1
|
Lissek T. Enhancement of physiology via adaptive transcription. Pflugers Arch 2024:10.1007/s00424-024-03037-5. [PMID: 39482558 DOI: 10.1007/s00424-024-03037-5] [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: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
The enhancement of complex physiological functions such as cognition and exercise performance in healthy individuals represents a challenging goal. Adaptive transcription programs that are naturally activated in animals to mediate cellular plasticity in response to stimulation can be leveraged to enhance physiological function above wild-type levels in young organisms and counteract complex functional decline in aging. In processes such as learning and memory and exercise-dependent muscle remodeling, a relatively small number of molecules such as certain stimulus-responsive transcription factors and immediate early genes coordinate widespread changes in cellular physiology. Adaptive transcription can be targeted by various methods including pharmaceutical compounds and gene transfer technologies. Important problems for leveraging adaptive transcription programs for physiological enhancement include a better understanding of their dynamical organization, more precise methods to influence the underlying molecular components, and the integration of adaptive transcription into multi-scale physiological enhancement concepts.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Jasim SA, Altalbawy FMA, Abohassan M, Oghenemaro EF, Bishoyi AK, Singh RP, Kaur P, Sivaprasad GV, Mohammed JS, Hulail HM. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem Biophys 2024:10.1007/s12013-024-01587-0. [PMID: 39419931 DOI: 10.1007/s12013-024-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The histone acetyl transferases (HATs) and histone deacetylases (HDACs), which are mostly recognized for their involvement in regulating chromatin remodeling via histone acetylation/deacetylation, have been shown to also change several non-histone proteins to regulate other cellular processes. Acetylation affects the activity or function of cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors in connection to inflammation. Some small-molecule HDAC inhibitors are utilized as anticancer medications in clinical settings due to their capability to regulate cellular growth arrest, differentiation, and death. Here, we summarize our present knowledge of the innate and adaptive immunological pathways that classical HDAC enzymes control. The aim is to justify the targeted (or non-targeted) use of inhibitors against certain HDAC enzymes in inflammatory diseases such as arthritis, inflammatory bowel diseases (IBD), airways inflammation and neurological diseases.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
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] [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
|
Rebboah E, Rezaie N, Williams BA, Weimer AK, Shi M, Yang X, Liang HY, Dionne LA, Reese F, Trout D, Jou J, Youngworth I, Reinholdt L, Morabito S, Snyder MP, Wold BJ, Mortazavi A. The ENCODE mouse postnatal developmental time course identifies regulatory programs of cell types and cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598567. [PMID: 38915583 PMCID: PMC11195270 DOI: 10.1101/2024.06.12.598567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Postnatal genomic regulation significantly influences tissue and organ maturation but is under-studied relative to existing genomic catalogs of adult tissues or prenatal development in mouse. The ENCODE4 consortium generated the first comprehensive single-nucleus resource of postnatal regulatory events across a diverse set of mouse tissues. The collection spans seven postnatal time points, mirroring human development from childhood to adulthood, and encompasses five core tissues. We identified 30 cell types, further subdivided into 69 subtypes and cell states across adrenal gland, left cerebral cortex, hippocampus, heart, and gastrocnemius muscle. Our annotations cover both known and novel cell differentiation dynamics ranging from early hippocampal neurogenesis to a new sex-specific adrenal gland population during puberty. We used an ensemble Latent Dirichlet Allocation strategy with a curated vocabulary of 2,701 regulatory genes to identify regulatory "topics," each of which is a gene vector, linked to cell type differentiation, subtype specialization, and transitions between cell states. We find recurrent regulatory topics in tissue-resident macrophages, neural cell types, endothelial cells across multiple tissues, and cycling cells of the adrenal gland and heart. Cell-type-specific topics are enriched in transcription factors and microRNA host genes, while chromatin regulators dominate mitosis topics. Corresponding chromatin accessibility data reveal dynamic and sex-specific regulatory elements, with enriched motifs matching transcription factors in regulatory topics. Together, these analyses identify both tissue-specific and common regulatory programs in postnatal development across multiple tissues through the lens of the factors regulating transcription.
Collapse
Affiliation(s)
- Elisabeth Rebboah
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Narges Rezaie
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Brian A. Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Annika K. Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Minyi Shi
- Department of Next Generation Sequencing and Microchemistry, Proteomics and Lipidomics, Genentech, San Francisco, USA
| | - Xinqiong Yang
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Heidi Yahan Liang
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | | | - Fairlie Reese
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | - Diane Trout
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Jennifer Jou
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Ingrid Youngworth
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | | | - Samuel Morabito
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Barbara J. Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Ali Mortazavi
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| |
Collapse
|
5
|
Zhao Z, Guo D, Wei Y, Li J, Jia X, Niu Y, Liu Z, Bai Y, Chen Z, Shi B, Zhang X, Hu J, Wang J, Liu X, Li S. Integrative ATAC-seq and RNA-seq Analysis of the Longissimus Dorsi Muscle of Gannan Yak and Jeryak. Int J Mol Sci 2024; 25:6029. [PMID: 38892214 PMCID: PMC11172533 DOI: 10.3390/ijms25116029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Jeryak is the F1 generation of the cross between Gannan yak and Jersey cattle, which has the advantages of fast growth and high adaptability. The growth and development of skeletal muscle is closely linked to meat production and the quality of meat. However, the molecular regulatory mechanisms of muscle growth differences between Gannan yak and Jeryak analyzed from the perspective of chromatin opening have not been reported. In this study, ATAC-seq was used to analyze the difference of chromatin openness in longissimus muscle of Gannan yak and Jeryak. It was found that chromatin accessibility was more enriched in Jeryak compared to Gannan yak, especially in the range of the transcription start site (TSS) ± 2 kb. GO and KEGG enrichment analysis indicate that differential peak-associated genes are involved in the negative regulation of muscle adaptation and the Hippo signaling pathway. Integration analysis of ATAC-seq and RNA-seq revealed overlapping genes were significantly enriched during skeletal muscle cell differentiation and muscle organ morphogenesis. At the same time, we screened FOXO1, ZBED6, CRY2 and CFL2 for possible involvement in skeletal muscle development, constructed a genes and transcription factors network map, and found that some transcription factors (TFs), including YY1, KLF4, KLF5 and Bach1, were involved in skeletal muscle development. Overall, we have gained a comprehensive understanding of the key factors that impact skeletal muscle development in various breeds of cattle, providing new insights for future analysis of the molecular regulatory mechanisms involved in muscle growth and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730000, China; (Z.Z.); (D.G.); (Y.W.); (J.L.); (X.J.); (Y.N.); (Z.L.); (Y.B.); (Z.C.); (B.S.); (X.Z.); (J.W.); (X.L.); (S.L.)
| | | | | | | |
Collapse
|
6
|
Li Y, Feng Y, Chen X, He J, Luo Y, Yu B, Chen D, Huang Z. Dietary short-term supplementation of grape seed proanthocyanidin extract improves pork quality and promotes skeletal muscle fiber type conversion in finishing pigs. Meat Sci 2024; 210:109436. [PMID: 38266434 DOI: 10.1016/j.meatsci.2024.109436] [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: 09/30/2023] [Revised: 12/18/2023] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Plant extracts are commonly used as feed additives to improve pork quality. However, due to their high cost, shortening the duration of supplement use can help reduce production costs. In this study, we aimed to investigate the effects of grape seed proanthocyanidin extract (GSPE) on meat quality and muscle fiber characteristics of finishing pigs during the late stage of fattening, which was 30 days in our experimental design. The results indicated that short-term dietary supplementation of GSPE significantly reduced backfat thickness, but increased loin eye area and improved meat color and tenderness. Moreover, GSPE increased slow myosin heavy chain (MyHC) expression and malate dehydrogenase (MDH) activity, while decreasing fast MyHC expression and lactate dehydrogenase (LDH) activity in the Longissimus thoracis (LT) muscle. Additionally, GSPE increased the expression of Sirt1 and PGC-1α proteins in the LT muscle of finishing pigs and upregulated AMP-activated protein kinase α 1 (AMPKα1), AMPKα2, nuclear respiratory factor 1 (NRF1), and calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ) mRNA expression levels. These findings suggest that even during the late stage of fattening, GSPE treatment can regulate skeletal muscle fiber type transformation through the AMPK signaling pathway, thereby affecting the muscle quality of finishing pigs. Therefore, by incorporating GSPE into the diet of pigs during the late stage of fattening, producers can enhance pork quality while reducing production costs.
Collapse
Affiliation(s)
- Yiqiang Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yadi Feng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.
| |
Collapse
|
7
|
Mozzetta C, Sartorelli V, Steinkuhler C, Puri PL. HDAC inhibitors as pharmacological treatment for Duchenne muscular dystrophy: a discovery journey from bench to patients. Trends Mol Med 2024; 30:278-294. [PMID: 38408879 PMCID: PMC11095976 DOI: 10.1016/j.molmed.2024.01.007] [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: 11/11/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/28/2024]
Abstract
Earlier evidence that targeting the balance between histone acetyltransferases (HATs) and deacetylases (HDACs), through exposure to HDAC inhibitors (HDACis), could enhance skeletal myogenesis, prompted interest in using HDACis to promote muscle regeneration. Further identification of constitutive HDAC activation in dystrophin-deficient muscles, caused by dysregulated nitric oxide (NO) signaling, provided the rationale for HDACi-based therapeutic interventions for Duchenne muscular dystrophy (DMD). In this review, we describe the molecular, preclinical, and clinical evidence supporting the efficacy of HDACis in countering disease progression by targeting pathogenic networks of gene expression in multiple muscle-resident cell types of patients with DMD. Given that givinostat is paving the way for HDACi-based interventions in DMD, next-generation HDACis with optimized therapeutic profiles and efficacy could be also explored for synergistic combinations with other therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy, Rome, Italy
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Pier Lorenzo Puri
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
8
|
Wu H, Hu Y, Jiang C, Chen C. Global scientific trends in research of epigenetic response to exercise: A bibliometric analysis. Heliyon 2024; 10:e25644. [PMID: 38370173 PMCID: PMC10869857 DOI: 10.1016/j.heliyon.2024.e25644] [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: 11/02/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024] Open
Abstract
The purpose of this work is to comprehensively understand the adaptive response of multiple epigenetic modifications on gene expression changes driven by exercise. Here, we retrieved literatures from publications in the PubMed and Web of Science Core Collection databases up to and including October 15, 2023. After screening with the exclusion criteria, 1910 publications were selected in total, comprising 1399 articles and 511 reviews. Specifically, a total of 512, 224, and 772 publications is involved in DNA methylation, histone modification, and noncoding RNAs, respectively. The correlations between publication number, authors, institutions, countries, references, and the characteristics of hotspots were explored by CiteSpace. Here, the USA (621 publications) ranked the world's most-influential countries, the University of California System (68 publications) was the most productive, and Tiago Fernandes (14 publications) had the most-published publications. A comprehensive keyword analysis revealed that cardiovascular disease, cancer, skeletal muscle development, and metabolic syndrome, and are the research hotspots. The detailed impact of exercise was further discussed in different aspects of these three categories of epigenetic modifications. Detailed analysis of epigenetic modifications in response to exercise, including DNA methylation, histone modification, and changes in noncoding RNAs, will offer valuable information to help researchers understand hotspots and emerging trends.
