pathways that increase GLUT4 expression. Conversely, ROS can also inhibit GLUT4 translocation and expression in metabolic disease states. The opposing roles of ROS in GLUT4 regulation are ultimately linked to the metabolic state of skeletal muscle and the intricate mechanisms involved give insights into pathways critical for exercise performance and implicated in metabolic health and disease.
Collapse
Affiliation(s)
- Sean L McGee
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Waurn Ponds, 3217, Australia.
| | - Mark Hargreaves
- Department of Anatomy & Physiology, University of Melbourne, 3010, Australia.
| |
Collapse
|
4
|
Zhang J, Tian Z, Qin C, Momeni MR. The effects of exercise on epigenetic modifications: focus on DNA methylation, histone modifications and non-coding RNAs. Hum Cell 2024; 37:887-903. [PMID: 38587596 DOI: 10.1007/s13577-024-01057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/10/2024] [Indexed: 04/09/2024]
Abstract
Physical activity on a regular basis has been shown to bolster the overall wellness of an individual; research is now revealing that these changes are accompanied by epigenetic modifications. Regular exercise has been proven to make intervention plans more successful and prolong adherence to them. When it comes to epigenetic changes, there are four primary components. This includes changes to the DNA, histones, expression of particular non-coding RNAs and DNA methylation. External triggers, such as physical activity, can lead to modifications in the epigenetic components, resulting in changes in the transcription process. This report pays attention to the current knowledge that pertains to the epigenetic alterations that occur after exercise, the genes affected and the resulting characteristics.
Collapse
Affiliation(s)
- Junxiong Zhang
- Xiamen Academy of Art and Design, Fuzhou University, Xiamen, 361024, Fujian, China.
| | - Zhongxin Tian
- College of Physical Education, Taiyuan University of Technology, Taiyuan, 030024, Shanxi, China.
| | - Chao Qin
- College of Physical Education, Taiyuan University of Technology, Taiyuan, 030024, Shanxi, China
| | | |
Collapse
|
5
|
Carrillo ED, Hernández DI, Clara MV, Lezama I, García MC, Sánchez JA. Exercise increases MEF2A abundance in rat cardiac muscle by downregulating microRNA-223-5p. Sci Rep 2023; 13:14481. [PMID: 37660209 PMCID: PMC10475133 DOI: 10.1038/s41598-023-41696-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023] Open
Abstract
Exercise plays an important role in cardiac health and enhances the transport of glucose in cardiac muscle by increasing the glucose transporter-4 (GLUT4) content at the cell membrane. The GLUT4 gene is a target of myocyte enhancer transcription factor 2A (MEF2A). Several transcription factors are regulated by microRNAs (miRs), small non-coding RNAs that control gene expression at the posttranscriptional level. In this study we tested the hypothesis that exercise regulates the expression of miR-223 and that MEF2A is a direct target of miR-223. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot experiments showed that GLUT4 gene expression and protein abundance increased by 30 and 23%, respectively, in the microsomal fraction immediately after exercise, and had returned to control levels after 18 h. In contrast, the increase in GLUT4 in the membrane fraction was delayed. Exercise also increased the protein abundance of transcription factors involved in GLUT4 expression. Immediately after exercise, the protein abundance of MEF2A, nuclear respiratory factor 1 (NRF1), and forkhead box O1 (FOXO1) increased by 18, 30, and 40%, respectively. qRT-PCR experiments showed that miR-223-3p and miR-223-5p expression decreased immediately after exercise by 60 and 30%, respectively, and luciferase assays indicated that MEF2A is a target of the 5p strand of miR-223. Overexpression of miR-223-5p in H9c2 cells decreased the protein abundance of MEF2A. Our results suggest that the exercise-induced increase in GLUT4 content in cardiac muscle is partly due to the posttranscriptional increase in MEF2A protein abundance caused by the decrease in miR-223-5p expression. The exercise-induced decrease in miR-223-3p expression likely contributes to the increases in NRF1 and FOXO1 abundance and GLUT4 content.
Collapse
Affiliation(s)
- Elba D Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico
| | - Dulce I Hernández
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico
| | - Maikel Valle Clara
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico
| | - Ivonne Lezama
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico
| | - María C García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico
| | - Jorge A Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional 2508, CP 07360, Mexico City, Mexico.
| |
Collapse
|
6
|
PGC-1α and MEF2 Regulate the Transcription of the Carnitine Transporter OCTN2 Gene in C2C12 Cells and in Mouse Skeletal Muscle. Int J Mol Sci 2022; 23:ijms232012304. [PMID: 36293168 PMCID: PMC9604316 DOI: 10.3390/ijms232012304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
OCTN2 (SLC22A5) is a carnitine transporter whose main function is the active transport of carnitine into cells. In skeletal muscle and other organs, the regulation of the SLC22A5 gene transcription has been shown to depend on the nuclear transcription factor PPAR-α. Due to the observation that the muscle OCTN2 mRNA level is maintained in PPAR-α knock-out mice and that PGC-1α overexpression in C2C12 myoblasts increases OCTN2 mRNA expression, we suspected additional regulatory pathways for SLC22A5 gene transcription. Indeed, we detected several binding sites of the myocyte-enhancing factor MEF2 in the upstream region of the SLC22A5 gene, and MEF2C/MEF2D stimulated the activity of the OCTN2 promoter in gene reporter assays. This stimulation was increased by PGC-1α and was blunted for a SLC22A5 promoter fragment with a mutated MEF2 binding site. Further, we demonstrated the specific binding of MEF2 to the SLC22A5 gene promoter, and a supershift of the MEF2/DNA complex in electrophoretic mobility shift assays. In immunoprecipitation experiments, we could demonstrate the interaction between PGC-1α and MEF2. In addition, SB203580, a specific inhibitor of p38 MAPK, blocked and interferon-γ stimulated the transcriptional activity of the SLC22A5 gene promoter. Finally, mice with muscle-specific overexpression of OCTN2 showed an increase in OCTN2 mRNA and protein expression in skeletal muscle. In conclusion, we detected and characterized a second stimulatory pathway of SLC22A5 gene transcription in skeletal muscle, which involves the nuclear transcription factor MEF2 and co-stimulation by PGC-1α and which is controlled by the p38 MAPK signaling cascade.
Collapse
|
7
|
Bowman PRT, Smith GL, Gould GW. Run for your life: can exercise be used to effectively target GLUT4 in diabetic cardiac disease? PeerJ 2021; 9:e11485. [PMID: 34113491 PMCID: PMC8162245 DOI: 10.7717/peerj.11485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
The global incidence, associated mortality rates and economic burden of diabetes are now such that it is considered one of the most pressing worldwide public health challenges. Considerable research is now devoted to better understanding the mechanisms underlying the onset and progression of this disease, with an ultimate aim of improving the array of available preventive and therapeutic interventions. One area of particular unmet clinical need is the significantly elevated rate of cardiomyopathy in diabetic patients, which in part contributes to cardiovascular disease being the primary cause of premature death in this population. This review will first consider the role of metabolism and more specifically the insulin sensitive glucose transporter GLUT4 in diabetic cardiac disease, before addressing how we may use exercise to intervene in order to beneficially impact key functional clinical outcomes.
Collapse
Affiliation(s)
- Peter R T Bowman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
8
|
Klymenko O, Brecklinghaus T, Dille M, Springer C, de Wendt C, Altenhofen D, Binsch C, Knebel B, Scheller J, Hardt C, Herwig R, Chadt A, Pfluger PT, Al-Hasani H, Kabra DG. Histone deacetylase 5 regulates interleukin 6 secretion and insulin action in skeletal muscle. Mol Metab 2020; 42:101062. [PMID: 32771698 PMCID: PMC7481569 DOI: 10.1016/j.molmet.2020.101062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Physical exercise training is associated with increased glucose uptake in skeletal muscle and improved glycemic control. HDAC5, a class IIa histone deacetylase, has been shown to regulate transcription of the insulin-responsive glucose transporter GLUT4 in cultured muscle cells. In this study, we analyzed the contribution of HDAC5 to the transcriptional network in muscle and the beneficial effect of muscle contraction and regular exercise on glucose metabolism. METHODS HDAC5 knockout mice (KO) and wild-type (WT) littermates were trained for 8 weeks on treadmills, metabolically phenotyped, and compared to sedentary controls. Hdac5-deficient skeletal muscle and cultured Hdac5-knockdown (KD) C2C12 myotubes were utilized for studies of gene expression and glucose metabolism. Chromatin immunoprecipitation (ChIP) studies were conducted to analyze Il6 promoter activity using H3K9ac and HDAC5 antibodies. RESULTS Global transcriptome analysis of Hdac5 KO gastrocnemius muscle demonstrated activation of the IL-6 signaling pathway. Accordingly, knockdown of Hdac5 in C2C12 myotubes led to higher expression and secretion of IL-6 with enhanced insulin-stimulated activation of AKT that was reversed by Il6 knockdown. Moreover, Hdac5-deficient myotubes exhibited enhanced glucose uptake, glycogen synthesis, and elevated expression levels of the glucose transporter GLUT4. Transcription of Il6 was further enhanced by electrical pulse stimulation in Hdac5-deficient C2C12 myotubes. ChIP identified a ∼1 kb fragment of the Il6 promoter that interacts with HDAC5 and demonstrated increased activation-associated histone marker AcH3K9 in Hdac5-deficient muscle cells. Exercise intervention of HDAC5 KO mice resulted in improved systemic glucose tolerance as compared to WT controls. CONCLUSIONS We identified HDAC5 as a negative epigenetic regulator of IL-6 synthesis and release in skeletal muscle. HDAC5 may exert beneficial effects through two different mechanisms, transcriptional control of genes required for glucose disposal and utilization, and HDAC5-dependent IL-6 signaling cross-talk to improve glucose uptake in muscle in response to exercise.
