1
|
Sermersheim TJ, Phillips LJ, Evans PL, Kahn BB, Welc SS, Witczak CA. Regulation of injury-induced skeletal myofiber regeneration by glucose transporter 4 (GLUT4). Skelet Muscle 2024; 14:33. [PMID: 39695900 DOI: 10.1186/s13395-024-00366-y] [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: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Insulin resistance and type 2 diabetes impair cellular regeneration in multiple tissues including skeletal muscle. The molecular basis for this impairment is largely unknown. Glucose uptake via glucose transporter GLUT4 is impaired in insulin resistance. In healthy muscle, acute injury stimulates glucose uptake. Whether decreased glucose uptake via GLUT4 impairs muscle regeneration is presently unknown. The goal of this study was to determine whether GLUT4 regulates muscle glucose uptake and/or regeneration following acute injury. METHODS Tibialis anterior and extensor digitorum longus muscles from wild-type, control, or muscle-specific GLUT4 knockout (mG4KO) mice were injected with the myotoxin barium chloride to induce muscle injury. After 3, 5, 7, 10, 14, or 21 days (in wild-type mice), or after 7 or 14 days (in control & mG4KO) mice, muscles were isolated to examine [3H]-2-deoxyglucose uptake, GLUT4 levels, extracellular fluid space, fibrosis, myofiber cross-sectional area, and myofiber centralized nuclei. RESULTS In wild-type mice, muscle glucose uptake was increased 3, 5, 7, and 10 days post-injury. There was a rapid decrease in GLUT4 protein levels that were restored to baseline at 5-7 days post-injury, followed by a super-compensation at 10-21 days. In mG4KO mice, there were no differences in muscle glucose uptake, extracellular fluid space, muscle fibrosis, myofiber cross-sectional areas, or percentage of centrally nucleated myofibers at 7 days post-injury. In contrast, at 14 days injured muscles from mG4KO mice exhibited decreased glucose uptake, muscle weight, myofiber cross sectional areas, and centrally nucleated myofibers, with no change in extracellular fluid space or fibrosis. CONCLUSIONS Collectively, these findings demonstrate that glucose uptake via GLUT4 regulates skeletal myofiber regeneration following acute injury.
Collapse
Affiliation(s)
- Tyler J Sermersheim
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - LeAnna J Phillips
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - Parker L Evans
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes & Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Steven S Welc
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| | - Carol A Witczak
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Diabetes & Metabolic Diseases, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Li X, Cao Y, Liu Y, Fang W, Xiao C, Cao Y, Zhao Y. Effect of IGF1 on Myogenic Proliferation and Differentiation of Bovine Skeletal Muscle Satellite Cells Through PI3K/AKT Signaling Pathway. Genes (Basel) 2024; 15:1494. [PMID: 39766763 PMCID: PMC11675145 DOI: 10.3390/genes15121494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Cultivated meat, an alternative to conventional meat, has substantial potential for alleviating environmental and ethical concerns. This method of manufacturing meat involves the isolation of skeletal muscle satellite cells (SMSCs) from donor animals, after which they proliferate in vitro and differentiate into primitive muscle fibers. The aim of this research was to evaluate how the insulin-like growth factor 1 (IGF1) gene regulates the myogenic differentiation of bovine skeletal muscle satellite cells (bSMSCs). Methods: bSMSCs isolated from newborn calves were cultured to the third generation in vitro and differentiated into myoblasts via the serum withdrawal method. An overexpression lentivirus and siRNA targeting the IGF1 gene were constructed and transduced into bSMSCs, which were subsequently analyzed via real-time fluorescence quantitative PCR(qRT-PCR) and Western blots. The mRNA and protein levels of the myogenic differentiation markers myosin heavy chain (MyHC) and myogenin (MyoG) were determined. Results: The results revealed that the lentivirus overexpressing the IGF1 gene significantly increased the expression of MyHC and MyoG, whereas the expression of both the MyHC and MyoG mRNAs and proteins was strongly reduced by si-IGF1. Conclusions: IGF1 positively regulates the myogenic differentiation of bSMSCs. This study provides a reference for further elucidating the molecular mechanism by which the IGF1 gene regulates the myogenic differentiation of bSMSCs via the PI3K/Akt signaling pathway and lays a foundation for establishing a regulatory network of bovine muscle growth and development.