Collapse
Affiliation(s)
- Huijuan Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yue Hu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Cai Jiang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Cong Chen
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, 350122 Fuzhou, Fujian, China
- Fujian Key Laboratory of Cognitive Rehabilitation, Fujian University of Traditional Chinese Medicine, 350122, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Pitzer CR, Paez HG, Ferrandi PJ, Mohamed J, Alway SE. Extracellular vesicles from obese and diabetic mouse plasma alter C2C12 myotube glucose uptake and gene expression. Physiol Rep 2024; 12:e15898. [PMID: 38169108 PMCID: PMC10761623 DOI: 10.14814/phy2.15898] [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: 06/29/2023] [Revised: 10/07/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024] Open
Abstract
Recent studies have indicated a role for circulating extracellular vesicles (EVs) in the pathogenesis of multiple diseases. However, most in vitro studies have used variable and arbitrary doses of EVs rather than interpreting EVs as an existing component of standard skeletal muscle cell culture media. The current study provides an initial investigation into the effects of circulating EVs on the metabolic phenotype of C2C12 myotubes by replacing EVs from fetal bovine serum with circulating EVs from control mice or mice with obesity and type 2 diabetes (OT2D). We report that EVs associated with OT2D decrease 2-NBDG uptake (a proxy measure of glucose uptake) in the insulin-stimulated state compared to controls. OT2D associated EV treatment also significantly decreased myosin heavy chain type 1 (MHCI) mRNA abundance in myotubes but had no effect on mRNA expression of any other myosin heavy chain isoforms. OT2D-associated circulating EVs also significantly increased lipid accumulation within myotubes without altering the expression of a selection of genes important for lipid entry, synthesis, or catabolism. The data indicate that, in a severely diabetic state, circulating EVs may contribute to insulin resistance and alter gene expression in myotubes in a manner consistent with the skeletal muscle phenotype observed in OT2D.
Collapse
Affiliation(s)
- Christopher R. Pitzer
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of Physiology, College of MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health SciencesThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Hector G. Paez
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of Physiology, College of MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health SciencesThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Peter J. Ferrandi
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health SciencesThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Junaith S. Mohamed
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Tennessee Institute of Regenerative MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Stephen E. Alway
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of Physiology, College of MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health ProfessionsThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Tennessee Institute of Regenerative MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
10
|
Lira Chavez FM, Gartzke LP, van Beuningen FE, Wink SE, Henning RH, Krenning G, Bouma HR. Restoring the infected powerhouse: Mitochondrial quality control in sepsis. Redox Biol 2023; 68:102968. [PMID: 38039825 PMCID: PMC10711241 DOI: 10.1016/j.redox.2023.102968] [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: 10/12/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Sepsis is a dysregulated host response to an infection, characterized by organ failure. The pathophysiology is complex and incompletely understood, but mitochondria appear to play a key role in the cascade of events that culminate in multiple organ failure and potentially death. In shaping immune responses, mitochondria fulfil dual roles: they not only supply energy and metabolic intermediates crucial for immune cell activation and function but also influence inflammatory and cell death pathways. Importantly, mitochondrial dysfunction has a dual impact, compromising both immune system efficiency and the metabolic stability of end organs. Dysfunctional mitochondria contribute to the development of a hyperinflammatory state and loss of cellular homeostasis, resulting in poor clinical outcomes. Already in early sepsis, signs of mitochondrial dysfunction are apparent and consequently, strategies to optimize mitochondrial function in sepsis should not only prevent the occurrence of mitochondrial dysfunction, but also cover the repair of the sustained mitochondrial damage. Here, we discuss mitochondrial quality control (mtQC) in the pathogenesis of sepsis and exemplify how mtQC could serve as therapeutic target to overcome mitochondrial dysfunction. Hence, replacing or repairing dysfunctional mitochondria may contribute to the recovery of organ function in sepsis. Mitochondrial biogenesis is a process that results in the formation of new mitochondria and is critical for maintaining a pool of healthy mitochondria. However, exacerbated biogenesis during early sepsis can result in accumulation of structurally aberrant mitochondria that fail to restore bioenergetics, produce excess reactive oxygen species (ROS) and exacerbate the disease course. Conversely, enhancing mitophagy can protect against organ damage by limiting the release of mitochondrial-derived damage-associated molecules (DAMPs). Furthermore, promoting mitophagy may facilitate the growth of healthy mitochondria by blocking the replication of damaged mitochondria and allow for post sepsis organ recovery through enabling mitophagy-coupled biogenesis. The remaining healthy mitochondria may provide an undamaged scaffold to reproduce functional mitochondria. However, the kinetics of mtQC in sepsis, specifically mitophagy, and the optimal timing for intervention remain poorly understood. This review emphasizes the importance of integrating mitophagy induction with mtQC mechanisms to prevent undesired effects associated with solely the induction of mitochondrial biogenesis.
Collapse
Affiliation(s)
- F M Lira Chavez
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands.
| | - L P Gartzke
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - F E van Beuningen
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - S E Wink
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - G Krenning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands; Sulfateq B.V, Admiraal de Ruyterlaan 5, 9726, GN Groningen, the Netherlands
| | - H R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands; Department of Internal Medicine, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| |
Collapse
|
11
|
Hota M, Barber JL, Ruiz-Ramie JJ, Schwartz CS, Lam DTUH, Rao P, Mi MY, Katz DH, Robbins JM, Clish CB, Gerszten RE, Sarzynski MA, Ghosh S, Bouchard C. Omics-driven investigation of the biology underlying intrinsic submaximal working capacity and its trainability. Physiol Genomics 2023; 55:517-543. [PMID: 37661925 PMCID: PMC11178266 DOI: 10.1152/physiolgenomics.00163.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/21/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Submaximal exercise capacity is an indicator of cardiorespiratory fitness with clinical and public health implications. Submaximal exercise capacity and its response to exercise programs are characterized by heritability levels of about 40%. Using physical working capacity (power output) at a heart rate of 150 beats/min (PWC150) as an indicator of submaximal exercise capacity in subjects of the HERITAGE Family Study, we have undertaken multi-omics and in silico explorations of the underlying biology of PWC150 and its response to 20 wk of endurance training. Our goal was to illuminate the biological processes and identify panels of genes associated with human variability in intrinsic PWC150 (iPWC150) and its trainability (dPWC150). Our bioinformatics approach was based on a combination of genome-wide association, skeletal muscle gene expression, and plasma proteomics and metabolomics experiments. Genes, proteins, and metabolites showing significant associations with iPWC150 or dPWC150 were further queried for the enrichment of biological pathways. We compared genotype-phenotype associations of emerging candidate genes with reported functional consequences of gene knockouts in mouse models. We investigated the associations between DNA variants and multiple muscle and cardiovascular phenotypes measured in HERITAGE subjects. Two panels of prioritized genes of biological relevance to iPWC150 (13 genes) and dPWC150 (6 genes) were identified, supporting the hypothesis that genes and pathways associated with iPWC150 are different from those underlying dPWC150. Finally, the functions of these genes and pathways suggested that human variation in submaximal exercise capacity is mainly driven by skeletal muscle morphology and metabolism and red blood cell oxygen-carrying capacity.NEW & NOTEWORTHY Multi-omics and in silico explorations of the genes and underlying biology of submaximal exercise capacity and its response to 20 wk of endurance training were undertaken. Prioritized genes were identified: 13 genes for variation in submaximal exercise capacity in the sedentary state and 5 genes for the response level to endurance training, with no overlap between them. Genes and pathways associated with submaximal exercise capacity in the sedentary state are different from those underlying trainability.
Collapse
Affiliation(s)
- Monalisa Hota
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Jacob L Barber
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Jonathan J Ruiz-Ramie
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
- Department of Kinesiology, Augusta University, Augusta, Georgia, United States
| | - Charles S Schwartz
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Do Thuy Uyen Ha Lam
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Prashant Rao
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Michael Y Mi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Daniel H Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Jeremy M Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Clary B Clish
- Metabolomics Platform, Broad Institute, Boston, Massachusetts, United States
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Mark A Sarzynski
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Sujoy Ghosh
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Bioinformatics Section, Human Genomics Core, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| |
Collapse
|
12
|
Takakura Y, Suzuki T, Hirai N, Araki T, Ohishi M, Sato H, Yamaguchi N, Takano H, Yamaguchi N. VGLL3 confers slow-twitch muscle differentiation via PGC-1α expression in C2C12 myocytes. Biochem Biophys Res Commun 2023; 669:30-37. [PMID: 37262950 DOI: 10.1016/j.bbrc.2023.05.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023]
Abstract
Vestigial-like family member 3 (VGLL3) is a cofactor for the TEA-domain transcription factor (TEAD) family. Although VGLL3 influences myogenic differentiation, its involvement in slow- and fast-twitch fiber specification remains unknown. In this study, we established a cell line stably overexpressing VGLL3 and analyzed effects of VGLL3 on the myogenic differentiation of murine myoblast C2C12 cells. We found that VGLL3 expression promotes slow-twitch muscle differentiation. Mechanistically, VGLL3 expression induced the expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master transcriptional regulator of slow-twitch muscle development. We also found that VGLL3 proteins are degraded by the proteasome, which causes switching of TEAD cofactors from VGLL3 to Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding motif (TAZ). These results suggest that the balance between the two kinds of TEAD cofactors VGLL3 and YAP/TAZ controls muscle fiber-type specification.
Collapse
Affiliation(s)
- Yuki Takakura
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba, 260-8675, Chiba University, Japan; Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Takayuki Suzuki
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Naoto Hirai
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Takuro Araki
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba, 260-8675, Chiba University, Japan; Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Mai Ohishi
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba, 260-8675, Chiba University, Japan
| | - Hiromi Sato
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Naoto Yamaguchi
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Hiroyuki Takano
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba, 260-8675, Chiba University, Japan
| | - Noritaka Yamaguchi
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba, 260-8675, Chiba University, Japan; Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan.
| |
Collapse
|
13
|
Dent JR, Stocks B, Campelj DG, Philp A. Transient changes to metabolic homeostasis initiate mitochondrial adaptation to endurance exercise. Semin Cell Dev Biol 2023; 143:3-16. [PMID: 35351374 DOI: 10.1016/j.semcdb.2022.03.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/26/2022] [Accepted: 03/19/2022] [Indexed: 12/14/2022]
Abstract
Endurance exercise is well established to increase mitochondrial content and function in skeletal muscle, a process termed mitochondrial biogenesis. Current understanding is that exercise initiates skeletal muscle mitochondrial remodeling via modulation of cellular nutrient, energetic and contractile stress pathways. These subtle changes in the cellular milieu are sensed by numerous transduction pathways that serve to initiate and coordinate an increase in mitochondrial gene transcription and translation. The result of these acute signaling events is the promotion of growth and assembly of mitochondria, coupled to a greater capacity for aerobic ATP provision in skeletal muscle. The aim of this review is to highlight the acute metabolic events induced by endurance exercise and the subsequent molecular pathways that sense this transient change in cellular homeostasis to drive mitochondrial adaptation and remodeling.