Collapse
Affiliation(s)
- Oleksiy Klymenko
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Tim Brecklinghaus
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Matthias Dille
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christian Springer
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Christian de Wendt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Delsi Altenhofen
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Christian Binsch
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Birgit Knebel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Heinrich Heine University, Medical faculty, Düsseldorf, Germany
| | - Christopher Hardt
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, 81675, München, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Dhiraj G Kabra
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Medical faculty, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
9
|
Yoshihara T, Naito H. Protective effects of acute exercise preconditioning on disuse-induced muscular atrophy in aged muscle: a narrative literature review. J Physiol Sci 2020; 70:55. [PMID: 33246401 PMCID: PMC10717045 DOI: 10.1186/s12576-020-00783-w] [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: 06/12/2020] [Accepted: 11/19/2020] [Indexed: 11/10/2022]
Abstract
Aging is associated with a progressive loss of skeletal muscle mass and strength, resulting in frailty and lower quality of life in older individuals. At present, a standard of clinical or pharmacological care to prevent the adverse effects of aging does not exist. Determining the mechanism(s) responsible for muscular atrophy in disused aged muscle is a required key step for the development of effective countermeasures. Studies suggest an age-related differential response of genes and signalings to muscle disuse in both rodents and humans, implying the possibility that effective countermeasures to prevent disuse muscle atrophy may be age-specific. Notably, exercise preconditioning can attenuate disuse-induced muscular atrophy in rodent and human skeletal muscles; however, information on age-specific mechanisms of this exercise-induced protection remains limited. This mini-review aimed to summarize the protective effects of acute exercise preconditioning on muscular atrophy in aged muscle and provide potential mechanisms for its preventive effect on skeletal muscle wasting.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| |
Collapse
|
10
|
Abstract
The glucose transporter GLUT4 is critical for skeletal muscle glucose uptake in response to insulin and muscle contraction/exercise. Exercise increases GLUT4 translocation to the sarcolemma and t-tubule and, over the longer term, total GLUT4 protein content. Here, we review key aspects of GLUT4 biology in relation to exercise, with a focus on exercise-induced GLUT4 translocation, postexercise metabolism and muscle insulin sensitivity, and exercise effects on GLUT4 expression.
Collapse
Affiliation(s)
- Marcelo Flores-Opazo
- Laboratory of Exercise and Physical Activity Sciences, Department of Physiotherapy, University Finis Terrae, Santiago, Chile
| | - Sean L McGee
- Metabolic Research Unit, School of Medicine and Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds
| | - Mark Hargreaves
- Department of Physiology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
11
|
Sukumaran A, Choi K, Dasgupta B. Insight on Transcriptional Regulation of the Energy Sensing AMPK and Biosynthetic mTOR Pathway Genes. Front Cell Dev Biol 2020; 8:671. [PMID: 32903688 PMCID: PMC7438746 DOI: 10.3389/fcell.2020.00671] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
The Adenosine Monophosphate-activated Protein Kinase (AMPK) and the Mechanistic Target of Rapamycin (mTOR) are two evolutionarily conserved kinases that together regulate nearly every aspect of cellular and systemic metabolism. These two kinases sense cellular energy and nutrient levels that in turn are determined by environmental nutrient availability. Because AMPK and mTOR are kinases, the large majority of studies remained focused on downstream substrate phosphorylation by these two proteins, and how AMPK and mTOR regulate signaling and metabolism in normal and disease physiology through phosphorylation of their substrates. Compared to the wealth of information known about the signaling and metabolic pathways modulated by these two kinases, much less is known about how the transcription of AMPK and mTOR pathway genes themselves are regulated, and the extent to which AMPK and mTOR regulate gene expression to cause durable changes in phenotype. Acute modification of cellular systems can be achieved through phosphorylation, however, induction of chronic changes requires modulation of gene expression. In this review we will assemble evidence from published studies on transcriptional regulation by AMPK and mTOR and discuss about the putative transcription factors that regulate expression of AMPK and mTOR complex genes.
Collapse
Affiliation(s)
- Abitha Sukumaran
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Biplab Dasgupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
12
|
Adaptations in GLUT4 Expression in Response to Exercise Detraining Linked to Downregulation of Insulin-Dependent Pathways in Cardiac but not in Skeletal Muscle Tissue. Int J Sport Nutr Exerc Metab 2020; 30:272-279. [PMID: 32454459 DOI: 10.1123/ijsnem.2019-0337] [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: 11/19/2019] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 11/18/2022]
Abstract
Insulin resistance is associated with cardiometabolic risk factors, and exercise training can improve insulin-mediated glucose uptake. However, few studies have demonstrated the reversibility of exercise-induced benefits. Thus, the authors examine the time-response effects of exercise training and detraining on glucose transporter 4 (GLUT4) content, insulin-dependent and insulin-independent pathways in cardiac and gastrocnemius muscle tissues of spontaneously hypertensive rats. Thirty-two male spontaneously hypertensive rats, 4 months old, were assigned to (n = 8/group): T (exercise training: 10-week treadmill exercise, 50-70% maximum effort capacity, 1 hr/day, 5 days/week); D2 (exercise training + 2-day detraining), D4 (exercise training + 4-day detraining); and S (no exercise). The authors evaluated insulin resistance, maximum effort capacity, GLUT4 content, p-IRS-1Tyr1179, p-AS160Ser588, p-AMPKα1Thr172, and p-CaMKIIThr286 in cardiac and gastrocnemius muscle tissues (Western blot). In response to exercise training, there were improvements in insulin resistance (15.4%; p = .010), increased GLUT4 content (microsomal, 29.4%; p = .012; plasma membrane, 27.1%; p < .001), p-IRS-1 (42.2%; p < .001), p-AS160 (60.0%; p < .001) in cardiac tissue, and increased GLUT4 content (microsomal, 29.4%; p = .009; plasma membrane, 55.5%; p < .001), p-IRS-1 (28.1%; p = .018), p-AS160 (76.0%; p < .001), p-AMPK-α1 (37.5%; p = .026), and p-CaMKII (30.0%; p = .040) in the gastrocnemius tissue. In D4 group, the exercise-induced increase in GLUT4 was reversed (plasma membrane, -21.3%; p = .027), p-IRS1 (-37.1%; p = .008), and p-AS160 (-82.6%; p < .001) in the cardiac tissue; p-AS160 expression (-35.7%; p = .034) was reduced in the gastrocnemius. In conclusion, the cardiac tissue is more susceptible to exercise adaptations in the GLUT4 content and signaling pathways than the gastrocnemius muscle. This finding may be explained by particular characteristics of insulin-dependent and insulin-independent pathways in the muscle tissues studied.
Collapse
|
13
|
Honda M, Tsuchimochi H, Hitachi K, Ohno S. Transcriptional cofactor Vgll2 is required for functional adaptations of skeletal muscle induced by chronic overload. J Cell Physiol 2019; 234:15809-15824. [PMID: 30724341 DOI: 10.1002/jcp.28239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
Skeletal muscle is composed of heterogeneous populations of myofibers classified as slow- and fast-twitch fibers. Myofiber size and composition are drastically changed in response to physiological demands. We previously showed that transcriptional cofactor vestigial-like (Vgll) 2 is a pivotal regulator of slow muscle gene programming under sedentary conditions. However, whether Vgll2 is required for skeletal muscle adaptations after chronic overload is unclear. Therefore, we investigated the role of Vgll2 in chronic overload-inducing skeletal muscle adaptations using synergist ablation (SA) on plantaris. We found that Vgll2 is an essential regulator of the switch towards a slow-contractile phenotype and oxidative metabolism during chronic overload. Mice lacking Vgll2 exhibited limited fiber type transition and downregulation of genes related to lactate metabolism and their regulator peroxisome proliferator-activated receptor gamma coactivator 1α1, after SA, was augmented in Vgll2-deficient mice compared with in wild-type mice. Mechanistically, increased muscle usage elevated Vgll2 levels and promoted the interaction between Vgll2 and its transcription partners such as TEA domain1 (TEAD1), MEF2c, and NFATc1. Calcium ionophore treatment promoted nuclear translocation of Vgll2 and increased TEAD-dependent MYH7 promotor activity in a Vgll2-dependent manner. Taken together, these data demonstrate that Vgll2 plays an important role for functional adaptation of skeletal muscle to chronic overload.
Collapse
Affiliation(s)
- Masahiko Honda
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Centre Research Institute, Suita, Osaka, Japan
| | - Keisuke Hitachi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| |
Collapse
|
14
|
NURR1 activation in skeletal muscle controls systemic energy homeostasis. Proc Natl Acad Sci U S A 2019; 116:11299-11308. [PMID: 31110021 DOI: 10.1073/pnas.1902490116] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Skeletal muscle plays a central role in the control of metabolism and exercise tolerance. Analysis of muscle enhancers activated after exercise in mice revealed the orphan nuclear receptor NURR1/NR4A2 as a prominent component of exercise-responsive enhancers. We show that exercise enhances the expression of NURR1, and transgenic overexpression of NURR1 in skeletal muscle enhances physical performance in mice. NURR1 expression in skeletal muscle is also sufficient to prevent hyperglycemia and hepatic steatosis, by enhancing muscle glucose uptake and storage as glycogen. Furthermore, treatment of obese mice with putative NURR1 agonists increases energy expenditure, improves glucose tolerance, and confers a lean phenotype, mimicking the effects of exercise. These findings identify a key role for NURR1 in governance of skeletal muscle glucose metabolism, and reveal a transcriptional link between exercise and metabolism. Our findings also identify NURR1 agonists as possible exercise mimetics with the potential to ameliorate obesity and other metabolic abnormalities.
