1
|
Horwath O, Cornet L, Strömlind H, Moberg M, Edman S, Söderlund K, Checa A, Ruas JL, Blomstrand E. Endurance exercise with reduced muscle glycogen content influences substrate utilization and attenuates acute mTORC1- and autophagic signaling in human type I and type II muscle fibers. Skelet Muscle 2025; 15:9. [PMID: 40128889 PMCID: PMC11934587 DOI: 10.1186/s13395-025-00377-3] [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: 10/14/2024] [Accepted: 02/28/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Exercising with low muscle glycogen content can improve training adaptation, but the mechanisms underlying the muscular adaptation are still largely unknown. In this study, we measured substrate utilization and cell signaling in different muscle fiber types during exercise and investigated a possible link between these variables. METHODS Five subjects performed a single leg cycling exercise in the evening (day 1) with the purpose of reducing glycogen stores. The following morning (day 2), they performed two-legged cycling at ∼70% of VO2peak for 1 h. Muscle biopsies were taken from both legs pre- and post-exercise for enzymatic analyses of glycogen, metabolite concentrations using LC-MS/MS-based quantification, and protein signaling using Western blot in pools of type I or type II fibers. RESULTS Glycogen content was 60-65% lower for both fiber types (P < 0.01) in the leg that exercised on day 1 (low leg) compared to the other leg with normal level of glycogen (normal leg) before the cycling exercise on day 2. Glycogen utilization during exercise was significantly less in both fiber types in the low compared to the normal leg (P < 0.05). In the low leg, there was a 14- and 6-fold increase in long-chain fatty acids conjugated to carnitine in type I and type II fibers, respectively, post-exercise. This increase was 3-4 times larger than in the normal leg (P < 0.05). Post-exercise, mTORSer2448 phosphorylation was increased in both fiber types in the normal leg (P < 0.05) but remained unchanged in both fiber types in the low leg together with an increase in eEF2Thr56 phosphorylation in type I fibers (P < 0.01). Exercise induced a reduction in the autophagy marker LC3B-II in both fiber types and legs, but the post-exercise level was higher in both fiber types in the low leg (P < 0.05). Accordingly, the LC3B-II/I ratio decreased only in the normal leg (75% for type I and 87% for type II, P < 0.01). CONCLUSIONS Starting an endurance exercise session with low glycogen availability leads to profound changes in substrate utilization in both type I and type II fibers. This may reduce the mTORC1 signaling response, primarily in type I muscle fibers, and attenuate the normally observed reduction in autophagy.
Collapse
Affiliation(s)
- Oscar Horwath
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Lucas Cornet
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Henrik Strömlind
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Marcus Moberg
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Sebastian Edman
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Karin Söderlund
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
- Department of Pharmacology and Stanley & Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eva Blomstrand
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Holmes TL, Chabronova A, Denning C, James V, Peffers MJ, Smith JGW. Footprints in the Sno: investigating the cellular and molecular mechanisms of SNORD116. Open Biol 2025; 15:240371. [PMID: 40101781 PMCID: PMC11919532 DOI: 10.1098/rsob.240371] [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: 11/29/2024] [Revised: 01/11/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
The small nucleolar RNA (snoRNA) SNORD116 is a small non-coding RNA of interest across multiple biomedical fields of research. Much of the investigation into SNORD116 has been undertaken in the context of the congenital disease Prader-Willi syndrome, wherein SNORD116 expression is lost. However, emerging evidence indicates wider roles in various disease and tissue contexts such as cellular growth, metabolism and signalling. Nevertheless, a conclusive mechanism of action for SNORD116 remains to be established. Here, we review the key findings from these investigations, with the aim of identifying common elements from which to elucidate potential targets and mechanisms of SNORD116. A key recurring element identified is disruption to the insulin/IGF-1 and PI3K/mTOR signalling pathways, contributing to many of the phenotypes associated with SNORD116 modulation explored in this review.
Collapse
Affiliation(s)
- Terri L Holmes
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, UK
| | - Alzbeta Chabronova
- Department of Musculoskeletal Ageing Science, University of Liverpool, Liverpool, UK
| | - Chris Denning
- Department of Stem Cell Biology, University of Nottingham, Nottingham, UK
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Ageing Science, University of Liverpool, Liverpool, UK
| | - James G W Smith
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich, Norfolk NR4 7UQ, UK
| |
Collapse
|
3
|
Von Ruff ZD, Miller MJ, Moro T, Reidy PT, Ebert SM, Volpi E, Adams CM, Rasmussen BB. Resistance exercise training in older men reduces ATF4-activated and senescence-associated mRNAs in skeletal muscle. GeroScience 2025:10.1007/s11357-025-01564-2. [PMID: 40011348 DOI: 10.1007/s11357-025-01564-2] [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] [Received: 10/22/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Sarcopenia increases the risk of frailty, morbidity, and mortality in older adults. Resistance exercise training improves muscle size and function; however, the response to exercise training is variable in older adults. The objective of our study was to determine both the age-independent and age-dependent changes to the transcriptome following progressive resistance exercise training. Skeletal muscle biopsies were obtained before and after 12 weeks of resistance exercise training in 8 young (24 ± 3.3 years) and 10 older (72 ± 4.9 years) men. RNA was extracted from each biopsy and prepared for analysis via RNA sequencing. We performed differential mRNA expression, gene ontology, and gene set enrichment analyses. We report that when comparing post-training vs pre-training 226 mRNAs and 959 mRNAs were differentially expressed in the skeletal muscle of young and older men, respectively. Additionally, 94 mRNAs increased, and 17 mRNAs decreased in both young and old, indicating limited overlap in response to resistance exercise training. Furthermore, the differential gene expression was larger in older skeletal muscle. Finally, we report three novel findings: 1) resistance exercise training decreased the abundance of ATF4-activated and senescence-associated skeletal muscle mRNAs in older men; 2) resistance exercise-induced increases in lean mass correlate with increased mRNAs encoding mitochondrial proteins; and 3) increases in muscle strength following resistance exercise positively correlate with increased mRNAs involved in translation, rRNA processing, and polyamine metabolism. We conclude that resistance exercise training elicits a differential gene expression response in young and old skeletal muscle, including reduced ATF-4 activated and senescence-associated gene expression.