Collapse
Affiliation(s)
- Jessica R Dent
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Dean G Campelj
- Mitochondrial Metabolism and Ageing Laboratory, Healthy Ageing Research Theme, Garvan Institute of Medical Research, Sydney, Australia
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing Laboratory, Healthy Ageing Research Theme, Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Medical School, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
14
|
Zhang Y, Ramirez-Martinez A, Chen K, McAnally JR, Cai C, Durbacz MZ, Chemello F, Wang Z, Xu L, Bassel-Duby R, Liu N, Olson EN. Net39 protects muscle nuclei from mechanical stress during the pathogenesis of Emery-Dreifuss muscular dystrophy. J Clin Invest 2023; 133:e163333. [PMID: 37395273 PMCID: PMC10313361 DOI: 10.1172/jci163333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Mutations in genes encoding nuclear envelope proteins lead to diseases known as nuclear envelopathies, characterized by skeletal muscle and heart abnormalities, such as Emery-Dreifuss muscular dystrophy (EDMD). The tissue-specific role of the nuclear envelope in the etiology of these diseases has not been extensively explored. We previously showed that global deletion of the muscle-specific nuclear envelope protein NET39 in mice leads to neonatal lethality due to skeletal muscle dysfunction. To study the potential role of the Net39 gene in adulthood, we generated a muscle-specific conditional knockout (cKO) of Net39 in mice. cKO mice recapitulated key skeletal muscle features of EDMD, including muscle wasting, impaired muscle contractility, abnormal myonuclear morphology, and DNA damage. The loss of Net39 rendered myoblasts hypersensitive to mechanical stretch, resulting in stretch-induced DNA damage. Net39 was downregulated in a mouse model of congenital myopathy, and restoration of Net39 expression through AAV gene delivery extended life span and ameliorated muscle abnormalities. These findings establish NET39 as a direct contributor to the pathogenesis of EDMD that acts by protecting against mechanical stress and DNA damage.
Collapse
Affiliation(s)
- Yichi Zhang
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Andres Ramirez-Martinez
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Kenian Chen
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, and
| | - John R. McAnally
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Chunyu Cai
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Mateusz Z. Durbacz
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Francesco Chemello
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Zhaoning Wang
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, and
| | - Rhonda Bassel-Duby
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Ning Liu
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Eric N. Olson
- Department of Molecular Biology
- Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| |
Collapse
|
15
|
Sato T, Umebayashi S, Senoo N, Akahori T, Ichida H, Miyoshi N, Yoshida T, Sugiura Y, Goto-Inoue N, Kawana H, Shindou H, Baba T, Maemoto Y, Kamei Y, Shimizu T, Aoki J, Miura S. LPGAT1/LPLAT7 regulates acyl chain profiles at the sn-1 position of phospholipids in murine skeletal muscles. J Biol Chem 2023:104848. [PMID: 37217003 PMCID: PMC10285227 DOI: 10.1016/j.jbc.2023.104848] [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: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 05/24/2023] Open
Abstract
Skeletal muscle consists of both fast- and slow-twitch fibers. Phospholipids are important structural components of cellular membranes, and the diversity of their fatty acid composition affects membrane fluidity and permeability. Although some studies have shown that acyl chain species in phospholipids differ among various muscle fiber types, the mechanisms underlying these differences are unclear. To investigate this, we analyzed phosphatidylcholine (PC) and phosphatidylethanolamine (PE) molecules in the murine extensor digitorum longus (EDL; fast-twitch) and soleus (slow-twitch) muscles. In the EDL muscle, the vast majority (93.6%) of PC molecules was palmitate-containing PC (16:0-PC), whereas in the soleus muscle, in addition to 16:0-PC, 27.9% of PC molecules was stearate-containing PC (18:0-PC). Most palmitate and stearate were bound at the sn-1 position of 16:0- and 18:0-PC, respectively, and 18:0-PC was found in type I and IIa fibers. The amount of 18:0-PE was higher in the soleus than in the EDL muscle. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) increased the amount of 18:0-PC in the EDL. Lysophosphatidylglycerol acyltransferase 1 (LPGAT1) was highly expressed in the soleus compared with that in the EDL muscle and was upregulated by PGC-1α. LPGAT1 knockout decreased the incorporation of stearate into PC and PE in vitro and ex vivo and the amount of 18:0-PC and 18:0-PE in murine skeletal muscle with an increase in the level of 16:0-PC and 16:0-PE. Moreover, knocking out LPGAT1 decreased the amount of stearate-containing-phosphatidylserine (18:0-PS), suggesting that LPGAT1 regulated the acyl chain profiles of phospholipids, namely PC, PE, and PS, in the skeletal muscle.
Collapse
Affiliation(s)
- Tomoki Sato
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Shuhei Umebayashi
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Nanami Senoo
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Takumi Akahori
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Hiyori Ichida
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Noriyuki Miyoshi
- Laboratory of Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Takuya Yoshida
- Laboratory of Clinical Nutrition, Graduate School of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, 862-8502, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Naoko Goto-Inoue
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Fujisawa, 252-0880, Japan
| | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takashi Baba
- Laboratory of Molecular Cell Biology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, 192-0392, Japan
| | - Yuki Maemoto
- Laboratory of Molecular Cell Biology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, 192-0392, Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition, Graduate School of Environmental and Life Science, Kyoto Prefectural University, Kyoto, 606-8522, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Institute of Microbial Chemistry, Tokyo, 141-0021, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan.
| |
Collapse
|
16
|
Abu Shelbayeh O, Arroum T, Morris S, Busch KB. PGC-1α Is a Master Regulator of Mitochondrial Lifecycle and ROS Stress Response. Antioxidants (Basel) 2023; 12:antiox12051075. [PMID: 37237941 DOI: 10.3390/antiox12051075] [Citation(s) in RCA: 67] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria play a major role in ROS production and defense during their life cycle. The transcriptional activator PGC-1α is a key player in the homeostasis of energy metabolism and is therefore closely linked to mitochondrial function. PGC-1α responds to environmental and intracellular conditions and is regulated by SIRT1/3, TFAM, and AMPK, which are also important regulators of mitochondrial biogenesis and function. In this review, we highlight the functions and regulatory mechanisms of PGC-1α within this framework, with a focus on its involvement in the mitochondrial lifecycle and ROS metabolism. As an example, we show the role of PGC-1α in ROS scavenging under inflammatory conditions. Interestingly, PGC-1α and the stress response factor NF-κB, which regulates the immune response, are reciprocally regulated. During inflammation, NF-κB reduces PGC-1α expression and activity. Low PGC-1α activity leads to the downregulation of antioxidant target genes resulting in oxidative stress. Additionally, low PGC-1α levels and concomitant oxidative stress promote NF-κB activity, which exacerbates the inflammatory response.
Collapse
Affiliation(s)
- Othman Abu Shelbayeh
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, 48149 Münster, Germany
| | - Tasnim Arroum
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, 48149 Münster, Germany
- Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48202, USA
| | - Silke Morris
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, 48149 Münster, Germany
| | - Karin B Busch
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, 48149 Münster, Germany
| |
Collapse
|
17
|
Sadaki S, Fujita R, Hayashi T, Nakamura A, Okamura Y, Fuseya S, Hamada M, Warabi E, Kuno A, Ishii A, Muratani M, Okada R, Shiba D, Kudo T, Takeda S, Takahashi S. Large Maf transcription factor family is a major regulator of fast type IIb myofiber determination. Cell Rep 2023; 42:112289. [PMID: 36952339 DOI: 10.1016/j.celrep.2023.112289] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
Myofibers are broadly characterized as fatigue-resistant slow-twitch (type I) fibers and rapidly fatiguing fast-twitch (type IIa/IIx/IIb) fibers. However, the molecular regulation of myofiber type is not entirely understood; particularly, information on regulators of fast-twitch muscle is scarce. Here, we demonstrate that the large Maf transcription factor family dictates fast type IIb myofiber specification in mice. Remarkably, the ablation of three large Mafs leads to the drastic loss of type IIb myofibers, resulting in enhanced endurance capacity and the reduction of muscle force. Conversely, the overexpression of each large Maf in the type I soleus muscle induces type IIb myofibers. Mechanistically, a large Maf directly binds to the Maf recognition element on the promoter of myosin heavy chain 4, which encodes the type IIb myosin heavy chain, driving its expression. This work identifies the large Maf transcription factor family as a major regulator for fast type IIb muscle determination.
Collapse
Affiliation(s)
- Shunya Sadaki
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Ph.D. Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Ryo Fujita
- Division of Regenerative Medicine, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| | - Takuto Hayashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Ayano Nakamura
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yui Okamura
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Sayaka Fuseya
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Michito Hamada
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Eiji Warabi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akihiro Kuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akiko Ishii
- Department of Neurology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Risa Okada
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Ibaraki 305-8505, Japan
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Tsukuba, Ibaraki 305-8505, Japan
| | - Takashi Kudo
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
18
|
Molinari S, Imbriano C, Moresi V, Renzini A, Belluti S, Lozanoska-Ochser B, Gigli G, Cedola A. Histone deacetylase functions and therapeutic implications for adult skeletal muscle metabolism. Front Mol Biosci 2023; 10:1130183. [PMID: 37006625 PMCID: PMC10050567 DOI: 10.3389/fmolb.2023.1130183] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Skeletal muscle is a highly adaptive organ that sustains continuous metabolic changes in response to different functional demands. Healthy skeletal muscle can adjust fuel utilization to the intensity of muscle activity, the availability of nutrients and the intrinsic characteristics of muscle fibers. This property is defined as metabolic flexibility. Importantly, impaired metabolic flexibility has been associated with, and likely contributes to the onset and progression of numerous pathologies, including sarcopenia and type 2 diabetes. Numerous studies involving genetic and pharmacological manipulations of histone deacetylases (HDACs) in vitro and in vivo have elucidated their multiple functions in regulating adult skeletal muscle metabolism and adaptation. Here, we briefly review HDAC classification and skeletal muscle metabolism in physiological conditions and upon metabolic stimuli. We then discuss HDAC functions in regulating skeletal muscle metabolism at baseline and following exercise. Finally, we give an overview of the literature regarding the activity of HDACs in skeletal muscle aging and their potential as therapeutic targets for the treatment of insulin resistance.
Collapse
Affiliation(s)
- Susanna Molinari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Viviana Moresi
- Institute of Nanotechnology, Department of Physics, National Research Council (CNR-NANOTEC), Sapienza University of Rome, Rome, Italy
- *Correspondence: Viviana Moresi,
| | - Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Lecce, Italy
| | - Alessia Cedola
- Institute of Nanotechnology, Department of Physics, National Research Council (CNR-NANOTEC), Sapienza University of Rome, Rome, Italy
| |
Collapse
|
19
|
Sandonà M, Cavioli G, Renzini A, Cedola A, Gigli G, Coletti D, McKinsey TA, Moresi V, Saccone V. Histone Deacetylases: Molecular Mechanisms and Therapeutic Implications for Muscular Dystrophies. Int J Mol Sci 2023; 24:4306. [PMID: 36901738 PMCID: PMC10002075 DOI: 10.3390/ijms24054306] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Histone deacetylases (HDACs) are enzymes that regulate the deacetylation of numerous histone and non-histone proteins, thereby affecting a wide range of cellular processes. Deregulation of HDAC expression or activity is often associated with several pathologies, suggesting potential for targeting these enzymes for therapeutic purposes. For example, HDAC expression and activity are higher in dystrophic skeletal muscles. General pharmacological blockade of HDACs, by means of pan-HDAC inhibitors (HDACi), ameliorates both muscle histological abnormalities and function in preclinical studies. A phase II clinical trial of the pan-HDACi givinostat revealed partial histological improvement and functional recovery of Duchenne Muscular Dystrophy (DMD) muscles; results of an ongoing phase III clinical trial that is assessing the long-term safety and efficacy of givinostat in DMD patients are pending. Here we review the current knowledge about the HDAC functions in distinct cell types in skeletal muscle, identified by genetic and -omic approaches. We describe the signaling events that are affected by HDACs and contribute to muscular dystrophy pathogenesis by altering muscle regeneration and/or repair processes. Reviewing recent insights into HDAC cellular functions in dystrophic muscles provides new perspectives for the development of more effective therapeutic approaches based on drugs that target these critical enzymes.