Collapse
|
15
|
Koh JH, Hancock CR, Han DH, Holloszy JO, Nair KS, Dasari S. AMPK and PPARβ positive feedback loop regulates endurance exercise training-mediated GLUT4 expression in skeletal muscle. Am J Physiol Endocrinol Metab 2019; 316:E931-E939. [PMID: 30888859 PMCID: PMC6580175 DOI: 10.1152/ajpendo.00460.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The objective of this study is to determine whether AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), or peroxisome proliferator-activated receptor β (PPARβ) can independently mediate the increase of glucose transporter type 4 (GLUT4) expression that occurs in response to exercise training. We found that PPARβ can regulate GLUT4 expression without PGC-1α. We also found AMPK and PPARβ are important for maintaining normal physiological levels of GLUT4 protein in the sedentary condition as well following exercise training. However, AMPK and PPARβ are not essential for the increase in GLUT4 protein expression that occurs in response to exercise training. We discovered that AMPK activation increases PPARβ via myocyte enhancer factor 2A (MEF2A), which acted as a transcription factor for PPARβ. Furthermore, exercise training increases the cooperation of AMPK and PPARβ to regulate glucose uptake. In conclusion, cooperation between AMPK and PPARβ via NRF-1/MEF2A pathway enhances the exercise training mediated adaptive increase in GLUT4 expression and subsequent glucose uptake in skeletal muscle.
Collapse
Affiliation(s)
- Jin-Ho Koh
- Department of Internal Medicine, Mayo Clinic , Rochester, Minnesota
- Department of Physiology, College of Medicine, Yeungnam University , Daegu , Korea
| | - Chad R Hancock
- Department of Nutrition, Dietetics and Food Science, Brigham Young University , Provo, Utah
| | - Dong-Ho Han
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | - John O Holloszy
- Division of Geriatrics and Nutritional Sciences, Department of Medicine, Washington University School of Medicine , St. Louis, Missouri
| | | | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
16
|
Yoshihara T, Tsuzuki T, Chang SW, Kakigi R, Sugiura T, Naito H. Exercise preconditioning attenuates hind limb unloading-induced gastrocnemius muscle atrophy possibly via the HDAC4/Gadd45 axis in old rats. Exp Gerontol 2019; 122:34-41. [PMID: 31009659 DOI: 10.1016/j.exger.2019.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 11/25/2022]
Abstract
The mechanisms involved in unloading-induced skeletal muscle loss may be age-specific, and the evidence for exercise preconditioning-induced protection against disuse muscle atrophy in aged rats is limited. Therefore, in this study, we investigated age-related differences in the activation of the HDAC4/Gadd45α pathway following hindlimb unloading (HU). We also assessed the protective effect of preconditioning exercise on this pathway in young and old rat gastrocnemius muscle. Three-month-old (young, n = 18) and 24-month-old (old, n = 18) male Wistar rats were assigned to the following groups: control group (n = 6), seven days of HU group (n = 6), and a bout of exercise preconditioning prior to HU (Ex+HU) group (n = 6). Rats of both ages in the Ex + HU group ran continuously on a motor-driven treadmill (0° slope, 20 m/min, 15 min) prior to HU. The gastrocnemius muscles were removed after 7 days of HU and analyzed for protein content and mRNA expression. Gastrocnemius muscle weight was significantly higher in the Ex+HU group than in the HU group of old rats, but not in young rats. Levels of HDAC4 protein and mRNA were significantly increased in the old HU group. However, the increase was significantly suppressed in the old Ex+HU group. Moreover, the protective effect of exercise preconditioning had a positive effect on Gadd45α mRNA and protein levels only in the old Ex+HU group. No exercise preconditioning-related protection was observed in the young rats. Our data indicated that a single bout of preconditioning exercise prior to HU may exert a protective effect in disuse muscle atrophy in old rats and that these effects may be partially mediated by the HDAC4/Gadd45α axis.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| | - Takamasa Tsuzuki
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan.
| | - Shuo-Wen Chang
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| | - Ryo Kakigi
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Takao Sugiura
- Faculty of Education, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8513, Japan.
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| |
Collapse
|
17
|
Kjøbsted R, Hingst JR, Fentz J, Foretz M, Sanz MN, Pehmøller C, Shum M, Marette A, Mounier R, Treebak JT, Wojtaszewski JFP, Viollet B, Lantier L. AMPK in skeletal muscle function and metabolism. FASEB J 2018; 32:1741-1777. [PMID: 29242278 PMCID: PMC5945561 DOI: 10.1096/fj.201700442r] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle possesses a remarkable ability to adapt to various physiologic conditions. AMPK is a sensor of intracellular energy status that maintains energy stores by fine-tuning anabolic and catabolic pathways. AMPK’s role as an energy sensor is particularly critical in tissues displaying highly changeable energy turnover. Due to the drastic changes in energy demand that occur between the resting and exercising state, skeletal muscle is one such tissue. Here, we review the complex regulation of AMPK in skeletal muscle and its consequences on metabolism (e.g., substrate uptake, oxidation, and storage as well as mitochondrial function of skeletal muscle fibers). We focus on the role of AMPK in skeletal muscle during exercise and in exercise recovery. We also address adaptations to exercise training, including skeletal muscle plasticity, highlighting novel concepts and future perspectives that need to be investigated. Furthermore, we discuss the possible role of AMPK as a therapeutic target as well as different AMPK activators and their potential for future drug development.—Kjøbsted, R., Hingst, J. R., Fentz, J., Foretz, M., Sanz, M.-N., Pehmøller, C., Shum, M., Marette, A., Mounier, R., Treebak, J. T., Wojtaszewski, J. F. P., Viollet, B., Lantier, L. AMPK in skeletal muscle function and metabolism.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Fentz
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marc Foretz
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria-Nieves Sanz
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Christian Pehmøller
- Internal Medicine Research Unit, Pfizer Global Research and Development, Cambridge, Massachusetts, USA
| | - Michael Shum
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - André Marette
- Axe Cardiologie, Quebec Heart and Lung Research Institute, Laval University, Québec, Canada.,Institute for Nutrition and Functional Foods, Laval University, Québec, Canada
| | - Remi Mounier
- Institute NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM Unité 1217, CNRS UMR, Villeurbanne, France
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Benoit Viollet
- INSERM, Unité 1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
18
|
Xian HM, Che H, Qin Y, Yang F, Meng SY, Li XG, Bai YL, Wang LH. Coriolus versicolor aqueous extract ameliorates insulin resistance with PI3K/Akt and p38 MAPK signaling pathways involved in diabetic skeletal muscle. Phytother Res 2017; 32:551-560. [PMID: 29243310 DOI: 10.1002/ptr.6007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/13/2017] [Accepted: 11/13/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Hui-min Xian
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Hui Che
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Ying Qin
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Department of Pharmacology; College of Pharmacy, Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Fan Yang
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Song-yan Meng
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Xiao-guang Li
- Academician Workstation; Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Yun-long Bai
- Department of Pharmacology; College of Pharmacy, Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Institute of Chronic Disease; Heilongjiang Academy of Medical Science; Harbin Heilongjiang Province 150001 China
| | - Li-hong Wang
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Institute of Chronic Disease; Heilongjiang Academy of Medical Science; Harbin Heilongjiang Province 150001 China
| |
Collapse
|
19
|
Chen X, Gao B, Ponnusamy M, Lin Z, Liu J. MEF2 signaling and human diseases. Oncotarget 2017; 8:112152-112165. [PMID: 29340119 PMCID: PMC5762387 DOI: 10.18632/oncotarget.22899] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 09/09/2017] [Indexed: 01/01/2023] Open
Abstract
The members of myocyte Enhancer Factor 2 (MEF2) protein family was previously believed to function in the development of heart and muscle. Recent reports indicate that they are also closely associated with development and progression of many human diseases. Although their role in cancer biology is well established, the molecular mechanisms underlying their action is yet largely unknown. MEF2 family is closely associated with various signaling pathways, including Ca2+ signaling, MAP kinase signaling, Wnt signaling, PI3K/Akt signaling, etc. microRNAs also contribute to regulate the activities of MEF2. In this review, we summarize the known molecular mechanism by which MEF2 family contribute to human diseases.
Collapse
Affiliation(s)
- Xiao Chen
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Bing Gao
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Zhijuan Lin
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Jia Liu
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
20
|
Lamon S, Zacharewicz E, Butchart LC, Orellana L, Mikovic J, Grounds MD, Russell AP. MicroRNA expression patterns in post-natal mouse skeletal muscle development. BMC Genomics 2017; 18:52. [PMID: 28061746 PMCID: PMC5219731 DOI: 10.1186/s12864-016-3399-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/08/2016] [Indexed: 01/28/2023] Open
Abstract
Background MiRNAs are essential regulators of skeletal muscle development and homeostasis. To date, the role and regulation of miRNAs in myogenesis have been mostly studied in tissue culture and during embryogenesis. However, little information relating to miRNA regulation during early post-natal skeletal muscle growth in mammals is available. Using a high-throughput miRNA qPCR-based array, followed by stringent statistical and bioinformatics analysis, we describe the expression pattern and putative role of 768 miRNAs in the quadriceps muscle of mice aged 2 days, 2 weeks, 4 weeks and 12 weeks. Results Forty-six percent of all measured miRNAs were expressed in mouse quadriceps muscle during the first 12 weeks of life. We report unprecedented changes in miRNA expression levels over time. The expression of a majority of miRNAs significantly decreased with post-natal muscle maturation in vivo. MiRNA clustering identified 2 subsets of miRNAs that are potentially involved in cell proliferation and differentiation, mainly via the regulation of non-muscle specific targets. Conclusion Collective miRNA expression in mouse quadriceps muscle is subjected to substantial levels of regulation during the first 12 weeks of age. This study identified a new suite of highly conserved miRNAs that are predicted to influence early muscle development. As such it provides novel knowledge pertaining to post-natal myogenesis and muscle regeneration in mammals. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3399-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Séverine Lamon
- Deakin University, School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (I-PAN), Geelong, Australia.