Collapse
Affiliation(s)
- Xin Li
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yang Cao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yu Liu
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Wenwen Fang
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Cheng Xiao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| | - Yang Cao
- Institute of Animal Biotechnology, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China
| | - Yumin Zhao
- Institute of Animal Husbandry and Veterinary Medicine, Ji Lin Academy of Agricultural Sciences, Gongzhuling 136100, China; (X.L.); (Y.C.); (Y.L.); (W.F.); (C.X.)
| |
Collapse
|
3
|
Burke BI, Ismaeel A, McCarthy JJ. The utility of the rodent synergist ablation model in identifying molecular and cellular mechanisms of skeletal muscle hypertrophy. Am J Physiol Cell Physiol 2024; 327:C601-C606. [PMID: 39069822 PMCID: PMC11427019 DOI: 10.1152/ajpcell.00362.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024]
Abstract
Skeletal muscle exhibits remarkable plasticity to adapt to stimuli such as mechanical loading. The mechanisms that regulate skeletal muscle hypertrophy due to mechanical overload have been thoroughly studied. Remarkably, our understanding of many of the molecular and cellular mechanisms that regulate hypertrophic growth were first identified using the rodent synergist ablation (SA) model and subsequently corroborated in human resistance exercise training studies. To demonstrate the utility of the SA model, we briefly summarize the hypertrophic mechanisms identified using the model and the following translation of these mechanism to human skeletal muscle hypertrophy induced by resistance exercise training.
Collapse
Affiliation(s)
- Benjamin I Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
4
|
Burke BI, Ismaeel A, von Walden F, Murach KA, McCarthy JJ. Skeletal muscle hypertrophy: cell growth is cell growth. Am J Physiol Cell Physiol 2024; 327:C614-C618. [PMID: 39069829 PMCID: PMC11901336 DOI: 10.1152/ajpcell.00418.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Roberts et al. have provided an insightful counterpoint to our review article on the utility of the synergist ablation model. The purpose of this review is to provide some further dialogue regarding the strengths and weaknesses of the synergist ablation model. Specifically, we highlight that the robustness of the model overshadows surgical limitations. We also compare the transcriptomic responses to synergist ablation in mice and resistance exercise in humans to identify common pathways. We conclude that "cell growth is cell growth" and that the mechanisms available to cells to accumulate biomass and increase in size are similar across cell types and independent of the rate of growth.
Collapse
Affiliation(s)
- Benjamin I Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - Ferdinand von Walden
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Kevin A Murach
- Department Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
5
|
Ismaeel A, Peck BD, Montgomery MM, Burke BI, Goh J, Kang G, Franco AB, Xia Q, Goljanek-Whysall K, McDonagh B, McLendon JM, Koopmans PJ, Jacko D, Schaaf K, Bloch W, Gehlert S, Wen Y, Murach KA, Peterson CA, Boudreau RL, Fisher-Wellman KH, McCarthy JJ. microRNA-1 Regulates Metabolic Flexibility in Skeletal Muscle via Pyruvate Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607377. [PMID: 39149347 PMCID: PMC11326265 DOI: 10.1101/2024.08.09.607377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
MicroRNA-1 (miR-1) is the most abundant miRNA in adult skeletal muscle. To determine the function of miR-1 in adult skeletal muscle, we generated an inducible, skeletal muscle-specific miR-1 knockout (KO) mouse. Integration of RNA-sequencing (RNA-seq) data from miR-1 KO muscle with Argonaute 2 enhanced crosslinking and immunoprecipitation sequencing (AGO2 eCLIP-seq) from human skeletal muscle identified miR-1 target genes involved with glycolysis and pyruvate metabolism. The loss of miR-1 in skeletal muscle induced cancer-like metabolic reprogramming, as shown by higher pyruvate kinase muscle isozyme M2 (PKM2) protein levels, which promoted glycolysis. Comprehensive bioenergetic and metabolic phenotyping combined with skeletal muscle proteomics and metabolomics further demonstrated that miR-1 KO induced metabolic inflexibility as a result of pyruvate oxidation resistance. While the genetic loss of miR-1 reduced endurance exercise performance in mice and in C. elegans, the physiological down-regulation of miR-1 expression in response to a hypertrophic stimulus in both humans and mice causes a similar metabolic reprogramming that supports muscle cell growth. Taken together, these data identify a novel post-translational mechanism of adult skeletal muscle metabolism regulation mediated by miR-1.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Bailey D Peck