Collapse
Affiliation(s)
| | - Giorgia Cavioli
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alessia Cedola
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), 73100 Lecce, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Human Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “La Sapienza”, 00161 Rome, Italy
- CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Aging B2A, Sorbonne Université, 75005 Paris, France
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Viviana Moresi
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), University of Rome “La Sapienza”, 00181 Rome, Italy
| | - Valentina Saccone
- IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
20
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
21
|
Cuttini E, Goi C, Pellarin E, Vida R, Brancolini C. HDAC4 in cancer: A multitasking platform to drive not only epigenetic modifications. Front Mol Biosci 2023; 10:1116660. [PMID: 36762207 PMCID: PMC9902726 DOI: 10.3389/fmolb.2023.1116660] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Controlling access to genomic information and maintaining its stability are key aspects of cell life. Histone acetylation is a reversible epigenetic modification that allows access to DNA and the assembly of protein complexes that regulate mainly transcription but also other activities. Enzymes known as histone deacetylases (HDACs) are involved in the removal of the acetyl-group or in some cases of small hydrophobic moieties from histones but also from the non-histone substrate. The main achievement of HDACs on histones is to repress transcription and promote the formation of more compact chromatin. There are 18 different HDACs encoded in the human genome. Here we will discuss HDAC4, a member of the class IIa family, and its possible contribution to cancer development.
Collapse
Affiliation(s)
- Emma Cuttini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Camilla Goi
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Ester Pellarin
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Riccardo Vida
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Claudio Brancolini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy,Laboratory of Epigenomics, Department of Medicine, Università degli Studi di Udine, Udine, Italy,*Correspondence: Claudio Brancolini,
| |
Collapse
|
22
|
Pan P, Qin Z, Xie W, Chen B, Guan Z, Xie B. Identification of Differentially Expressed Genes in the Longissimus Dorsi Muscle of Luchuan and Duroc Pigs by Transcriptome Sequencing. Genes (Basel) 2023; 14:132. [PMID: 36672873 PMCID: PMC9859529 DOI: 10.3390/genes14010132] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/13/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
The Duroc pig originated in the United States and is a typical lean-meat pig. The breed grows fast, and the body size is large, but the meat quality is poor. The Luchuan pig is one of eight excellent local breeds in China; it has tender meat but is small in size. To study the factors that determine growth, we selected the longissimus dorsi muscle of Luchuan and Duroc pigs for transcriptome sequencing. The results of the transcriptome showed that 3682 genes were differentially expressed (DEGs) in the longissimus dorsi muscle of Duroc and Luchuan pigs. We screened out genes related to muscle development and selected the MYL2 (Myosin light chain-2) gene to perform preliminary research. Gene Ontology (GO) enrichment of biological functions and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the gene products were mainly involved in the Akt/FoxO signaling pathway, fatty acid metabolism, arachidonic acid metabolism and glycine, serine and threonine metabolism. Such pathways contributed to skeletal muscle growth, fatty acid metabolism and intramuscular fat deposition. These results provide insight into the mechanisms underlying the formation of skeletal muscle and provide candidate genes to improve growth traits, as well as contribute to improving the growth and development traits of pigs through molecular breeding.
Collapse
Affiliation(s)
- Pengcheng Pan
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Agricultural Vocational and Technical University, Nanning 530001, China
| | - Zhaoxian Qin
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Agricultural Vocational and Technical University, Nanning 530001, China
| | - Wan Xie
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Baojian Chen
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Agricultural Vocational and Technical University, Nanning 530001, China
| | - Zhihui Guan
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Agricultural Vocational and Technical University, Nanning 530001, China
| | - Bingkun Xie
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Agricultural Vocational and Technical University, Nanning 530001, China
| |
Collapse
|
23
|
Oikawa S, Akimoto T. Functional Analysis of MicroRNAs in Skeletal Muscle. Methods Mol Biol 2023; 2640:339-349. [PMID: 36995606 DOI: 10.1007/978-1-0716-3036-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are highly conserved in vertebrates and play important roles in diverse biological processes. miRNAs function to fine-tune gene expression by accelerating the degradation of mRNA and/or by inhibiting protein translation. Identification of muscle-specific miRNAs has extended our knowledge of the molecular network in skeletal muscle. Here we describe methods that are commonly used to analyze the function of miRNAs in skeletal muscle.
Collapse
Affiliation(s)
- Satoshi Oikawa
- Laboratory of Muscle Biology, Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | - Takayuki Akimoto
- Laboratory of Muscle Biology, Faculty of Sport Sciences, Waseda University, Saitama, Japan.
| |
Collapse
|
24
|
Metformin Attenuates Slow-to-Fast Fiber Shift and Proteolysis Markers Increase in Rat Soleus after 7 Days of Rat Hindlimb Unloading. Int J Mol Sci 2022; 24:ijms24010503. [PMID: 36613942 PMCID: PMC9820761 DOI: 10.3390/ijms24010503] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Muscle unloading leads to signaling alterations that cause muscle atrophy and weakness. The cellular energy sensor AMPK can regulate myofiber-type shift, calcium-dependent signaling and ubiquitin-proteasome system markers. We hypothesized that the prevention of p-AMPK downregulation during the first week of muscle unloading would impede atrophy development and the slow-to-fast shift of soleus muscle fibers, and the aim of the study was to test this hypothesis. Thirty-two male Wistar rats were randomly assigned to four groups: placebo control (C), control rats treated with metformin (C + M), 7 days of hindlimb suspension (HS) + placebo (7HS), and 7 days of HS + metformin administration (7HS + M). In the soleus of the 7HS rats, we detected a slow-to-fast fiber-type shift as well as a significant downregulation of MEF-2D and p300 in the nuclei. In the 7HS group, we also found decreases in p-ACC (AMPK target) protein level and in the expression of E3 ubiquitin ligases and p-CaMK II protein level vs. the C group. The 7-day metformin treatment for soleus muscle unloading (1) prevented slow-to-fast fiber-type shift; (2) counteracted changes in the p-ACC protein level; (3) hindered changes in the nuclear protein level of the slow myosin expression activators MEF-2D and p300, but did not affect NFATc1 signaling; and (4) attenuated the unloading-induced upregulation of MuRF-1, atrogin-1, ubiquitin and myostatin mRNA expression, but did not prevent soleus muscle atrophy. Thus, metformin treatment during muscle disuse could be useful to prevent the decrease in the percentage of slow-type fatigue-resistant muscle fibers.
Collapse
|
25
|
Kinase signalling in excitatory neurons regulates sleep quantity and depth. Nature 2022; 612:512-518. [PMID: 36477539 DOI: 10.1038/s41586-022-05450-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/14/2022] [Indexed: 12/12/2022]
Abstract
Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1-SIK3-HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.
Collapse
|
26
|
Liu M, Liu Y, Li X, Pei M, Han M, Qi F. Dexmedetomidine inhibits abnormal muscle hypertrophy of myofascial trigger points via TNF-α/ NF-κB signaling pathway in rats. Front Pharmacol 2022; 13:1031804. [PMID: 36408215 PMCID: PMC9669483 DOI: 10.3389/fphar.2022.1031804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Myofascial pain syndrome (MPS) is a chronic pain disorder with inflammation-related primarily characterized by the presence of myofascial trigger points (MTrPs). Myocyte enhancer factor 2C (MEF2C) is involved in the occurrence of a variety of skeletal muscle diseases. However, it is not yet clear if MEF2C is involved in MTrPs. The purpose of this study was to investigate whether MEF2C was involved in the inflammatory pathogenesis of MTrPs. In the present study, we used RNA sequencing (RNA-seq) to compare the differential expression of myocyte enhancer factor 2C (MEF2C) in healthy participants and MTrPs participants. The widely used rat MTrPs model was established to research the upstream and downstream regulatory mechanism of MEF2C and found that MEF2C was significantly increased in patients with MTrPs. Dexmedetomidine (Dex) was injected intramuscularly in the MTrPs animal to assess its effects on MEF2C. The expression of MEF2C protein and mRNA in skeletal muscle of rats in the MTrPs group were up-regulated. In addition, the expression of TNF- α, p-P65, MLCK, and Myocilin (MyoC) was up-regulated and the mechanical pain threshold was decreased. Peripheral TNF- α injection significantly decreased the mechanical pain threshold and increased the expression of p-P65, MLCK, MEF2C, and MyoC in healthy rats. Maslinic acid increased the mechanical pain threshold and inhibited the expression of p-P65, MLCK, MEF2C, and MyoC. In addition, peripheral injection of DEX in MTrPs rats also inhibited the expression of TNF- α, p-P65, MLCK, MEF2C, and MyoC. These results suggest that MEF2C is involved in the inflammatory pathogenesis of MTrPs and DEX serves as a potential therapeutic strategy for the treatment of MPS.
Collapse
Affiliation(s)
- Mingjian Liu
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yu Liu
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xuan Li
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Miao Pei
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
- Department of Anesthesiology Clinic, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, China
| | - Mei Han
- Department of the Quality Management, The Second Hospital of Shandong University, Jinan, China
- *Correspondence: Mei Han, ; Feng Qi,
| | - Feng Qi
- Department of Anesthesiology and Pain Clinic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Mei Han, ; Feng Qi,
| |
Collapse
|
27
|
Huang S, Zheng X, Zhang X, Jin Z, Liu S, Fu L, Niu Y. Exercise improves high-fat diet-induced metabolic disorder by promoting HDAC5 degradation through the ubiquitin-proteasome system in skeletal muscle. Appl Physiol Nutr Metab 2022; 47:1062-1074. [PMID: 35998371 DOI: 10.1139/apnm-2022-0174] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Histone deacetylase 4/5 are essential for regulating metabolic gene expression, AMPKα2 regulates HDAC4/5 activity and the expression of MuRF1 during exercise. In this study, we used wild type and AMPKα2-/- mice to explore the potential regulatory relationship between AMPKα2 and HDAC4/5 expression during exercise. Firstly, we fed C57BL/6J mice with high-fat diet for eight-week to assess the effects of high-fat diet on skeletal muscle metabolism and HDAC4/5 expression. We then performed a six-week treadmill exercise on both wild type and AMPKα2-/- mice. After exercise, the expressions of HDAC4/5 were examined in both gastrocnemius and soleus. The citrate synthase activity and proteins involved in skeletal muscle oxidative process were assessed. To determine the relationship of HDAC4/5 and skeletal muscle oxidative capacity, citrate synthase activity was assessed after silencing HDAC4/5. Moreover, HDAC5 ubiquitination and the association of MuRF1 to HDAC5 were also investigated. Our results showed that six-week exercise increased the skeletal muscle oxidative capacity and decreased HDAC4/5 expression only in soleus. HDAC5 silencing increased C2C12 cells oxidative capacity. Proteasome inhibition by MG132 abolished exercise-induced HDAC5 degradation mediated by MuRF1-ubiquitin-proteasome system. However, the UPS did not dominantly account for exercise-induced HDAC4 degradation. Exercise up-regulated MuRF1-HDAC5 association in wild type mice but not in AMPKα2-/- mice. Our results revealed that six-week exercise increased the skeletal muscle oxidative capacity and promoted HDAC5 degradation in soleus through the UPS, MuRF1 mediated HDAC5 ubiquitination. Although AMPKα2 played partial role in regulating MuRF1 expression and HDAC5 ubiquitination, exercise-induced HDAC5 degradation did not fully depend on AMPKα2.