| | - Evelyn Zacharewicz
- Deakin University, School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (I-PAN), Geelong, Australia
| | - Lauren C Butchart
- The University of Western Australia, School of Anatomy, Physiology and Human Biology, Perth, WA, Australia
| | - Liliana Orellana
- Deakin University, Biostatistics Unit, Faculty of Health, Geelong, Australia
| | - Jasmine Mikovic
- Deakin University, School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (I-PAN), Geelong, Australia
| | - Miranda D Grounds
- The University of Western Australia, School of Anatomy, Physiology and Human Biology, Perth, WA, Australia
| | - Aaron P Russell
- Deakin University, School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (I-PAN), Geelong, Australia
| |
Collapse
|
21
|
Pereira RM, Moura LPD, Muñoz VR, Silva ASRD, Gaspar RS, Ropelle ER, Pauli JR. Molecular mechanisms of glucose uptake in skeletal muscle at rest and in response to exercise. MOTRIZ: REVISTA DE EDUCACAO FISICA 2017. [DOI: 10.1590/s1980-6574201700si0004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
| | | | | | | | | | | | - José Rodrigo Pauli
- Universidade Estadual de Campinas, Brazil; Universidade Estadual de Campinas, Brazil
| |
Collapse
|
22
|
Close GL, Hamilton DL, Philp A, Burke LM, Morton JP. New strategies in sport nutrition to increase exercise performance. Free Radic Biol Med 2016; 98:144-158. [PMID: 26855422 DOI: 10.1016/j.freeradbiomed.2016.01.016] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 02/03/2023]
Abstract
Despite over 50 years of research, the field of sports nutrition continues to grow at a rapid rate. Whilst the traditional research focus was one that centred on strategies to maximise competition performance, emerging data in the last decade has demonstrated how both macronutrient and micronutrient availability can play a prominent role in regulating those cell signalling pathways that modulate skeletal muscle adaptations to endurance and resistance training. Nonetheless, in the context of exercise performance, it is clear that carbohydrate (but not fat) still remains king and that carefully chosen ergogenic aids (e.g. caffeine, creatine, sodium bicarbonate, beta-alanine, nitrates) can all promote performance in the correct exercise setting. In relation to exercise training, however, it is now thought that strategic periods of reduced carbohydrate and elevated dietary protein intake may enhance training adaptations whereas high carbohydrate availability and antioxidant supplementation may actually attenuate training adaptation. Emerging evidence also suggests that vitamin D may play a regulatory role in muscle regeneration and subsequent hypertrophy following damaging forms of exercise. Finally, novel compounds (albeit largely examined in rodent models) such as epicatechins, nicotinamide riboside, resveratrol, β-hydroxy β-methylbutyrate, phosphatidic acid and ursolic acid may also promote or attenuate skeletal muscle adaptations to endurance and strength training. When taken together, it is clear that sports nutrition is very much at the heart of the Olympic motto, Citius, Altius, Fortius (faster, higher, stronger).
Collapse
Affiliation(s)
- G L Close
- Research Institute for Sport and Exercise Science (RISES), Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool L3 3AF, United Kingdom.
| | - D L Hamilton
- Health and Exercise Sciences Research Group, University of Stirling, Stirling, United Kingdom
| | - A Philp
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - L M Burke
- Sports Nutrition, Australian Institute of Sport, Canberra, ACT, Australia; Mary Mackillop Institute for Health Research, Melbourne, Australia
| | - J P Morton
- Research Institute for Sport and Exercise Science (RISES), Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| |
Collapse
|
23
|
Gaur V, Connor T, Sanigorski A, Martin S, Bruce C, Henstridge D, Bond S, McEwen K, Kerr-Bayles L, Ashton T, Fleming C, Wu M, Pike Winer L, Chen D, Hudson G, Schwabe J, Baar K, Febbraio M, Gregorevic P, Pfeffer F, Walder K, Hargreaves M, McGee S. Disruption of the Class IIa HDAC Corepressor Complex Increases Energy Expenditure and Lipid Oxidation. Cell Rep 2016; 16:2802-2810. [DOI: 10.1016/j.celrep.2016.08.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 06/20/2016] [Accepted: 07/31/2016] [Indexed: 10/21/2022] Open
|
24
|
Yoshihara T, Machida S, Kurosaka Y, Kakigi R, Sugiura T, Naito H. Immobilization induces nuclear accumulation of HDAC4 in rat skeletal muscle. J Physiol Sci 2016; 66:337-43. [PMID: 26759025 PMCID: PMC10717107 DOI: 10.1007/s12576-015-0432-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/13/2015] [Indexed: 11/25/2022]
Abstract
The study described herein aimed to examine changes in HDAC4 and its downstream targets in immobilization-induced rat skeletal muscle atrophy. Eleven male Wistar rats were used, and one hindlimb was immobilized in the plantar flexion position using a plaster cast. The contralateral, non-immobilized leg served as an internal control. After 10 days, the gastrocnemius muscles were removed from both hindlimbs. Ten days of immobilization resulted in a significant reduction (-27.3 %) in gastrocnemius muscle weight. A significant decrease in AMPK phosphorylation was also observed in nuclear fractions from immobilized legs relative to the controls. HDAC4 expression was significantly increased in immobilized legs in both the cytoplasmic and nuclear fractions. Moreover, Myogenin and MyoD mRNA levels were upregulated in immobilized legs, resulting in increased Atrogin-1 mRNA expression. Our data suggest that nuclear HDAC4 accumulation is partly related to immobilization-induced muscle atrophy.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Yuka Kurosaka
- Faculty of Human Ecology, Wayo Women's University, 2-3-1 konodai, Ichikawa, Chiba, 272-8533, Japan
| | - Ryo Kakigi
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takao Sugiura
- Faculty of Education, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, Yamaguchi, 753-8513, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| |
Collapse
|
25
|
trans-Cinnamaldehyde stimulates mitochondrial biogenesis through PGC-1α and PPARβ/δ leading to enhanced GLUT4 expression. Biochimie 2015; 119:45-51. [DOI: 10.1016/j.biochi.2015.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/01/2015] [Indexed: 11/22/2022]
|
26
|
Olson AL. Insulin resistance: cross-talk between adipose tissue and skeletal muscle, through free fatty acids, liver X receptor, and peroxisome proliferator-activated receptor-α signaling. Horm Mol Biol Clin Investig 2015; 15:115-21. [PMID: 25436738 DOI: 10.1515/hmbci-2013-0019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/18/2013] [Indexed: 11/15/2022]
Abstract
Skeletal muscle and adipose tissue play a major role in the regulation of whole-body glucose homeostasis. Much of the coordinated regulation of whole-body glucose homeostasis results from the regulation of lipid storage and release by adipose tissue and efficient switching between glucose oxidation and fatty acid oxidation in skeletal muscle. A control point for these biochemical actions center around the regulation of the insulin responsive glucose transporter, GLUT4. This review examines the regulation of GLUT4 in adipose tissue and skeletal muscle, in the context of the steroid nuclear hormone receptor signaling.
Collapse
Affiliation(s)
- Ann Louise Olson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, P. O. Box 26901, BMSB 964, Oklahoma City, OK 73190, USA.
| |
Collapse
|
27
|
Abstract
Acute and transient changes in gene transcription following a single exercise bout, if reinforced by repeated exercise stimuli, result in the longer lasting effects on protein expression and function that form the basis of skeletal muscle training adaptations. Changes in skeletal muscle gene expression occur in response to multiple stimuli associated with skeletal muscle contraction, various signaling kinases that respond to these stimuli, and numerous downstream pathways and targets of these kinases. In addition, DNA methylation, histone acetylation and phosphorylation, and micro-RNAs can alter gene expression via epigenetic mechanisms. Contemporary studies rely upon "big omics data," in combination with computational and systems biology, to interrogate, and make sense of, the complex interactions underpinning exercise adaptations. The exciting potential is a greater understanding of the integrative biology of exercise.
Collapse
Affiliation(s)
- Mark Hargreaves
- Department of Physiology, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
28
|
Pecorella SRH, Potter JVF, Cherry AD, Peacher DF, Welty-Wolf KE, Moon RE, Piantadosi CA, Suliman HB. The HO-1/CO system regulates mitochondrial-capillary density relationships in human skeletal muscle. Am J Physiol Lung Cell Mol Physiol 2015; 309:L857-71. [PMID: 26186946 DOI: 10.1152/ajplung.00104.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/14/2015] [Indexed: 12/14/2022] Open
Abstract
The heme oxygenase-1 (HO-1)/carbon monoxide (CO) system induces mitochondrial biogenesis, but its biological impact in human skeletal muscle is uncertain. The enzyme system generates CO, which stimulates mitochondrial proliferation in normal muscle. Here we examined whether CO breathing can be used to produce a coordinated metabolic and vascular response in human skeletal muscle. In 19 healthy subjects, we performed vastus lateralis muscle biopsies and tested one-legged maximal O2 uptake (V̇o2max) before and after breathing air or CO (200 ppm) for 1 h daily for 5 days. In response to CO, there was robust HO-1 induction along with increased mRNA levels for nuclear-encoded mitochondrial transcription factor A (Tfam), cytochrome c, cytochrome oxidase subunit IV (COX IV), and mitochondrial-encoded COX I and NADH dehydrogenase subunit 1 (NDI). CO breathing did not increase V̇o2max (1.96 ± 0.51 pre-CO, 1.87 ± 0.50 post-CO l/min; P = not significant) but did increase muscle citrate synthase, mitochondrial density (139.0 ± 34.9 pre-CO, 219.0 ± 36.2 post-CO; no. of mitochondrial profiles/field), myoglobin content and glucose transporter (GLUT4) protein level and led to GLUT4 localization to the myocyte membrane, all consistent with expansion of the tissue O2 transport system. These responses were attended by increased cluster of differentiation 31 (CD31)-positive muscle capillaries (1.78 ± 0.16 pre-CO, 2.37 ± 0.59 post-CO; capillaries/muscle fiber), implying the enrichment of microvascular O2 reserve. The findings support that induction of the HO-1/CO system by CO not only improves muscle mitochondrial density, but regulates myoglobin content, GLUT4 localization, and capillarity in accordance with current concepts of skeletal muscle plasticity.