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - McLane M Montgomery
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Benjamin I Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Jensen Goh
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Gyumin Kang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Abigail B Franco
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Mass Spectrometry and Proteomics Core, University of Kentucky, Lexington, KY, USA
| | - Qin Xia
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Katarzyna Goljanek-Whysall
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Jared M McLendon
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Pieter J Koopmans
- Department Health, Human Performance, & Recreation, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA
| | - Daniel Jacko
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Olympic Base Center, North Rhine-Westphalia/Rhineland, Cologne, Germany
| | - Kirill Schaaf
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Olympic Base Center, North Rhine-Westphalia/Rhineland, Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Department for the Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department Health, Human Performance, & Recreation, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA
| | - Charlotte A Peterson
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Ryan L Boudreau
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kelsey H Fisher-Wellman
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
6
|
Baumert P, Mäntyselkä S, Schönfelder M, Heiber M, Jacobs MJ, Swaminathan A, Minderis P, Dirmontas M, Kleigrewe K, Meng C, Gigl M, Ahmetov II, Venckunas T, Degens H, Ratkevicius A, Hulmi JJ, Wackerhage H. Skeletal muscle hypertrophy rewires glucose metabolism: An experimental investigation and systematic review. J Cachexia Sarcopenia Muscle 2024; 15:989-1002. [PMID: 38742477 PMCID: PMC11154753 DOI: 10.1002/jcsm.13468] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/06/2024] [Accepted: 03/15/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Proliferating cancer cells shift their metabolism towards glycolysis, even in the presence of oxygen, to especially generate glycolytic intermediates as substrates for anabolic reactions. We hypothesize that a similar metabolic remodelling occurs during skeletal muscle hypertrophy. METHODS We used mass spectrometry in hypertrophying C2C12 myotubes in vitro and plantaris mouse muscle in vivo and assessed metabolomic changes and the incorporation of the [U-13C6]glucose tracer. We performed enzyme inhibition of the key serine synthesis pathway enzyme phosphoglycerate dehydrogenase (Phgdh) for further mechanistic analysis and conducted a systematic review to align any changes in metabolomics during muscle growth with published findings. Finally, the UK Biobank was used to link the findings to population level. RESULTS The metabolomics analysis in myotubes revealed insulin-like growth factor-1 (IGF-1)-induced altered metabolite concentrations in anabolic pathways such as pentose phosphate (ribose-5-phosphate/ribulose-5-phosphate: +40%; P = 0.01) and serine synthesis pathway (serine: -36.8%; P = 0.009). Like the hypertrophy stimulation with IGF-1 in myotubes in vitro, the concentration of the dipeptide l-carnosine was decreased by 26.6% (P = 0.001) during skeletal muscle growth in vivo. However, phosphorylated sugar (glucose-6-phosphate, fructose-6-phosphate or glucose-1-phosphate) decreased by 32.2% (P = 0.004) in the overloaded muscle in vivo while increasing in the IGF-1-stimulated myotubes in vitro. The systematic review revealed that 10 metabolites linked to muscle hypertrophy were directly associated with glycolysis and its interconnected anabolic pathways. We demonstrated that labelled carbon from [U-13C6]glucose is increasingly incorporated by ~13% (P = 0.001) into the non-essential amino acids in hypertrophying myotubes, which is accompanied by an increased depletion of media serine (P = 0.006). The inhibition of Phgdh suppressed muscle protein synthesis in growing myotubes by 58.1% (P < 0.001), highlighting the importance of the serine synthesis pathway for maintaining muscle size. Utilizing data from the UK Biobank (n = 450 243), we then discerned genetic variations linked to the serine synthesis pathway (PHGDH and PSPH) and to its downstream enzyme (SHMT1), revealing their association with appendicular lean mass in humans (P < 5.0e-8). CONCLUSIONS Understanding the mechanisms that regulate skeletal muscle mass will help in developing effective treatments for muscle weakness. Our results provide evidence for the metabolic rewiring of glycolytic intermediates into anabolic pathways during muscle growth, such as in serine synthesis.