Collapse
Affiliation(s)
- Song Huang
- Tianjin Medical University, Department of Rehabilitation, Tianjin, Tianjin, China;
| | - Xinyue Zheng
- Tianjin Medical University, Department of Rehabilitation, Tianjin, Tianjin, China;
| | - Xinyu Zhang
- Tianjin Medical University, Physiology and Pathophysiology, Tianjin, Tianjin, China;
| | - Zhe Jin
- Tianjin Yaohua binhai, School of Yaohua binhai, Tianjin, China;
| | - Sujuan Liu
- Tianjin Medical University, Tianjin, Tianjin, China;
| | - Li Fu
- Tianjin Medical University, Physiology, Tianjin, China;
| | - Yanmei Niu
- Tianjin Medical University, Tianjin, Tianjin, China;
| |
Collapse
|
28
|
Epigenetic Alterations in Sports-Related Injuries. Genes (Basel) 2022; 13:genes13081471. [PMID: 36011382 PMCID: PMC9408207 DOI: 10.3390/genes13081471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
It is a well-known fact that physical activity benefits people of all age groups. However, highly intensive training, maladaptation, improper equipment, and lack of sufficient rest lead to contusions and sports-related injuries. From the perspectives of sports professionals and those performing regular–amateur sports activities, it is important to maintain proper levels of training, without encountering frequent injuries. The bodily responses to physical stress and intensive physical activity are detected on many levels. Epigenetic modifications, including DNA methylation, histone protein methylation, acetylation, and miRNA expression occur in response to environmental changes and play fundamental roles in the regulation of cellular activities. In the current review, we summarise the available knowledge on epigenetic alterations present in tissues and organs (e.g., muscles, the brain, tendons, and bones) as a consequence of sports-related injuries. Epigenetic mechanism observations have the potential to become useful tools in sports medicine, as predictors of approaching pathophysiological alterations and injury biomarkers that have already taken place.
Collapse
|
29
|
Xue J, Fang C, Mu R, Zhuo R, Xiao Y, Qing Y, Tang J, Fang R. Potential Mechanism and Effects of Different Selenium Sources and Different Effective Microorganism Supplementation Levels on Growth Performance, Meat Quality, and Muscle Fiber Characteristics of Three-Yellow Chickens. Front Nutr 2022; 9:869540. [PMID: 35495956 PMCID: PMC9051370 DOI: 10.3389/fnut.2022.869540] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
A trial was conducted to investigate the effects of different Se sources, including sodium selenite (S-Se) and selenium yeast (Y-Se) and different effective microorganism (EM) addition levels on growth performance, meat quality, and muscle fiber characteristics of three-yellow chickens and its potential mechanism. A total of 400 birds were randomly distributed into 4 groups (S-Se, S-Se + EM, Y-Se, and Y-Se + EM groups) consisting of a 2 × 2 factorial arrangement. The main factors were the source of Se (ISe = inorganic Se: 0.2 mg/kg S-Se; OSe = organic Se: 0.2 mg/kg Y-Se) and the level of EM (HEMB = high EM: 0.5% EM; ZEMB = low EM: 0% EM). Each treatment had 5 replicates and each replicate consisted of 20 broiler chickens. The trial lasted for 70 days. The results showed that, in breast muscle, the broiler chickens fed OSe source decreased the pH24h, drip loss, shear force, perimeter, cross-sectional area, and diameter, but increased the a24h* and density compared with the broiler chickens fed ISe source (p < 0.05); broiler chickens supplied with HEMB level decreased the cross-sectional area and diameter, but increased the pH24h, a24h,* and density compared with the broiler chickens supplied with ZEMB level (p < 0.05). In thigh muscle, OSe source and HEMB level also could improve the meat quality and change muscle fiber characteristics of broiler chickens (p < 0.05). Meat quality was correlated with the muscle fiber characteristics (p < 0.05). OSe source and HEMB level could regulate the expression levels of muscle fiber-relative genes in the breast and thigh muscles (p < 0.05). In conclusion, OSe source and HEMB level could improve the meat quality of the breast and thigh muscles of three-yellow chickens by changing the muscle fiber characteristics, and they changed the muscle fiber characteristics by regulating the expression levels of muscle fiber-relative genes.
Collapse
Affiliation(s)
- Junjing Xue
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Chengkun Fang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Rui Mu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Ruiwen Zhuo
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Yuanyuan Xiao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Yiqing Qing
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Jiaxi Tang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
| | - Rejun Fang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha, China
- *Correspondence: Rejun Fang
| |
Collapse
|
30
|
Fu X, Li S, Jia M, Xu B, Yang L, Ma R, Cheng H, Yang W, Hu P. Myogenesis controlled by a long non-coding RNA 1700113A16RIK and post-transcriptional regulation. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:13. [PMID: 35366685 PMCID: PMC8977255 DOI: 10.1186/s13619-022-00114-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/22/2022] [Indexed: 01/05/2023]
Abstract
Long non-coding (lnc) RNA plays important roles in many cellular processes. The function of the vast majority of lncRNAs remains unknown. Here we identified that lncRNA-1700113A16RIK existed in skeletal muscle stem cells (MuSCs) and was significantly elevated during MuSC differentiation. Knockdown of 1700113A16RIK inhibits the differentiation of muscle stem cells. In contrast, overexpression of 1700113A16RIK promotes the differentiation of muscle stem cells. Further study shows the muscle specific transcription factor Myogenin (MyoG) positively regulates the expression of 1700113A16RIK by binding to the promoter region of 1700113A16RIK. Mechanistically, 1700113A16RIK may regulate the expression of myogenic genes by directly binding to 3'UTR of an important myogenic transcription factor MEF2D, which in turn promotes the translation of MEF2D. Taken together, our results defined 1700113A16RIK as a positive regulator of MuSC differentiation and elucidated a mechanism as to how 1700113A16RIK regulated MuSC differentiation.
Collapse
Affiliation(s)
- Xin Fu
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Sheng Li
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Minzhi Jia
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Bo Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lele Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ruimiao Ma
- Guangzhou Laboratory, Guangzhou, 510700, Guangdong, China
| | - Hong Cheng
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenjun Yang
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
| | - Ping Hu
- Spine Center, Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China. .,Guangzhou Laboratory, Guangzhou, 510700, Guangdong, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
31
|
Gomez-Sanchez JA, Patel N, Martirena F, Fazal SV, Mutschler C, Cabedo H. Emerging Role of HDACs in Regeneration and Ageing in the Peripheral Nervous System: Repair Schwann Cells as Pivotal Targets. Int J Mol Sci 2022; 23:ijms23062996. [PMID: 35328416 PMCID: PMC8951080 DOI: 10.3390/ijms23062996] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
The peripheral nervous system (PNS) has a remarkable regenerative capacity in comparison to the central nervous system (CNS), a phenomenon that is impaired during ageing. The ability of PNS axons to regenerate after injury is due to Schwann cells (SC) being reprogrammed into a repair phenotype called Repair Schwann cells. These repair SCs are crucial for supporting axonal growth after injury, myelin degradation in a process known as myelinophagy, neurotropic factor secretion, and axonal growth guidance through the formation of Büngner bands. After regeneration, repair SCs can remyelinate newly regenerated axons and support nonmyelinated axons. Increasing evidence points to an epigenetic component in the regulation of repair SC gene expression changes, which is necessary for SC reprogramming and regeneration. One of these epigenetic regulations is histone acetylation by histone acetyl transferases (HATs) or histone deacetylation by histone deacetylases (HDACs). In this review, we have focused particularly on three HDAC classes (I, II, and IV) that are Zn2+-dependent deacetylases. These HDACs are important in repair SC biology and remyelination after PNS injury. Another key aspect explored in this review is HDAC genetic compensation in SCs and novel HDAC inhibitors that are being studied to improve nerve regeneration.
Collapse
Affiliation(s)
- Jose A. Gomez-Sanchez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (N.P.); (H.C.)
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
- Correspondence: ; Tel.: +34-965-919-594
| | - Nikiben Patel
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (N.P.); (H.C.)
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Fernanda Martirena
- Department of Hematology, General University Hospital of Elda, 03600 Elda, Spain;
| | - Shaline V. Fazal
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK; (S.V.F.); (C.M.)
- Wellcome—MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Clara Mutschler
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK; (S.V.F.); (C.M.)
| | - Hugo Cabedo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (N.P.); (H.C.)
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
32
|
Lavin KM, Coen PM, Baptista LC, Bell MB, Drummer D, Harper SA, Lixandrão ME, McAdam JS, O’Bryan SM, Ramos S, Roberts LM, Vega RB, Goodpaster BH, Bamman MM, Buford TW. State of Knowledge on Molecular Adaptations to Exercise in Humans: Historical Perspectives and Future Directions. Compr Physiol 2022; 12:3193-3279. [PMID: 35578962 PMCID: PMC9186317 DOI: 10.1002/cphy.c200033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For centuries, regular exercise has been acknowledged as a potent stimulus to promote, maintain, and restore healthy functioning of nearly every physiological system of the human body. With advancing understanding of the complexity of human physiology, continually evolving methodological possibilities, and an increasingly dire public health situation, the study of exercise as a preventative or therapeutic treatment has never been more interdisciplinary, or more impactful. During the early stages of the NIH Common Fund Molecular Transducers of Physical Activity Consortium (MoTrPAC) Initiative, the field is well-positioned to build substantially upon the existing understanding of the mechanisms underlying benefits associated with exercise. Thus, we present a comprehensive body of the knowledge detailing the current literature basis surrounding the molecular adaptations to exercise in humans to provide a view of the state of the field at this critical juncture, as well as a resource for scientists bringing external expertise to the field of exercise physiology. In reviewing current literature related to molecular and cellular processes underlying exercise-induced benefits and adaptations, we also draw attention to existing knowledge gaps warranting continued research effort. © 2021 American Physiological Society. Compr Physiol 12:3193-3279, 2022.