Collapse
Affiliation(s)
- Shelly R H Pecorella
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| | - Jennifer V F Potter
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| | - Anne D Cherry
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| | - Dionne F Peacher
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| | - Karen E Welty-Wolf
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Richard E Moon
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Department of Medicine, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| | - Claude A Piantadosi
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina; Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina; and
| |
Collapse
|
29
|
Chen S, Yin C, Lao T, Liang D, He D, Wang C, Sang N. AMPK-HDAC5 pathway facilitates nuclear accumulation of HIF-1α and functional activation of HIF-1 by deacetylating Hsp70 in the cytosol. Cell Cycle 2015; 14:2520-36. [PMID: 26061431 PMCID: PMC4614078 DOI: 10.1080/15384101.2015.1055426] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) transcriptionally promotes production of adenosine triphosphate (ATP) whereas AMPK senses and regulates cellular energy homeostasis. A histone deacetylase (HDAC) activity has been proven to be critical for HIF-1 activation but the underlying mechanism and its role in energy homesostasis remain unclear. Here, we demonstrate that HIF-1 activation depends on a cytosolic, enzymatically active HDAC5. HDAC5 knockdown impairs hypoxia-induced HIF-1α accumulation and HIF-1 transactivation, whereas HDAC5 overexpression enhances HIF-1α stabilization and nuclear translocation. Mechanistically, we show that Hsp70 is a cytosolic substrate of HDAC5; and hyperacetylation renders Hsp70 higher affinity for HIF-1α binding, which correlates with accelerated degradation and attenuated nuclear accumulation of HIF-1α. Physiologically, AMPK-triggered cytosolic shuttling of HDAC5 is critical; inhibition of either AMPK or HDAC5 impairs HIF-1α nuclear accumulation under hypoxia or low glucose conditions. Finally, we show specifically suppressing HDAC5 is sufficient to inhibit tumor cell proliferation under hypoxic conditions. Our data delineate a novel link between AMPK, the energy sensor, and HIF-1, the major driver of ATP production, indicating that specifically inhibiting HDAC5 may selectively suppress the survival and proliferation of hypoxic tumor cells.
Collapse
Affiliation(s)
- Shuyang Chen
- a Department of Biology and Graduate Program of Biological Sciences; CoAS; Department of Pathology & Laboratory Medicine; DUCOM; Drexel University ; Philadelphia , PA USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Steinbacher P, Feichtinger RG, Kedenko L, Kedenko I, Reinhardt S, Schönauer AL, Leitner I, Sänger AM, Stoiber W, Kofler B, Förster H, Paulweber B, Ring-Dimitriou S. The single nucleotide polymorphism Gly482Ser in the PGC-1α gene impairs exercise-induced slow-twitch muscle fibre transformation in humans. PLoS One 2015; 10:e0123881. [PMID: 25886402 PMCID: PMC4401702 DOI: 10.1371/journal.pone.0123881] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/23/2015] [Indexed: 12/13/2022] Open
Abstract
PGC-1α (peroxisome proliferator-activated receptor γ co-activator 1α) is an important regulator of mitochondrial biogenesis and a master regulator of enzymes involved in oxidative phosphorylation. Recent evidence demonstrated that the Gly482Ser single nucleotide polymorphism (SNP) in the PGC-1α gene affects insulin sensitivity, blood lipid metabolism and binding to myocyte enhancer factor 2 (MEF2). Individuals carrying this SNP were shown to have a reduced cardiorespiratory fitness and a higher risk to develop type 2 diabetes. Here, we investigated the responses of untrained men with the Gly482Ser SNP to a 10 week programme of endurance training (cycling, 3 x 60 min/week, heart rate at 70-90% VO2peak). Quantitative data from analysis of biopsies from vastus lateralis muscle revealed that the SNP group, in contrast to the control group, lacked a training-induced increase in content of slow contracting oxidative fibres. Capillary supply, mitochondrial density, mitochondrial enzyme activities and intramyocellular lipid content increased similarly in both groups. These results indicate that the impaired binding of MEF2 to PGC-1α in humans with this SNP impedes exercise-induced fast-to-slow muscle fibre transformation.
Collapse
Affiliation(s)
- Peter Steinbacher
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
- * E-mail:
| | - René G. Feichtinger
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pedicatrics, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Lyudmyla Kedenko
- First Department of Internal Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Igor Kedenko
- First Department of Internal Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Sandra Reinhardt
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
| | - Anna-Lena Schönauer
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
| | - Isabella Leitner
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
| | - Alexandra M. Sänger
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
| | - Walter Stoiber
- Department of Cell Biology, Paris Lodron-University of Salzburg, Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pedicatrics, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Holger Förster
- Medical Office in Pediatrics and Sports Medicine, Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Susanne Ring-Dimitriou
- Department of Sport Science and Kinesiology, Paris Lodron-University of Salzburg, Hallein, Austria
| |
Collapse
|
31
|
Lee WH, Wu HH, Huang WJ, Li YN, Lin RJ, Lin SY, Liang YC. N-hydroxycinnamide derivatives of osthole ameliorate hyperglycemia through activation of AMPK and p38 MAPK. Molecules 2015; 20:4516-29. [PMID: 25768846 PMCID: PMC6272315 DOI: 10.3390/molecules20034516] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/04/2015] [Accepted: 03/05/2015] [Indexed: 01/19/2023] Open
Abstract
Our previous studies found that osthole markedly reduced blood glucose levels in both db/db and ob/ob mice. To improve the antidiabetic activity of osthole, a series of N-hydroxycinnamide derivatives of osthole were synthesized, and their hypoglycemia activities were examined in vitro and in vivo. Both N-hydroxycinnamide derivatives of osthole, OHC-4p and OHC-2m, had the greatest potential for activating AMPK and increasing glucose uptake by L6 skeletal muscle cells. In addition, OHC-4p and OHC-2m time- and dose-dependently increased phosphorylation levels of AMPK and p38 MAPK. The AMPK inhibitor, compound C, and the p38 MAPK inhibitor, SB203580, significantly reversed activation of AMPK and p38 MAPK, respectively, in OHC-4p- and OHC-2m-treated cells. Compound C and SB203580 also inhibited glucose uptake induced by OHC-4p and OHC-2m. Next, we found that OHC-4p and OHC-2m significantly increased glucose transporter 4 (GLUT4) translocation to plasma membranes and counteracted hyperglycemia in mice with streptozotocin-induced diabetes. These results suggest that activation of AMPK and p38 MAPK by OHC-4p and OHC-2m is associated with increased glucose uptake and GLUT4 translocation and subsequently led to amelioration of hyperglycemia. Therefore, OHC-4p and OHC-2m might have potential as antidiabetic agents for treating type 2 diabetes. Our previous studies found that osthole markedly reduced blood glucose levels in both db/db and ob/ob mice. To improve the antidiabetic activity of osthole, a series of N-hydroxycinnamide derivatives of osthole were synthesized, and their hypoglycemia activities were examined in vitro and in vivo. Both N-hydroxycinnamide derivatives of osthole, OHC-4p and OHC-2m, had the greatest potential for activating AMPK and increasing glucose uptake by L6 skeletal muscle cells. In addition, OHC-4p and OHC-2m time- and dose-dependently increased phosphorylation levels of AMPK and p38 MAPK. The AMPK inhibitor, compound C, and the p38 MAPK inhibitor, SB203580, significantly reversed activation of AMPK and p38 MAPK, respectively, in OHC-4p- and OHC-2m-treated cells. Compound C and SB203580 also inhibited glucose uptake induced by OHC-4p and OHC-2m. Next, we found that OHC-4p and OHC-2m significantly increased glucose transporter 4 (GLUT4) translocation to plasma membranes and counteracted hyperglycemia in mice with streptozotocin-induced diabetes. These results suggest that activation of AMPK and p38 MAPK by OHC-4p and OHC-2m is associated with increased glucose uptake and GLUT4 translocation and subsequently led to amelioration of hyperglycemia. Therefore, OHC-4p and OHC-2m might have potential as antidiabetic agents for treating type 2 diabetes.
Collapse
Affiliation(s)
- Wei-Hwa Lee
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, 291 Zhongzheng Rd., New Taipei City 23561, Taiwan.
| | - Hsueh-Hsia Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| | - Wei-Jan Huang
- Graduate Institute of Pharmacognosy Science, College of Pharmacy, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| | - Yi-Ning Li
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| | - Ren-Jye Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, 252 Wuxing St., Taipei 11031, Taiwan.
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, 252 Wuxing St., Taipei 11031, Taiwan.
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| | - Yu-Chih Liang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, 252 Wuxing St., Taipei 11031, Taiwan.
| |
Collapse
|
32
|
Raciti GA, Nigro C, Longo M, Parrillo L, Miele C, Formisano P, Béguinot F. Personalized medicine and type 2 diabetes: lesson from epigenetics. Epigenomics 2015; 6:229-38. [PMID: 24811791 DOI: 10.2217/epi.14.10] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Similarly to genetic polymorphisms, epigenetic modifications may alter transcriptional activity and contribute to different traits of the Type 2 diabetes phenotype. The establishment of these epigenetic marks may precede diabetes onset and predict the disease. Current evidence now indicates that epigenetic differences represent markers of diabetes risk. Studies on epigenome plasticity revealed that cytokines and other metabolites, by affecting DNA methylation, may acutely reprogram gene expression and contribute to the Type 2 diabetes phenotype even in the adult life. The available evidence further indicates that epigenetic marks across the genome are subject to dynamic variations in response to environmental cues. Finally, different genes responsible for the interindividual variability in antidiabetic drug response are subjected to epigenetic regulation. Determining how specific epigenetic profiles determine diabetes is a challenging task. In the near future, the identification of epigenetic marks predictive of diabetes risk or response to treatment may offer unanticipated opportunities to personalize Type 2 diabetes management.