Collapse
Affiliation(s)
- Philipp Baumert
- School of Medicine and HealthTechnical University of MunichMunichGermany
- Research Institute for Sport and Exercise SciencesLiverpool John Moores UniversityLiverpoolUK
- Research Unit for Orthopaedic Sports Medicine and Injury Prevention (OSMI)UMIT TIROL ‐ Private University for Health Sciences and Health TechnologyInnsbruckAustria
| | - Sakari Mäntyselkä
- Faculty of Sport and Health Sciences, NeuroMuscular Research CenterUniversity of JyväskyläJyväskyläFinland
| | - Martin Schönfelder
- School of Medicine and HealthTechnical University of MunichMunichGermany
| | - Marie Heiber
- School of Medicine and HealthTechnical University of MunichMunichGermany
- Institute of Sport ScienceUniversity of the Bundeswehr MunichNeubibergGermany
| | - Mika Jos Jacobs
- School of Medicine and HealthTechnical University of MunichMunichGermany
| | - Anandini Swaminathan
- Institute of Sport Science and InnovationsLithuanian Sports UniversityKaunasLithuania
| | - Petras Minderis
- Institute of Sport Science and InnovationsLithuanian Sports UniversityKaunasLithuania
| | - Mantas Dirmontas
- Department of Health Promotion and RehabilitationLithuanian Sports UniversityKaunasLithuania
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass SpectrometryTechnical University of MunichMunichGermany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass SpectrometryTechnical University of MunichMunichGermany
| | - Michael Gigl
- Bavarian Center for Biomolecular Mass SpectrometryTechnical University of MunichMunichGermany
| | - Ildus I. Ahmetov
- Research Institute for Sport and Exercise SciencesLiverpool John Moores UniversityLiverpoolUK
| | - Tomas Venckunas
- Institute of Sport Science and InnovationsLithuanian Sports UniversityKaunasLithuania
| | - Hans Degens
- Institute of Sport Science and InnovationsLithuanian Sports UniversityKaunasLithuania
- Department of Life SciencesManchester Metropolitan UniversityManchesterUK
| | - Aivaras Ratkevicius
- Institute of Sport Science and InnovationsLithuanian Sports UniversityKaunasLithuania
- Sports and Exercise Medicine CentreQueen Mary University of LondonLondonUK
| | - Juha J. Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research CenterUniversity of JyväskyläJyväskyläFinland
| | - Henning Wackerhage
- School of Medicine and HealthTechnical University of MunichMunichGermany
| |
Collapse
|
7
|
Meinhold M, Verbrugge S, Shi A, Schönfelder M, Becker L, Jaspers RT, Zammit PS, Wackerhage H. Yap/Taz activity is associated with increased expression of phosphoglycerate dehydrogenase that supports myoblast proliferation. Cell Tissue Res 2024; 395:271-283. [PMID: 38183459 PMCID: PMC10904560 DOI: 10.1007/s00441-023-03851-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/24/2023] [Indexed: 01/08/2024]
Abstract
In skeletal muscle, the Hippo effector Yap promotes satellite cell, myoblast, and rhabdomyoblast proliferation but prevents myogenic differentiation into multinucleated muscle fibres. We previously noted that Yap drives expression of the first enzyme of the serine biosynthesis pathway, phosphoglycerate dehydrogenase (Phgdh). Here, we examined the regulation and function of Phgdh in satellite cells and myoblasts and found that Phgdh protein increased during satellite cell activation. Analysis of published data reveal that Phgdh mRNA in mouse tibialis anterior muscle was highly expressed at day 3 of regeneration after cardiotoxin injection, when markers of proliferation are also robustly expressed and in the first week of synergist-ablated muscle. Finally, siRNA-mediated knockdown of PHGDH significantly reduced myoblast numbers and the proliferation rate. Collectively, our data suggest that Phgdh is a proliferation-enhancing metabolic enzyme that is induced when quiescent satellite cells become activated.
Collapse
Affiliation(s)
- Marius Meinhold
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany.