Collapse
Affiliation(s)
- Kaleen M. Lavin
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Liliana C. Baptista
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Margaret B. Bell
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin Drummer
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara A. Harper
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manoel E. Lixandrão
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeremy S. McAdam
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samia M. O’Bryan
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sofhia Ramos
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Lisa M. Roberts
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rick B. Vega
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Bret H. Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Marcas M. Bamman
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Thomas W. Buford
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
33
|
Ruiz A, Benucci S, Duthaler U, Bachmann C, Franchini M, Noreen F, Pietrangelo L, Protasi F, Treves S, Zorzato F. Improvement of muscle strength in a mouse model for congenital myopathy treated with HDAC and DNA methyltransferase inhibitors. eLife 2022; 11:73718. [PMID: 35238775 PMCID: PMC8956288 DOI: 10.7554/elife.73718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
To date there are no therapies for patients with congenital myopathies, muscle disorders causing poor quality of life of affected individuals. In approximately 30% of the cases, patients with congenital myopathies carry either dominant or recessive mutations in the RYR1 gene; recessive RYR1 mutations are accompanied by reduction of RyR1 expression and content in skeletal muscles and are associated with fiber hypotrophy and muscle weakness. Importantly, muscles of patients with recessive RYR1 mutations exhibit increased content of class II histone de-acetylases and of DNA genomic methylation. We recently created a mouse model knocked-in for the p.Q1970fsX16+p.A4329D RyR1 mutations, which are isogenic to those carried by a severely affected child suffering from a recessive form of RyR1-related multi-mini core disease. The phenotype of the RyR1 mutant mice recapitulates many aspects of the clinical picture of patients carrying recessive RYR1 mutations. We treated the compound heterozygous mice with a combination of two drugs targeting DNA methylases and class II histone de-acetylases. Here we show that treatment of the mutant mice with drugs targeting epigenetic enzymes improves muscle strength, RyR1 protein content and muscle ultrastructure. This study provides proof of concept for the pharmacological treatment of patients with congenital myopathies linked to recessive RYR1 mutations.
Collapse
Affiliation(s)
- Alexis Ruiz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sofia Benucci
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Urs Duthaler
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Christoph Bachmann
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Martina Franchini
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Laura Pietrangelo
- Department of Neuroscience, Imaging and Clinical Science, University G d' Annunzio of Chieti, Chieti, Italy
| | - Feliciano Protasi
- Department of Neuroscience, Imaging and Clinical Science, University G d' Annunzio of Chieti, Chieti, Italy
| | - Susan Treves
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Francesco Zorzato
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
34
|
Renzini A, D’Onghia M, Coletti D, Moresi V. Histone Deacetylases as Modulators of the Crosstalk Between Skeletal Muscle and Other Organs. Front Physiol 2022; 13:706003. [PMID: 35250605 PMCID: PMC8895239 DOI: 10.3389/fphys.2022.706003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle plays a major role in controlling body mass and metabolism: it is the most abundant tissue of the body and a major source of humoral factors; in addition, it is primarily responsible for glucose uptake and storage, as well as for protein metabolism. Muscle acts as a metabolic hub, in a crosstalk with other organs and tissues, such as the liver, the brain, and fat tissue. Cytokines, adipokines, and myokines are pivotal mediators of such crosstalk. Many of these circulating factors modulate histone deacetylase (HDAC) expression and/or activity. HDACs form a numerous family of enzymes, divided into four classes based on their homology to their orthologs in yeast. Eleven family members are considered classic HDACs, with a highly conserved deacetylase domain, and fall into Classes I, II, and IV, while class III members are named Sirtuins and are structurally and mechanistically distinct from the members of the other classes. HDACs are key regulators of skeletal muscle metabolism, both in physiological conditions and following metabolic stress, participating in the highly dynamic adaptative responses of the muscle to external stimuli. In turn, HDAC expression and activity are closely regulated by the metabolic demands of the skeletal muscle. For instance, NAD+ levels link Class III (Sirtuin) enzymatic activity to the energy status of the cell, and starvation or exercise affect Class II HDAC stability and intracellular localization. SUMOylation or phosphorylation of Class II HDACs are modulated by circulating factors, thus establishing a bidirectional link between HDAC activity and endocrine, paracrine, and autocrine factors. Indeed, besides being targets of adipo-myokines, HDACs affect the synthesis of myokines by skeletal muscle, altering the composition of the humoral milieu and ultimately contributing to the muscle functioning as an endocrine organ. In this review, we discuss recent findings on the interplay between HDACs and circulating factors, in relation to skeletal muscle metabolism and its adaptative response to energy demand. We believe that enhancing knowledge on the specific functions of HDACs may have clinical implications leading to the use of improved HDAC inhibitors for the treatment of metabolic syndromes or aging.
Collapse
Affiliation(s)
- Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Marco D’Onghia
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Viviana Moresi
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Institute of Nanotechnology (Nanotec), National Research Council, Rome, Italy
| |
Collapse
|
35
|
Velasco-Aviles S, Patel N, Casillas-Bajo A, Frutos-Rincón L, Velasco E, Gallar J, Arthur-Farraj P, Gomez-Sanchez JA, Cabedo H. A genetic compensatory mechanism regulated by Jun and Mef2d modulates the expression of distinct class IIa Hdacs to ensure peripheral nerve myelination and repair. eLife 2022; 11:e72917. [PMID: 35076395 PMCID: PMC8853665 DOI: 10.7554/elife.72917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/24/2022] [Indexed: 11/21/2022] Open
Abstract
The class IIa histone deacetylases (HDACs) have pivotal roles in the development of different tissues. Of this family, Schwann cells express Hdac4, 5, and 7 but not Hdac9. Here, we show that a transcription factor regulated genetic compensatory mechanism within this family of proteins, blocks negative regulators of myelination ensuring peripheral nerve developmental myelination and remyelination after injury. Thus, when Hdac4 and 5 are knocked-out from Schwann cells in mice, a JUN-dependent mechanism induces the compensatory overexpression of Hdac7 permitting, although with a delay, the formation of the myelin sheath. When Hdac4, 5, and 7 are simultaneously removed, the myocyte-specific enhancer-factor d (MEF2D) binds to the promoter and induces the de novo expression of Hdac9, and although several melanocytic lineage genes are misexpressed and Remak bundle structure is disrupted, myelination proceeds after a long delay. Thus, our data unveil a finely tuned compensatory mechanism within the class IIa Hdac family, coordinated by distinct transcription factors, that guarantees the ability of Schwann cells to myelinate during development and remyelinate after nerve injury.
Collapse
Affiliation(s)
- Sergio Velasco-Aviles
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
| | - Nikiben Patel
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
| | - Angeles Casillas-Bajo
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
| | - Laura Frutos-Rincón
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- The European University of Brain and Technology-NeurotechEUAlicanteSpain
| | - Enrique Velasco
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- The European University of Brain and Technology-NeurotechEUAlicanteSpain
| | - Juana Gallar
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- The European University of Brain and Technology-NeurotechEUAlicanteSpain
- RICORS en enfermedades inflamatoriasSant Joan d'AlacantSpain
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| | | | - Hugo Cabedo
- Instituto de Neurociencias de Alicante UMH-CSICAlicanteSpain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
| |
Collapse
|
36
|
Chambers PJ, Juracic ES, Fajardo VA, Tupling AR. The role of SERCA and sarcolipin in adaptive muscle remodeling. Am J Physiol Cell Physiol 2022; 322:C382-C394. [PMID: 35044855 DOI: 10.1152/ajpcell.00198.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sarcolipin (SLN) is a small integral membrane protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, where large increases in SLN content are found in response to development, atrophy, overload and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious to muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism which protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fibre type shifts and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.
Collapse
Affiliation(s)
- Paige J Chambers
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Emma S Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Val A Fajardo
- Department Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
37
|
Pozo MR, Meredith GW, Entcheva E. Human iPSC-Cardiomyocytes as an Experimental Model to Study Epigenetic Modifiers of Electrophysiology. Cells 2022; 11:200. [PMID: 35053315 PMCID: PMC8774228 DOI: 10.3390/cells11020200] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 02/04/2023] Open
Abstract
The epigenetic landscape and the responses to pharmacological epigenetic regulators in each human are unique. Classes of epigenetic writers and erasers, such as histone acetyltransferases, HATs, and histone deacetylases, HDACs, control DNA acetylation/deacetylation and chromatin accessibility, thus exerting transcriptional control in a tissue- and person-specific manner. Rapid development of novel pharmacological agents in clinical testing-HDAC inhibitors (HDACi)-targets these master regulators as common means of therapeutic intervention in cancer and immune diseases. The action of these epigenetic modulators is much less explored for cardiac tissue, yet all new drugs need to be tested for cardiotoxicity. To advance our understanding of chromatin regulation in the heart, and specifically how modulation of DNA acetylation state may affect functional electrophysiological responses, human-induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology can be leveraged as a scalable, high-throughput platform with ability to provide patient-specific insights. This review covers relevant background on the known roles of HATs and HDACs in the heart, the current state of HDACi development, applications, and any adverse cardiac events; it also summarizes relevant differential gene expression data for the adult human heart vs. hiPSC-CMs along with initial transcriptional and functional results from using this new experimental platform to yield insights on epigenetic control of the heart. We focus on the multitude of methodologies and workflows needed to quantify responses to HDACis in hiPSC-CMs. This overview can help highlight the power and the limitations of hiPSC-CMs as a scalable experimental model in capturing epigenetic responses relevant to the human heart.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA; (M.R.P.); (G.W.M.)
| |
Collapse
|
38
|
Sakuma S, Zhu EY, Raices M, Zhang P, Murad R, D'Angelo MA. Loss of Nup210 results in muscle repair delays and age-associated alterations in muscle integrity. Life Sci Alliance 2021; 5:5/3/e202101216. [PMID: 34911810 PMCID: PMC8711851 DOI: 10.26508/lsa.202101216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/29/2022] Open
Abstract
This study describes the the role of a nuclear pore complex protein in mammalian in skeletal muscle maintenance, repair, and function. Nuclear pore complexes, the channels connecting the nucleus with the cytoplasm, are built by multiple copies of ∼30 proteins called nucleoporins. Recent evidence has exposed that nucleoporins can play cell type-specific functions. Despite novel discoveries into the cellular functions of nucleoporins, their role in the regulation of mammalian tissue physiology remains mostly unexplored because of a limited number of nucleoporin mouse models. Here we show that ablation of Nup210/Gp210, a nucleoporin previously identified to play a role in myoblast differentiation and Zebrafish muscle maturation, is dispensable for skeletal muscle formation and growth in mice. We found that although primary satellite cells from Nup210 knockout mice can differentiate, these animals show delayed muscle repair after injury. Moreover, Nup210 knockout mice display an increased percentage of centrally nucleated fibers and abnormal fiber type distribution as they age. Muscle function experiments also exposed that Nup210 is required for muscle endurance during voluntary running. Our findings indicate that in mammals, Nup210 is important for the maintenance of skeletal muscle integrity and for proper muscle function providing novel insights into the in vivo roles of nuclear pore complex components.