Collapse
Affiliation(s)
- Gregory Alexander Raciti
- Dipartimento di Scienze Mediche Traslazionali, 'Federico II' University of Naples Medical School & Istituto per l' Endocrinologia e l' Oncologia Sperimentale del CNR, Via Sergio Pansini, 5 - Naples, 80131, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
Russell AP, Lamon S. Exercise, Skeletal Muscle and Circulating microRNAs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 135:471-96. [DOI: 10.1016/bs.pmbts.2015.07.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Yuan H, Niu Y, Liu X, Fu L. Exercise increases the binding of MEF2A to the Cpt1b promoter in mouse skeletal muscle. Acta Physiol (Oxf) 2014; 212:283-92. [PMID: 25213552 DOI: 10.1111/apha.12395] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/09/2014] [Accepted: 09/09/2014] [Indexed: 12/11/2022]
Abstract
AIM Muscle-type carnitine palmitoyltransferase 1 (CPT1b) plays an important role in skeletal muscle mitochondrial fatty acid β-oxidation. A myocyte-specific enhancer factor (MEF2) site that binds MEF2A in the promoter of Cpt1b has been identified. The aim of this study is to determine whether Cpt1b expression is regulated by MEF2 in response to exercise training. METHODS Twelve male, 14-week-old C57BL/6 mice underwent 6 weeks of treadmill exercise at 12 m min(-1), for 60 min day(-1), 5 days week(-1). Quadriceps muscles were analysed by real-time PCR, Western blot, chromatin immunoprecipitation. RESULTS The binding of MEF2A to the Cpt1b promoter was elevated in the quadriceps muscle of mice after 6-week aerobic exercise intervention. The repressor partner of MEF2, histone deacetylase 5 (HDAC5), binding to the Cpt1b promoter was decreased by exercise training. In C2C12 myoblast, MEF2A overexpression increased the Cpt1b mRNA expression and the Cpt1b promoter transcriptional activity, which were suppressed by HDAC5. Moreover, exercise training induced MEF2A hyperacetylation, which is strongly associated with its enhanced DNA-binding ability. The binding of both HDAC5 and histone deacetylase 3 (HDAC3) to MEF2A was decreased by exercise training. Overexpression of HDAC5 decreased exogenous MEF2A acetylation level with an increased binding of endogenous HDAC3 to MEF2A. Finally, the phosphorylation levels of HDAC5 Ser259 and Ser498 were increased by exercise training. Exercise training decreased the abundance of HDAC5 in the nucleus while increasing its abundance in the cytoplasm. CONCLUSION Our results indicated that exercise-induced CPT1b expression was at least in part mediated by HDAC5/MEF2A interaction.
Collapse
Affiliation(s)
- H. Yuan
- Department of Physiology and Pathophysiology; School of Basic Medical Science; Tianjin Medical University; Tianjin China
| | - Y. Niu
- Department of Physiology and Pathophysiology; School of Basic Medical Science; Tianjin Medical University; Tianjin China
- Department of Rehabilitation and Sports Medicine; Tianjin Medical University; Tianjin China
| | - X. Liu
- Department of Physiology and Pathophysiology; School of Basic Medical Science; Tianjin Medical University; Tianjin China
| | - L. Fu
- Department of Physiology and Pathophysiology; School of Basic Medical Science; Tianjin Medical University; Tianjin China
| |
Collapse
|
35
|
Gannon NP, Conn CA, Vaughan RA. Dietary stimulators of GLUT4 expression and translocation in skeletal muscle: a mini-review. Mol Nutr Food Res 2014; 59:48-64. [PMID: 25215442 DOI: 10.1002/mnfr.201400414] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/07/2014] [Accepted: 09/08/2014] [Indexed: 12/23/2022]
Abstract
Chronic insulin resistance can lead to type II diabetes mellitus, which is also directly influenced by an individual's genetics as well as their lifestyle. Under normal circumstances, insulin facilitates glucose uptake in skeletal muscle and adipose tissue by stimulating glucose transporter 4 (GLUT4) translocation and activity. GLUT4 activity is directly correlated with the ability to clear elevated blood glucose and insulin sensitivity. In diabetes, energy excess and prolonged hyperinsulinemia suppress muscle and adipose response to insulin, in part through reduced GLUT4 membrane levels. This work uniquely describes much of the experimental data demonstrating the effects of various dietary components on GLUT4 expression and translocation in skeletal muscle. These observations implicate several individual dietary chemicals as potential adjuvant therapies in the maintenance of diabetes and insulin resistance.
Collapse
Affiliation(s)
- Nicholas P Gannon
- Department of Biochemistry and Molecular Biology, University of New Mexico, Health Sciences Center, School of Medicine, Albuquerque, NM, USA
| | | | | |
Collapse
|
36
|
Bianco A, Pomara F, Patti A, Thomas E, Petrucci M, Bellafiore M, Battaglia G, Paoli A, Palma A. The surprising influence of family history to type 2 diabetes on anaerobic performance of young male élite athletes. SPRINGERPLUS 2014; 3:224. [PMID: 25045605 PMCID: PMC4101127 DOI: 10.1186/2193-1801-3-224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/29/2014] [Indexed: 11/10/2022]
Abstract
AIMS/HYPOTHESIS It is known that family history to type 2 diabetes induces anthropometric changes in various populations. Regular physical activity can induce adaptations in these subjects regularizing body composition and anthropometric parameters. The aim of this study is therefore to understand if family history to type 2 diabetes affects anaerobic performance in young male élite athletes. METHODS Forty six young male élite athletes were tested. Thirty three without family history to type 2 diabetes (FH-) and thirteen with family history to type 2 diabetes (FH+). Anthropometric parameters, body composition, physiological parameters and athletic performance were assessed. RESULTS Weight (p 0,0050), BMI (p 0,0019), waist circumference (p 0,0090), hips circumference (p 0,0490) and WHR (p 0,0339) were different between the two groups, showing greater values for the FH + subjects. Body composition showed lower FM and higher FFM percentages for the FH + group compared to the FH-. Anaerobic performance tests showed differences between the groups highlighting that the FH + group had higher anaerobic performance values (Wingate test for FH + 512,77 ± 107,93 W vs Wingate test for FH- 447,94 ± 56,95 W). CONCLUSIONS/INTERPRETATION The élite athletes with FH + showed better anaerobic performances and a higher body mass. At this stage we cannot generalise, but in a sample of athletes we tested, all who were with FH + showed both interesting and unexpected results; if confirmed, this evidence may represent a remarkable knowledge for fitness coaches and professionals who are daily dealing with track and field athletes and their performances.
Collapse
Affiliation(s)
- Antonino Bianco
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | | | - Antonino Patti
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | - Ewan Thomas
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | - Marco Petrucci
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | - Marianna Bellafiore
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | - Giuseppe Battaglia
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| | - Antonio Paoli
- Department of Biomedical Science, University of Padua, Padova, Italy
| | - Antonio Palma
- Sport and Exercise Sciences Research Unit, University of Palermo, Via Eleonora Duse 2, 90146 Palermo, Italy
| |
Collapse
|
37
|
Ling C, Rönn T. Epigenetic adaptation to regular exercise in humans. Drug Discov Today 2014; 19:1015-8. [PMID: 24632002 DOI: 10.1016/j.drudis.2014.03.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/06/2014] [Indexed: 01/10/2023]
Abstract
Regular exercise has numerous health benefits, for example, it reduces the risk of cardiovascular disease and cancer. It has also been shown that the risk of type 2 diabetes can be halved in high-risk groups through nonpharmacological lifestyle interventions involving exercise and diet. Nevertheless, the number of people living a sedentary life is dramatically increasing worldwide. Researchers have searched for molecular mechanisms explaining the health benefits of regular exercise for decades and it is well established that exercise alters the gene expression pattern in multiple tissues. However, until recently it was unknown that regular exercise can modify the genome-wide DNA methylation pattern in humans. This review will focus on recent progress in the field of regular exercise and epigenetics.
Collapse
Affiliation(s)
- Charlotte Ling
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, CRC, Scania University Hospital, 205 02 Malmö, Sweden.
| | - Tina Rönn
- Department of Clinical Sciences, Epigenetics and Diabetes Unit, Lund University Diabetes Centre, CRC, Scania University Hospital, 205 02 Malmö, Sweden
| |
Collapse
|
38
|
Zacharewicz E, Lamon S, Russell AP. MicroRNAs in skeletal muscle and their regulation with exercise, ageing, and disease. Front Physiol 2013; 4:266. [PMID: 24137130 PMCID: PMC3786223 DOI: 10.3389/fphys.2013.00266] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/09/2013] [Indexed: 01/08/2023] Open
Abstract
Skeletal muscle makes up approximately 40% of the total body mass, providing structural support and enabling the body to maintain posture, to control motor movements and to store energy. It therefore plays a vital role in whole body metabolism. Skeletal muscle displays remarkable plasticity and is able to alter its size, structure and function in response to various stimuli; an essential quality for healthy living across the lifespan. Exercise is an important stimulator of extracellular and intracellular stress signals that promote positive adaptations in skeletal muscle. These adaptations are controlled by changes in gene transcription and protein translation, with many of these molecules identified as potential therapeutic targets to pharmacologically improve muscle quality in patient groups too ill to exercise. MicroRNAs (miRNAs) are recently identified regulators of numerous gene networks and pathways and mainly exert their effect by binding to their target messenger RNAs (mRNAs), resulting in mRNA degradation or preventing protein translation. The role of exercise as a regulatory stimulus of skeletal muscle miRNAs is now starting to be investigated. This review highlights our current understanding of the regulation of skeletal muscle miRNAs with exercise and disease as well as how they may control skeletal muscle health.