| | - Sander Verbrugge
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| | - Andi Shi
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Martin Schönfelder
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| | - Lore Becker
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, German Mouse Clinic, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Henning Wackerhage
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| |
Collapse
|
8
|
Mäntyselkä S, Kolari K, Baumert P, Ylä-Outinen L, Kuikka L, Lahtonen S, Permi P, Wackerhage H, Kalenius E, Kivelä R, Hulmi JJ. Serine synthesis pathway enzyme PHGDH is critical for muscle cell biomass, anabolic metabolism, and mTORC1 signaling. Am J Physiol Endocrinol Metab 2024; 326:E73-E91. [PMID: 37991454 DOI: 10.1152/ajpendo.00151.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Cells use glycolytic intermediates for anabolism, e.g., via the serine synthesis and pentose phosphate pathways. However, we still understand poorly how these metabolic pathways contribute to skeletal muscle cell biomass generation. The first aim of this study was therefore to identify enzymes that limit protein synthesis, myotube size, and proliferation in skeletal muscle cells. We inhibited key enzymes of glycolysis, the pentose phosphate pathway, and the serine synthesis pathway to evaluate their importance in C2C12 myotube protein synthesis. Based on the results of this first screen, we then focused on the serine synthesis pathway enzyme phosphoglycerate dehydrogenase (PHGDH). We used two different PHGDH inhibitors and mouse C2C12 and human primary muscle cells to study the importance and function of PHGDH. Both myoblasts and myotubes incorporated glucose-derived carbon into proteins, RNA, and lipids, and we showed that PHGDH is essential in these processes. PHGDH inhibition decreased protein synthesis, myotube size, and myoblast proliferation without cytotoxic effects. The decreased protein synthesis in response to PHGDH inhibition appears to occur mainly mechanistic target of rapamycin complex 1 (mTORC1)-dependently, as was evident from experiments with insulin-like growth factor 1 and rapamycin. Further metabolomics analyses revealed that PHGDH inhibition accelerated glycolysis and altered amino acid, nucleotide, and lipid metabolism. Finally, we found that supplementing an antioxidant and redox modulator, N-acetylcysteine, partially rescued the decreased protein synthesis and mTORC1 signaling during PHGDH inhibition. The data suggest that PHGDH activity is critical for skeletal muscle cell biomass generation from glucose and that it regulates protein synthesis and mTORC1 signaling.NEW & NOTEWORTHY The use of glycolytic intermediates for anabolism was demonstrated in both myoblasts and myotubes, which incorporate glucose-derived carbon into proteins, RNA, and lipids. We identify phosphoglycerate dehydrogenase (PHGDH) as a critical enzyme in those processes and also for muscle cell hypertrophy, proliferation, protein synthesis, and mTORC1 signaling. Our results thus suggest that PHGDH in skeletal muscle is more than just a serine-synthesizing enzyme.
Collapse
Affiliation(s)
- Sakari Mäntyselkä
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kalle Kolari
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Philipp Baumert
- Department for Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Laura Ylä-Outinen
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Lauri Kuikka
- Central Finland Health Care District Hospital District, Jyväskylä, Finland
| | - Suvi Lahtonen
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Perttu Permi
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Chemistry, University of Jyväskylä, Jyväskylä, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Henning Wackerhage
- Department for Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Elina Kalenius
- Department of Chemistry, University of Jyväskylä, Jyväskylä, Finland
| | - Riikka Kivelä
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Juha J Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
9
|
Ismaeel A, Thomas NT, McCashland M, Vechetti IJ, Edman S, Lanner JT, Figueiredo VC, Fry CS, McCarthy JJ, Wen Y, Murach KA, von Walden F. Coordinated Regulation of Myonuclear DNA Methylation, mRNA, and miRNA Levels Associates With the Metabolic Response to Rapid Synergist Ablation-Induced Skeletal Muscle Hypertrophy in Female Mice. FUNCTION 2023; 5:zqad062. [PMID: 38020067 PMCID: PMC10666992 DOI: 10.1093/function/zqad062] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/16/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
The central dogma of molecular biology dictates the general flow of molecular information from DNA that leads to a functional cellular outcome. In skeletal muscle fibers, the extent to which global myonuclear transcriptional alterations, accounting for epigenetic and post-transcriptional influences, contribute to an adaptive stress response is not clearly defined. In this investigation, we leveraged an integrated analysis of the myonucleus-specific DNA methylome and transcriptome, as well as myonuclear small RNA profiling to molecularly define the early phase of skeletal muscle fiber hypertrophy. The analysis of myonucleus-specific mature microRNA and other small RNA species provides new directions for exploring muscle adaptation and complemented the methylation and transcriptional information. Our integrated multi-omics interrogation revealed a coordinated myonuclear molecular landscape during muscle loading that coincides with an acute and rapid reduction of oxidative metabolism. This response may favor a biosynthesis-oriented metabolic program that supports rapid hypertrophic growth.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Mariah McCashland
- Department of Nutrition and Health Sciences, University of Nebraska–Lincoln, Lincoln, NE 68583, USA
| | - Ivan J Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska–Lincoln, Lincoln, NE 68583, USA
| | - Sebastian Edman
- Department of Women’s and Children’s Health, Karolinska Institutet, Solna 17177, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna 17177, Sweden
| | - Vandré C Figueiredo
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, AR 72701, USA
| | - Ferdinand von Walden
- Department of Women’s and Children’s Health, Karolinska Institutet, Solna 17177, Sweden
| |
Collapse
|