Collapse
Affiliation(s)
- Stephen Sakuma
- Cell and Molecular Biology of Cancer Program, National Cancer Institute (NCI)-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ethan Ys Zhu
- Cell and Molecular Biology of Cancer Program, National Cancer Institute (NCI)-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Marcela Raices
- Cell and Molecular Biology of Cancer Program, National Cancer Institute (NCI)-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Pan Zhang
- Bioinformatics Core, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rabi Murad
- Bioinformatics Core, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Maximiliano A D'Angelo
- Cell and Molecular Biology of Cancer Program, National Cancer Institute (NCI)-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
39
|
Paramonova II, Vilchinskaya NA, Shenkman BS. HDAC4 Is Indispensable for Reduced Slow Myosin Expression at the Early Stage of Hindlimb Unloading in Rat Soleus Muscle. Pharmaceuticals (Basel) 2021; 14:ph14111167. [PMID: 34832949 PMCID: PMC8617770 DOI: 10.3390/ph14111167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/05/2021] [Accepted: 11/13/2021] [Indexed: 11/16/2022] Open
Abstract
It is well known that reduced contractile activity of the main postural soleus muscle during long-term bedrest, immobilization, hindlimb unloading, and space flight leads to increased expression of fast isoforms and decreased expression of the slow isoform of myosin heavy chain (MyHC). The signaling cascade such as HDAC4/MEF2-D pathway is well-known to take part in regulating MyHC I gene expression. Earlier, we found a significant increase of HDAC4 in myonuclei due to AMPK dephosphorylation during 24 h of hindlimb unloading via hindlimb suspension (HU) and it had a significant impact on the expression of MyHC isoforms in rat soleus causing a decrease in MyHC I(β) pre-mRNA and mRNA expression as well as MyHC IIa mRNA expression. We hypothesized that dephosphorylated HDAC4 translocates into the nuclei and can lead to a reduced expression of slow MyHC. To test this hypothesis, Wistar rats were treated with HDAC4 inhibitor (Tasquinimod) for 7 days before HU as well as during 24 h of HU. We discovered that Tasquinimod treatment prevented a decrease in pre-mRNA expression of MyHC I. Furthermore, 24 h of hindlimb suspension resulted in HDAC4 nuclear accumulation of rat soleus but Tasquinimod pretreatment prevented this accumulation. The results of the study indicate that HDAC4 after 24 h of HU had a significant impact on the precursor MyHC I mRNA expression in rat soleus.
Collapse
|
40
|
Bengtsen M, Winje IM, Eftestøl E, Landskron J, Sun C, Nygård K, Domanska D, Millay DP, Meza-Zepeda LA, Gundersen K. Comparing the epigenetic landscape in myonuclei purified with a PCM1 antibody from a fast/glycolytic and a slow/oxidative muscle. PLoS Genet 2021; 17:e1009907. [PMID: 34752468 PMCID: PMC8604348 DOI: 10.1371/journal.pgen.1009907] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/19/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023] Open
Abstract
Muscle cells have different phenotypes adapted to different usage, and can be grossly divided into fast/glycolytic and slow/oxidative types. While most muscles contain a mixture of such fiber types, we aimed at providing a genome-wide analysis of the epigenetic landscape by ChIP-Seq in two muscle extremes, the fast/glycolytic extensor digitorum longus (EDL) and slow/oxidative soleus muscles. Muscle is a heterogeneous tissue where up to 60% of the nuclei can be of a different origin. Since cellular homogeneity is critical in epigenome-wide association studies we developed a new method for purifying skeletal muscle nuclei from whole tissue, based on the nuclear envelope protein Pericentriolar material 1 (PCM1) being a specific marker for myonuclei. Using antibody labelling and a magnetic-assisted sorting approach, we were able to sort out myonuclei with 95% purity in muscles from mice, rats and humans. The sorting eliminated influence from the other cell types in the tissue and improved the myo-specific signal. A genome-wide comparison of the epigenetic landscape in EDL and soleus reflected the differences in the functional properties of the two muscles, and revealed distinct regulatory programs involving distal enhancers, including a glycolytic super-enhancer in the EDL. The two muscles were also regulated by different sets of transcription factors; e.g. in soleus, binding sites for MEF2C, NFATC2 and PPARA were enriched, while in EDL MYOD1 and SIX1 binding sites were found to be overrepresented. In addition, more novel transcription factors for muscle regulation such as members of the MAF family, ZFX and ZBTB14 were identified. Complex tissues like skeletal muscle contain a variety of cells which confound the analysis of each cell type when based on homogenates, thus only about half of the cell nuclei in muscles reside inside the muscle cells. We here describe a labelling and sorting technique that allowed us to study the epigenetic landscape in purified muscle cell nuclei leaving the other cell types out. Differences between a fast/glycolytic and a slow/oxidative muscle were studied. While all skeletal muscle fibers have a similar make up and basic function, they differ in their physiology and the way they are used. Thus, some fibers are fast contracting but fatigable, and are used for short lasting explosive tasks such as sprinting. Other fibers are slow and are used for more prolonged tasks such as standing or long distance running. Since fiber type correlate with metabolic profile these features can also be related to metabolic diseases. We here show that the epigenetic landscape differed in gene loci corresponding to the differences in functional properties, and revealed that the two types are enriched in different gene regulatory networks. Exercise can alter muscle phenotype, and the epigenetic landscape might be related to how plastic different properties are.
Collapse
Affiliation(s)
- Mads Bengtsen
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Einar Eftestøl
- Department of Biosciences, University of Oslo, Oslo, Norway
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | | | - Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kamilla Nygård
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Diana Domanska
- Department of Pathology, University of Oslo, Oslo, Norway
| | - Douglas P. Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Leonardo A. Meza-Zepeda
- Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
41
|
Hyttinen J, Blasiak J, Tavi P, Kaarniranta K. Therapeutic potential of PGC-1α in age-related macular degeneration (AMD) - the involvement of mitochondrial quality control, autophagy, and antioxidant response. Expert Opin Ther Targets 2021; 25:773-785. [PMID: 34637373 DOI: 10.1080/14728222.2021.1991913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is the leading, cause of sight loss in the elderly in the Western world. Most patients remain still without any treatment options. The targeting of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a transcription co-factor, is a putative therapy against AMD. AREAS COVERED The characteristics of AMD and their possible connection with PGC-1α as well as the transcriptional and post-transcriptional control of PGC-1α are discussed. The PGC-1α-driven control of mitochondrial functions, and its involvement in autophagy and antioxidant responses are also examined. Therapeutic possibilities via drugs and epigenetic approaches to enhance PGC-1α expression are discussed. Authors conducted a search of literature mainly from the recent decade from the PubMed database. EXPERT OPINION Therapy options in AMD could include PGC-1α activation or stabilization. This could be achieved by a direct elevation of PGC-1α activity, a stabilization or modification of its upstream activators and inhibitors by chemical compounds, like 5-Aminoimidazole-4-carboxamide riboside, metformin, and resveratrol. Furthermore, manipulations with epigenetic modifiers of PGC-1α expression, including miRNAs, e.g. miR-204, are considered. A therapy aimed at PGC-1α up-regulation may be possible in other disorders besides AMD, if they are associated with disturbances in the mitochondria-antioxidant response-autophagy axis.
Collapse
Affiliation(s)
- Juha Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Sciences, University of Lodz, Lodz, Poland
| | - Pasi Tavi
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
42
|
Abbadi D, Andrews JJ, Katsara O, Schneider RJ. AUF1 gene transfer increases exercise performance and improves skeletal muscle deficit in adult mice. Mol Ther Methods Clin Dev 2021; 22:222-236. [PMID: 34485607 PMCID: PMC8399044 DOI: 10.1016/j.omtm.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022]
Abstract
Muscle function and mass begin declining in adults long before evidence of sarcopenia and include reduced mitochondrial function, although much remains to be characterized. We found that mRNA decay factor AU-rich mRNA binding factor 1 (AUF1), which stimulates myogenesis, is strongly reduced in skeletal muscle of adult and older mice in the absence of evidence of sarcopenia. Muscle-specific adeno-associated virus (AAV)8-AUF1 gene therapy increased expression of AUF1, muscle function, and mass. AAV8 AUF1 muscle gene transfer in 12-month-old mice increased the levels of activated muscle stem (satellite) cells, increased muscle mass, reduced markers of muscle atrophy, increased markers of mitochondrial content and muscle fiber oxidative capacity, and enhanced exercise performance to levels of 3-month-old mice. With wild-type and AUF1 knockout mice and cultured myoblasts, AUF1 supplementation of muscle fibers was found to increase expression of Peroxisome Proliferator-activated Receptor Gamma Co-activator 1-alpha (PGC1α), a major effector of skeletal muscle mitochondrial oxidative metabolism. AUF1 stabilized and increased translation of the pgc1α mRNA, which is strongly reduced in adult muscle in the absence of AUF1 supplementation. Skeletal muscle-specific gene transfer of AUF1 therefore restores muscle mass, increases exercise endurance, and may provide a therapeutic strategy for age-related muscle loss.
Collapse
Affiliation(s)
- Dounia Abbadi
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - John J. Andrews
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Olga Katsara
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Robert J. Schneider
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| |
Collapse
|
43
|
Zhou H, Su H, Chen W. Neddylation Regulates Class IIa and III Histone Deacetylases to Mediate Myoblast Differentiation. Int J Mol Sci 2021; 22:ijms22179509. [PMID: 34502418 PMCID: PMC8431717 DOI: 10.3390/ijms22179509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 11/16/2022] Open
Abstract
As the largest tissue in the body, skeletal muscle has multiple functions in movement and energy metabolism. Skeletal myogenesis is controlled by a transcriptional cascade including a set of muscle regulatory factors (MRFs) that includes Myogenic Differentiation 1 (MYOD1), Myocyte Enhancer Factor 2 (MEF2), and Myogenin (MYOG), which direct the fusion of myogenic myoblasts into multinucleated myotubes. Neddylation is a posttranslational modification that covalently conjugates ubiquitin-like NEDD8 (neural precursor cell expressed, developmentally downregulated 8) to protein targets. Inhibition of neddylation impairs muscle differentiation; however, the underlying molecular mechanisms remain less explored. Here, we report that neddylation is temporally regulated during myoblast differentiation. Inhibition of neddylation through pharmacological blockade using MLN4924 (Pevonedistat) or genetic deletion of NEDD8 Activating Enzyme E1 Subunit 1 (NAE1), a subunit of the E1 neddylation-activating enzyme, blocks terminal myoblast differentiation partially through repressing MYOG expression. Mechanistically, we found that neddylation deficiency enhances the mRNA and protein expressions of class IIa histone deacetylases 4 and 5 (HDAC4 and 5) and prevents the downregulation and nuclear export of class III HDAC (NAD-Dependent Protein Deacetylase Sirtuin-1, SIRT1), all of which have been shown to repress MYOD1-mediated MYOG transcriptional activation. Together, our findings for the first time identify the crucial role of neddylation in mediating class IIa and III HDAC co-repressors to control myogenic program and provide new insights into the mechanisms of muscle disease and regeneration.
Collapse
Affiliation(s)
- Hongyi Zhou
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Correspondence: ; Tel.: +1-706-721-8779
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
44
|
Abstract
Since ancient times, the health benefits of regular physical activity/exercise have been recognized and the classic studies of Morris and Paffenbarger provided the epidemiological evidence in support of such an association. Cardiorespiratory fitness, often measured by maximal oxygen uptake, and habitual physical activity levels are inversely related to mortality. Thus, studies exploring the biological bases of the health benefits of exercise have largely focused on the cardiovascular system and skeletal muscle (mass and metabolism), although there is increasing evidence that multiple tissues and organ systems are influenced by regular exercise. Communication between contracting skeletal muscle and multiple organs has been implicated in exercise benefits, as indeed has other interorgan "cross-talk." The application of molecular biology techniques and "omics" approaches to questions in exercise biology has opened new lines of investigation to better understand the beneficial effects of exercise and, in so doing, inform the optimization of exercise regimens and the identification of novel therapeutic strategies to enhance health and well-being.