Collapse
Affiliation(s)
- Evelyn Zacharewicz
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | | | | |
Collapse
|
39
|
Russell AP, Lamon S, Boon H, Wada S, Güller I, Brown EL, Chibalin AV, Zierath JR, Snow RJ, Stepto N, Wadley GD, Akimoto T. Regulation of miRNAs in human skeletal muscle following acute endurance exercise and short-term endurance training. J Physiol 2013; 591:4637-53. [PMID: 23798494 DOI: 10.1113/jphysiol.2013.255695] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The identification of microRNAs (miRNAs) has established new mechanisms that control skeletal muscle adaptation to exercise. The present study investigated the mRNA regulation of components of the miRNA biogenesis pathway (Drosha, Dicer and Exportin-5), muscle enriched miRNAs, (miR-1, -133a, -133b and -206), and several miRNAs dysregulated in muscle myopathies (miR-9, -23, -29, -31 and -181). Measurements were made in muscle biopsies from nine healthy untrained males at rest, 3 h following an acute bout of moderate-intensity endurance cycling and following 10 days of endurance training. Bioinformatics analysis was used to predict potential miRNA targets. In the 3 h period following the acute exercise bout, Drosha, Dicer and Exportin-5, as well as miR-1, -133a, -133-b and -181a were all increased. In contrast miR-9, -23a, -23b and -31 were decreased. Short-term training increased miR-1 and -29b, while miR-31 remained decreased. Negative correlations were observed between miR-9 and HDAC4 protein (r=-0.71; P=0.04), miR-31 and HDAC4 protein (r=-0.87; P=0.026) and miR-31 and NRF1 protein (r=-0.77; P=0.01) 3 h following exercise. miR-31 binding to the HDAC4 and NRF1 3 untranslated region (UTR) reduced luciferase reporter activity. Exercise rapidly and transiently regulates several miRNA species in muscle. Several of these miRNAs may be involved in the regulation of skeletal muscle regeneration, gene transcription and mitochondrial biogenesis. Identifying endurance exercise-mediated stress signals regulating skeletal muscle miRNAs, as well as validating their targets and regulatory pathways post exercise, will advance our understanding of their potential role/s in human health.
Collapse
Affiliation(s)
- Aaron P Russell
- A. P. Russell: Centre for Physical Activity and Nutrition Research (C-PAN), School of Exercise and Nutrition Sciences, Deakin University, 221 Burwood Highway 3125, Burwood, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kirchner H, Osler ME, Krook A, Zierath JR. Epigenetic flexibility in metabolic regulation: disease cause and prevention? Trends Cell Biol 2013; 23:203-9. [DOI: 10.1016/j.tcb.2012.11.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 12/21/2022]
|
41
|
Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, Yang BT, Lang S, Parikh H, Wessman Y, Weishaupt H, Attema J, Abels M, Wierup N, Almgren P, Jansson PA, Rönn T, Hansson O, Eriksson KF, Groop L, Ling C. Impact of an exercise intervention on DNA methylation in skeletal muscle from first-degree relatives of patients with type 2 diabetes. Diabetes 2012; 61:3322-32. [PMID: 23028138 PMCID: PMC3501844 DOI: 10.2337/db11-1653] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
To identify epigenetic patterns, which may predispose to type 2 diabetes (T2D) due to a family history (FH) of the disease, we analyzed DNA methylation genome-wide in skeletal muscle from individuals with (FH(+)) or without (FH(-)) an FH of T2D. We found differential DNA methylation of genes in biological pathways including mitogen-activated protein kinase (MAPK), insulin, and calcium signaling (P ≤ 0.007) and of individual genes with known function in muscle, including MAPK1, MYO18B, HOXC6, and the AMP-activated protein kinase subunit PRKAB1 in skeletal muscle of FH(+) compared with FH(-) men. We further validated our findings from FH(+) men in monozygotic twin pairs discordant for T2D, and 40% of 65 analyzed genes exhibited differential DNA methylation in muscle of both FH(+) men and diabetic twins. We further examined if a 6-month exercise intervention modifies the genome-wide DNA methylation pattern in skeletal muscle of the FH(+) and FH(-) individuals. DNA methylation of genes in retinol metabolism and calcium signaling pathways (P < 3 × 10(-6)) and with known functions in muscle and T2D including MEF2A, RUNX1, NDUFC2, and THADA decreased after exercise. Methylation of these human promoter regions suppressed reporter gene expression in vitro. In addition, both expression and methylation of several genes, i.e., ADIPOR1, BDKRB2, and TRIB1, changed after exercise. These findings provide new insights into how genetic background and environment can alter the human epigenome.
Collapse
Affiliation(s)
- Marloes Dekker Nitert
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Tasnim Dayeh
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Peter Volkov
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Targ Elgzyri
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Elin Hall
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Emma Nilsson
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Beatrice T. Yang
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Stefan Lang
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Hemang Parikh
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ylva Wessman
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Holger Weishaupt
- Immunology Unit, Institute for Experimental Medical Science, Lund University, Lund, Sweden
| | - Joanne Attema
- Immunology Unit, Institute for Experimental Medical Science, Lund University, Lund, Sweden
| | - Mia Abels
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Nils Wierup
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Peter Almgren
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tina Rönn
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Ola Hansson
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Karl-Fredrik Eriksson
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Leif Groop
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, CRC, Scania University Hospital, Malmö, Sweden
- Corresponding author: Charlotte Ling,
| |
Collapse
|
42
|
Olson AL. Regulation of GLUT4 and Insulin-Dependent Glucose Flux. ISRN MOLECULAR BIOLOGY 2012; 2012:856987. [PMID: 27335671 PMCID: PMC4890881 DOI: 10.5402/2012/856987] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022]
Abstract
GLUT4 has long been known to be an insulin responsive glucose transporter. Regulation of GLUT4 has been a major focus of research on the cause and prevention of type 2 diabetes. Understanding how insulin signaling alters the intracellular trafficking of GLUT4 as well as understanding the fate of glucose transported into the cell by GLUT4 will be critically important for seeking solutions to the current rise in diabetes and metabolic disease.
Collapse
Affiliation(s)
- Ann Louise Olson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, P.O. Box 26901, BMSB 964, Oklahoma City, OK 73190, USA
| |
Collapse
|
43
|
Hussey SE, McGee SL, Garnham A, McConell GK, Hargreaves M. Exercise increases skeletal muscle GLUT4 gene expression in patients with type 2 diabetes. Diabetes Obes Metab 2012; 14:768-71. [PMID: 22340256 DOI: 10.1111/j.1463-1326.2012.01585.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of the study was to determine the effect of a single bout of exercise on GLUT4 gene expression in muscle of patients with type 2 diabetes (T2D) and control subjects, matched for age and body mass index. Nine patients with T2D and nine control subjects performed 60 min of cycling exercise at ~55% peak power (W(max) ). Skeletal muscle biopsies were obtained at baseline, immediately post and 3-h post exercise. GLUT4 mRNA expression increased (p < 0.05) to a similar extent immediately post exercise in control (~60%) and T2D (~66%) subjects, and remained elevated (p < 0.05) 3-h post exercise with no differences between groups. Similarly, p-AMP-activated protein kinase, p38 mitogen-activated kinase and proliferator-activated receptor gamma co-activator-alpha mRNA expression were increased (p < 0.05) post exercise, and were not different between the groups. In conclusion, a single bout of exercise increased skeletal muscle GLUT4 mRNA expression in patients with T2D to a similar extent as in control subjects.
Collapse
|
44
|
White AT, Schenk S. NAD(+)/NADH and skeletal muscle mitochondrial adaptations to exercise. Am J Physiol Endocrinol Metab 2012; 303:E308-21. [PMID: 22436696 PMCID: PMC3423123 DOI: 10.1152/ajpendo.00054.2012] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/15/2012] [Indexed: 12/21/2022]
Abstract
The pyridine nucleotides, NAD(+) and NADH, are coenzymes that provide oxidoreductive power for the generation of ATP by mitochondria. In skeletal muscle, exercise perturbs the levels of NAD(+), NADH, and consequently, the NAD(+)/NADH ratio, and initial research in this area focused on the contribution of redox control to ATP production. More recently, numerous signaling pathways that are sensitive to perturbations in NAD(+)(H) have come to the fore, as has an appreciation for the potential importance of compartmentation of NAD(+)(H) metabolism and its subsequent effects on various signaling pathways. These pathways, which include the sirtuin (SIRT) proteins SIRT1 and SIRT3, the poly(ADP-ribose) polymerase (PARP) proteins PARP1 and PARP2, and COOH-terminal binding protein (CtBP), are of particular interest because they potentially link changes in cellular redox state to both immediate, metabolic-related changes and transcriptional adaptations to exercise. In this review, we discuss what is known, and not known, about the contribution of NAD(+)(H) metabolism and these aforementioned proteins to mitochondrial adaptations to acute and chronic endurance exercise.
Collapse
Affiliation(s)
- Amanda T White
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
45
|
White AT, Schenk S. NAD(+)/NADH and skeletal muscle mitochondrial adaptations to exercise. AMERICAN JOURNAL OF PHYSIOLOGY. ENDOCRINOLOGY AND METABOLISM 2012. [PMID: 22436696 DOI: 10.1152/ajpendo.00054.2012.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pyridine nucleotides, NAD(+) and NADH, are coenzymes that provide oxidoreductive power for the generation of ATP by mitochondria. In skeletal muscle, exercise perturbs the levels of NAD(+), NADH, and consequently, the NAD(+)/NADH ratio, and initial research in this area focused on the contribution of redox control to ATP production. More recently, numerous signaling pathways that are sensitive to perturbations in NAD(+)(H) have come to the fore, as has an appreciation for the potential importance of compartmentation of NAD(+)(H) metabolism and its subsequent effects on various signaling pathways. These pathways, which include the sirtuin (SIRT) proteins SIRT1 and SIRT3, the poly(ADP-ribose) polymerase (PARP) proteins PARP1 and PARP2, and COOH-terminal binding protein (CtBP), are of particular interest because they potentially link changes in cellular redox state to both immediate, metabolic-related changes and transcriptional adaptations to exercise. In this review, we discuss what is known, and not known, about the contribution of NAD(+)(H) metabolism and these aforementioned proteins to mitochondrial adaptations to acute and chronic endurance exercise.