Collapse
Affiliation(s)
- Mark Hargreaves
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Giagnorio E, Malacarne C, Mantegazza R, Bonanno S, Marcuzzo S. MyomiRs and their multifaceted regulatory roles in muscle homeostasis and amyotrophic lateral sclerosis. J Cell Sci 2021; 134:269129. [PMID: 34137441 DOI: 10.1242/jcs.258349] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by loss of both upper and lower motor neurons (MNs). The main clinical features of ALS are motor function impairment, progressive muscle weakness, muscle atrophy and, ultimately, paralysis. Intrinsic skeletal muscle deterioration plays a crucial role in the disease and contributes to ALS progression. Currently, there are no effective treatments for ALS, highlighting the need to obtain a deeper understanding of the molecular events underlying degeneration of both MNs and muscle tissue, with the aim of developing successful therapies. Muscle tissue is enriched in a group of microRNAs called myomiRs, which are effective regulators of muscle homeostasis, plasticity and myogenesis in both physiological and pathological conditions. After providing an overview of ALS pathophysiology, with a focus on the role of skeletal muscle, we review the current literature on myomiR network dysregulation as a contributing factor to myogenic perturbations and muscle atrophy in ALS. We argue that, in view of their critical regulatory function at the interface between MNs and skeletal muscle fiber, myomiRs are worthy of further investigation as potential molecular targets of therapeutic strategies to improve ALS symptoms and counteract disease progression.
Collapse
Affiliation(s)
- Eleonora Giagnorio
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.,PhD program in Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Claudia Malacarne
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.,PhD program in Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Silvia Bonanno
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Stefania Marcuzzo
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| |
Collapse
|
46
|
Di Giorgio E, Paluvai H, Dalla E, Ranzino L, Renzini A, Moresi V, Minisini M, Picco R, Brancolini C. HDAC4 degradation during senescence unleashes an epigenetic program driven by AP-1/p300 at selected enhancers and super-enhancers. Genome Biol 2021; 22:129. [PMID: 33966634 PMCID: PMC8108360 DOI: 10.1186/s13059-021-02340-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/06/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cellular senescence is a permanent state of replicative arrest defined by a specific pattern of gene expression. The epigenome in senescent cells is sculptured in order to sustain the new transcriptional requirements, particularly at enhancers and super-enhancers. How these distal regulatory elements are dynamically modulated is not completely defined. RESULTS Enhancer regions are defined by the presence of H3K27 acetylation marks, which can be modulated by class IIa HDACs, as part of multi-protein complexes. Here, we explore the regulation of class IIa HDACs in different models of senescence. We find that HDAC4 is polyubiquitylated and degraded during all types of senescence and it selectively binds and monitors H3K27ac levels at specific enhancers and super-enhancers that supervise the senescent transcriptome. Frequently, these HDAC4-modulated elements are also monitored by AP-1/p300. The deletion of HDAC4 in transformed cells which have bypassed oncogene-induced senescence is coupled to the re-appearance of senescence and the execution of the AP-1/p300 epigenetic program. CONCLUSIONS Overall, our manuscript highlights a role of HDAC4 as an epigenetic reader and controller of enhancers and super-enhancers that supervise the senescence program. More generally, we unveil an epigenetic checkpoint that has important consequences in aging and cancer.
Collapse
Affiliation(s)
- Eros Di Giorgio
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy
| | | | - Emiliano Dalla
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy
| | - Liliana Ranzino
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy
| | - Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, via Antonio Scarpa 16, 00161, Rome, Italy
| | - Viviana Moresi
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, via Antonio Scarpa 16, 00161, Rome, Italy
| | - Martina Minisini
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy
| | - Raffaella Picco
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy
| | - Claudio Brancolini
- Department of Medicine, Università degli Studi di Udine, p.le Kolbe 4, 33100, Udine, Italy.
| |
Collapse
|
47
|
Peng Y, Xu M, Dou M, Shi X, Yang G, Li X. MicroRNA-129-5p inhibits C2C12 myogenesis and represses slow fiber gene expression in vitro. Am J Physiol Cell Physiol 2021; 320:C1031-C1041. [PMID: 33826407 DOI: 10.1152/ajpcell.00578.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The miR-129 family is widely reported as tumor repressors, although their roles in skeletal muscle have not been fully investigated. Here, the function and mechanism of miR-129-5p in skeletal muscle, a member of the miR-129 family, were explored using C2C12 cell line. Our study showed that miR-129-5p was widely detected in mouse tissues, with the highest expression in skeletal muscle. Gain- and loss-of-function study showed that miR-129-5p could negatively regulate myogenic differentiation, indicated by reduced ratio of MyHC-positive myofibers and repressed expression of myogenic genes, such as MyoD, MyoG, and MyHC. Furthermore, miR-129-5p was more enriched in fast extensor digitorum longus (EDL) than in slow soleus (SOL). Enhanced miR-129-5p could significantly reduce the expression of mitochondrial cox family, together with that of MyHC I, and knockdown of miR-129-5p conversely increased the expression of cox genes and MyHC I. Mechanistically, miR-129-5p directly targeted the 3'-UTR of Mef2a, which was suppressed by miR-129-5p agomir at both mRNA and protein levels in C2C12 cells. Moreover, overexpression of Mef2a could rescue the inhibitory effects of miR-129-5p on the expression of myogenic factors and MyHC I. Collectively, our data revealed that miR-129-5p is a negative regulator of myogenic differentiation and slow fiber gene expression, thus affecting body metabolic homeostasis.
Collapse
Affiliation(s)
- Ying Peng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| | - Meixue Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| | - Mingle Dou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| | - Xin'E Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| | - Xiao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi, People's Republic of China
| |
Collapse
|
48
|
Loss of Mef2D function enhances TLR induced IL-10 production in macrophages. Biosci Rep 2021; 40:225925. [PMID: 32725155 PMCID: PMC7442974 DOI: 10.1042/bsr20201859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
Mef2 transcription factors comprise a family of four different isoforms that regulate a number of processes including neuronal and muscle development. While roles for Mef2C and Mef2D have been described in B-cell development their role in immunity has not been extensively studied. In innate immune cells such as macrophages, TLRs drive the production of both pro- and anti-inflammatory cytokines. IL-10 is an important anti-inflammatory cytokine produced by macrophages and it establishes an autocrine feedback loop to inhibit pro-inflammatory cytokine production. We show here that macrophages from Mef2D knockout mice have elevated levels of IL-10 mRNA induction compared with wild-type cells following LPS stimulation. The secretion of IL-10 was also higher from Mef2D knockout macrophages and this correlated to a reduction in the secretion of TNF, IL-6 and IL-12p40. The use of an IL-10 neutralising antibody showed that this reduction in pro-inflammatory cytokine production in the Mef2D knockouts was IL-10 dependent. As the IL-10 promoter has previously been reported to contain a potential binding site for Mef2D, it is possible that the binding of other Mef2 isoforms in the absence of Mef2D may result in a higher activation of the IL-10 gene. Further studies with compound Mef2 isoforms would be required to address this. We also show that Mef2D is highly expressed in the thymus, but that loss of Mef2D does not affect thymic T-cell development or the production of IFNγ from CD8 T cells.
Collapse
|
49
|
Xiao L, Liu J, Sun Z, Yin Y, Mao Y, Xu D, Liu L, Xu Z, Guo Q, Ding C, Sun W, Yang L, Zhou Z, Zhou D, Fu T, Zhou W, Zhu Y, Chen XW, Li JZ, Chen S, Xie X, Gan Z. AMPK-dependent and -independent coordination of mitochondrial function and muscle fiber type by FNIP1. PLoS Genet 2021; 17:e1009488. [PMID: 33780446 PMCID: PMC8031738 DOI: 10.1371/journal.pgen.1009488] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 04/08/2021] [Accepted: 03/12/2021] [Indexed: 01/01/2023] Open
Abstract
Mitochondria are essential for maintaining skeletal muscle metabolic homeostasis during adaptive response to a myriad of physiologic or pathophysiological stresses. The mechanisms by which mitochondrial function and contractile fiber type are concordantly regulated to ensure muscle function remain poorly understood. Evidence is emerging that the Folliculin interacting protein 1 (Fnip1) is involved in skeletal muscle fiber type specification, function, and disease. In this study, Fnip1 was specifically expressed in skeletal muscle in Fnip1-transgenic (Fnip1Tg) mice. Fnip1Tg mice were crossed with Fnip1-knockout (Fnip1KO) mice to generate Fnip1TgKO mice expressing Fnip1 only in skeletal muscle but not in other tissues. Our results indicate that, in addition to the known role in type I fiber program, FNIP1 exerts control upon muscle mitochondrial oxidative program through AMPK signaling. Indeed, basal levels of FNIP1 are sufficient to inhibit AMPK but not mTORC1 activity in skeletal muscle cells. Gain-of-function and loss-of-function strategies in mice, together with assessment of primary muscle cells, demonstrated that skeletal muscle mitochondrial program is suppressed via the inhibitory actions of FNIP1 on AMPK. Surprisingly, the FNIP1 actions on type I fiber program is independent of AMPK and its downstream PGC-1α. These studies provide a vital framework for understanding the intrinsic role of FNIP1 as a crucial factor in the concerted regulation of mitochondrial function and muscle fiber type that determine muscle fitness. Mitochondria provide an essential source of energy to drive cellular processes and the function of mitochondria is particularly important in skeletal muscle, a metabolically demanding tissue that depends critically on mitochondria, accounting for ~40% of total body mass. In this study, we discovered an essential function of adaptor protein FNIP1 in the coordinated regulation of the mitochondrial and structural programs controlling muscle fitness. Using both gain-of-function and loss-of-function strategies in mice and muscle cells, we provide clear genetic data that demonstrate FNIP1-dependent signaling is crucial for muscle mitochondrial remodeling as well as type I muscle fiber specification. We also uncover that FNIP1 exerts control upon muscle mitochondrial program through AMPK but not mTORC1 signaling. Furthermore, we demonstrate that FNIP1 acts independently of PGC-1α to regulate fiber type specification. Hence, our study emphasizes FNIP1 as a dominant factor that coordinates mitochondrial and muscle fiber type programs that govern muscle fitness.
Collapse
Affiliation(s)
- Liwei Xiao
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Jing Liu
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zongchao Sun
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yujing Yin
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yan Mao
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Dengqiu Xu
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Lin Liu
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zhisheng Xu
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Qiqi Guo
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Chenyun Ding
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wanping Sun
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Likun Yang
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zheng Zhou
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Danxia Zhou
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Tingting Fu
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wenjing Zhou
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yuangang Zhu
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao-Wei Chen
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Shuai Chen
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xiaoduo Xie
- Department of Biochemistry, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Zhenji Gan
- MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
- * E-mail:
| |
Collapse
|
50
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|