Collapse
Affiliation(s)
- Amanda T White
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
46
|
Ojuka EO, Goyaram V, Smith JAH. The role of CaMKII in regulating GLUT4 expression in skeletal muscle. Am J Physiol Endocrinol Metab 2012; 303:E322-31. [PMID: 22496345 DOI: 10.1152/ajpendo.00091.2012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contractile activity during physical exercise induces an increase in GLUT4 expression in skeletal muscle, helping to improve glucose transport capacity and insulin sensitivity. An important mechanism by which exercise upregulates GLUT4 is through the activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) in response to elevated levels of cytosolic Ca(2+) during muscle contraction. This review discusses the mechanism by which Ca(2+) activates CaMKII, explains research techniques currently used to alter CaMK activity in cells, and highlights various exercise models and pharmacological agents that have been used to provide evidence that CaMKII plays an important role in regulating GLUT4 expression. With regard to transcriptional mechanisms, the key research studies that identified myocyte enhancer factor 2 (MEF2) and GLUT4 enhancer factor as the major transcription factors regulating glut4 gene expression, together with their binding domains, are underlined. Experimental evidence showing that CaMK activation induces hyperacetylation of histones in the vicinity of the MEF2 domain and increases MEF2 binding to its cis element to influence MEF2-dependent Glut4 gene expression are also given along with data suggesting that p300 might be involved in acetylating histones on the Glut4 gene. Finally, an appraisal of the roles of other calcium- and non-calcium-dependent mechanisms, including the major HDAC kinases in GLUT4 expression, is also given.
Collapse
Affiliation(s)
- Edward O Ojuka
- University of Capetown/Medical Research Center Research Unit for Exercise Science & Sports Medicine, Department of Human Biology, Univeristy of Cape Town, Cape Town, South Africa.
| | | | | |
Collapse
|
47
|
Inazuka F, Sugiyama N, Tomita M, Abe T, Shioi G, Esumi H. Muscle-specific knock-out of NUAK family SNF1-like kinase 1 (NUAK1) prevents high fat diet-induced glucose intolerance. J Biol Chem 2012; 287:16379-89. [PMID: 22418434 PMCID: PMC3351321 DOI: 10.1074/jbc.m111.302687] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
NUAK1 is a member of the AMP-activated protein kinase-related kinase family. Recent studies have shown that NUAK1 is involved in cellular senescence and motility in epithelial cells and fibroblasts. However, the physiological roles of NUAK1 are poorly understood because of embryonic lethality in NUAK1 null mice. The purpose of this study was to elucidate the roles of NUAK1 in adult tissues. We determined the tissue distribution of NUAK1 and generated muscle-specific NUAK1 knock-out (MNUAK1KO) mice. For phenotypic analysis, whole body glucose homeostasis and muscle glucose metabolism were examined. Quantitative phosphoproteome analysis of soleus muscle was performed to understand the molecular mechanisms underlying the knock-out phenotype. Nuak1 mRNA was preferentially expressed in highly oxidative tissues such as brain, heart, and soleus muscle. On a high fat diet, MNUAK1KO mice had a lower fasting blood glucose level, greater glucose tolerance, higher insulin sensitivity, and higher concentration of muscle glycogen than control mice. Phosphoproteome analysis revealed that phosphorylation of IRS1 Ser-1097 was markedly decreased in NUAK1-deficient muscle. Consistent with this, insulin signaling was enhanced in the soleus muscle of MNUAK1KO mice, as evidenced by increased phosphorylation of IRS1 Tyr-608, AKT Thr-308, and TBC1D4 Thr-649. These observations suggest that a physiological role of NUAK1 is to suppress glucose uptake through negative regulation of insulin signaling in oxidative muscle.
Collapse
Affiliation(s)
- Fumika Inazuka
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8561, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Whitham M, Chan MHS, Pal M, Matthews VB, Prelovsek O, Lunke S, El-Osta A, Broenneke H, Alber J, Brüning JC, Wunderlich FT, Lancaster GI, Febbraio MA. Contraction-induced interleukin-6 gene transcription in skeletal muscle is regulated by c-Jun terminal kinase/activator protein-1. J Biol Chem 2012; 287:10771-9. [PMID: 22351769 DOI: 10.1074/jbc.m111.310581] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exercise increases the expression of the prototypical myokine IL-6, but the precise mechanism by which this occurs has yet to be identified. To mimic exercise conditions, C2C12 myotubes were mechanically stimulated via electrical pulse stimulation (EPS). We compared the responses of EPS with the pharmacological Ca(2+) carrier calcimycin (A23187) because contraction induces marked increases in cytosolic Ca(2+) levels or the classical IκB kinase/NFκB inflammatory response elicited by H(2)O(2). We demonstrate that, unlike H(2)O(2)-stimulated increases in IL-6 mRNA, neither calcimycin- nor EPS-induced IL-6 mRNA expression is under the transcriptional control of NFκB. Rather, we show that EPS increased the phosphorylation of JNK and the reporter activity of the downstream transcription factor AP-1. Furthermore, JNK inhibition abolished the EPS-induced increase in IL-6 mRNA and protein expression. Finally, we observed an exercise-induced increase in both JNK phosphorylation and IL-6 mRNA expression in the skeletal muscles of mice after 30 min of treadmill running. Importantly, exercise did not increase IL-6 mRNA expression in skeletal muscle-specific JNK-deficient mice. These data identify a novel contraction-mediated transcriptional regulatory pathway for IL-6 in skeletal muscle.
Collapse
Affiliation(s)
- Martin Whitham
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 8008, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gong H, Xie J, Zhang N, Yao L, Zhang Y. MEF2A binding to the Glut4 promoter occurs via an AMPKα2-dependent mechanism. Med Sci Sports Exerc 2011; 43:1441-50. [PMID: 21233771 DOI: 10.1249/mss.0b013e31820f6093] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE The role of AMP-activated protein kinase α2 (AMPKα2) in regulating MEF2A nucleus translocation, nuclear histone deacetylase 5 (HDAC5) association with MEF2, HDAC5 nuclear export, MEF2A binding to the Glut4 promoter, and GLUT4 expression was investigated. METHODS This was investigated in muscles from AMPKα2 overexpression (OE) mice, AMPKα2 knockout (KO) mice, and corresponding wild-type (WT) mice that had undertaken a 28-d program of treadmill training by: 1) AMPKα-Thr172 phosphorylation by Western blot, 2) total and nuclear MEF2A by Western blot, 3) nuclear HDAC5 association with MEF2 by coimmunoprecipitation, 4) total and nuclear HDAC5 by Western blot, 5) bound MEF2A at the Glut4 MEF2 cis-element by chromatin immunoprecipitation, and 6) GLUT4 expression by real-time polymerase chain reaction and Western blot. RESULTS OE or KO of AMPKα2 isoform heightened or attenuated the training-induced increase in nuclear MEF2A content, Glut4 promoter-bound MEF2A. However, OE or KO of the AMPKα2 isoform did not have any effect on the content of nuclear HDAC5 association with MEF2 after 28 d of exercise training, although 35% lower nuclear HDAC5 protein content was found in α2-OE training muscles. Lastly, OE of the α2-isoform was associated with 120% and 155% higher GLUT4 protein and mRNA in training muscles. However, the training-induced increases of GLUT4 protein and mRNA contents were normal in α2-KO muscles despite the reduced AMPK signaling. CONCLUSIONS Exercise training increases the nuclear MEF2A content and binding of MEF2A to their binding sites on the Glut4 gene by an AMPKα2-dependent mechanism, but intracellular signaling molecules other than AMPKα2 are important in regulating training-induced HDAC5 nuclear export. Furthermore, although AMPKα2 mediates the training-induced increase in Glut4 promoter-bound MEF2A, the present data do not support an essential role of AMPKα2 in regulating training-induced GLUT4 expression in skeletal muscle.
Collapse
|
50
|
Wallace MA, Hock MB, Hazen BC, Kralli A, Snow RJ, Russell AP. Striated muscle activator of Rho signalling (STARS) is a PGC-1α/oestrogen-related receptor-α target gene and is upregulated in human skeletal muscle after endurance exercise. J Physiol 2011; 589:2027-39. [PMID: 21486805 PMCID: PMC3090601 DOI: 10.1113/jphysiol.2011.205468] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 02/15/2011] [Indexed: 01/26/2023] Open
Abstract
The striated muscle activator of Rho signalling (STARS) is an actin-binding protein specifically expressed in cardiac, skeletal and smooth muscle. STARS has been suggested to provide an important link between the transduction of external stress signals to intracellular signalling pathways controlling genes involved in the maintenance of muscle function. The aims of this study were firstly, to establish if STARS, as well as members of its downstream signalling pathway, are upregulated following acute endurance cycling exercise; and secondly, to determine if STARS is a transcriptional target of peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC-1α) and oestrogen-related receptor-α (ERRα). When measured 3 h post-exercise, STARS mRNA and protein levels as well as MRTF-A and serum response factor (SRF) nuclear protein content, were significantly increased by 140, 40, 40 and 40%, respectively. Known SRF target genes, carnitine palmitoyltransferase-1β (CPT-1β) and jun B proto-oncogene (JUNB), as well as the exercise-responsive genes PGC-1α mRNA and ERRα were increased by 2.3-, 1.8-, 4.5- and 2.7-fold, 3 h post-exercise. Infection of C2C12 myotubes with an adenovirus-expressing human PGC-1α resulted in a 3-fold increase in Stars mRNA, a response that was abolished following the suppression of endogenous ERRα. Over-expression of PGC-1α also increased Cpt-1β, Cox4 and Vegf mRNA by 6.2-, 2.0- and 2.0-fold, respectively. Suppression of endogenous STARS reduced basal Cpt-1β levels by 8.2-fold and inhibited the PGC-1α-induced increase in Cpt-1β mRNA. Our results show for the first time that the STARS signalling pathway is upregulated in response to acute endurance exercise. Additionally, we show in C2C12 myotubes that the STARS gene is a PGC-1α/ERRα transcriptional target. Furthermore, our results suggest a novel role of STARS in the co-ordination of PGC-1α-induced upregulation of the fat oxidative gene, CPT-1β.
Collapse
Affiliation(s)
- Marita A Wallace
- Centre for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood 3125, Australia
| | | | | | | | | | | |
Collapse
|