Collapse
Affiliation(s)
| | - Matthew J Miller
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- University of Iowa, Iowa City, IA, USA
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Paul T Reidy
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, OH, USA
| | - Scott M Ebert
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Elena Volpi
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Blake B Rasmussen
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7756, San Antonio, TX, 78229, USA.
| |
Collapse
|
4
|
Murach KA, Bagley JR. A primer on global molecular responses to exercise in skeletal muscle: Omics in focus. JOURNAL OF SPORT AND HEALTH SCIENCE 2025:101029. [PMID: 39961420 DOI: 10.1016/j.jshs.2025.101029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/19/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Advances in skeletal muscle omics has expanded our understanding of exercise-induced adaptations at the molecular level. Over the past 2 decades, transcriptome studies in muscle have detailed acute and chronic responses to resistance, endurance, and concurrent exercise, focusing on variables such as training status, nutrition, age, sex, and metabolic health profile. Multi-omics approaches, such as the integration of transcriptomic and epigenetic data, along with emerging ribosomal RNA sequencing advancements, have further provided insights into how skeletal muscle adapts to exercise across the lifespan. Downstream of the transcriptome, proteomic and phosphoproteomic studies have identified novel regulators of exercise adaptations, while single-cell/nucleus and spatial sequencing technologies promise to evolve our understanding of cellular specialization and communication in and around skeletal muscle cells. This narrative review highlights (a) the historical foundations of exercise omics in skeletal muscle, (b) current research at 3 layers of the omics cascade (DNA, RNA, and protein), and (c) applications of single-cell omics and spatial sequencing technologies to study skeletal muscle adaptation to exercise. Further elaboration of muscle's global molecular footprint using multi-omics methods will help researchers and practitioners develop more effective and targeted approaches to improve skeletal muscle health as well as athletic performance.
Collapse
Affiliation(s)
- Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR 72701, USA.
| | - James R Bagley
- Muscle Physiology Laboratory, Department of Kinesiology, College of Health and Social Sciences, San Francisco State University, San Francisco, CA 94132, USA.
| |
Collapse
|
5
|
Dong H, Lyu Y, Huang CY, Tsai SY. Limiting cap-dependent translation increases 20S proteasomal degradation and protects the proteomic integrity in autophagy-deficient skeletal muscle. Autophagy 2025:1-16. [PMID: 39878121 DOI: 10.1080/15548627.2025.2457925] [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: 09/04/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
Postmitotic skeletal muscle critically depends on tightly regulated protein degradation to maintain proteomic stability. Impaired macroautophagy/autophagy-lysosomal or ubiquitin-proteasomal protein degradation causes the accumulation of damaged proteins, ultimately accelerating muscle dysfunction with age. While in vitro studies have demonstrated the complementary nature of these systems, their interplay at the organism levels remains poorly understood. Here, our study reveals novel insights into this complex relationship in autophagy-deficient skeletal muscle. We demonstrated that despite a compensatory increase in proteasome level in response to autophagy impairment, 26S proteasome activity was not proportionally enhanced in autophagy-deficient skeletal muscle. This functional deficit was partly attributed to reduced ATP levels to fuel the 26S proteasome. Remarkably, we found that activation of EIF4EBP1, a crucial inhibitor of cap-dependent translation, restored and even augmented proteasomal function through dual mechanisms. First, genetically activating EIF4EBP1 enhanced both ATP-dependent 26S proteasome and ATP-independent 20S proteasome activities, thereby expanding overall protein degradation capacity. Second, EIF4EBP1 activation caused muscle fiber transformation and increased mitochondrial biogenesis, thus replenishing ATP levels for 26S proteasome activation. Notably, the improved performance of the 20S proteasome in EIF4EBP1-activated skeletal muscle was attributed to an increased abundance of the immunoproteasome, a subtype specially adapted to function under oxidative stress conditions. This dual action of EIF4EBP1 activation preserved proteomic integrity in autophagy-deficient skeletal muscle. Our findings uncover a novel role of EIF4EBP1 in improving protein quality control, presenting a promising therapeutic strategy for autophagy-related muscular disorders and potentially other conditions characterized by proteostatic imbalance.Abbreviations: 3-MA: 3-methyladenine; ACAC/ACC: acetyl-Coenzyme A carboxylase; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATP: adenosine triphosphate; ATP5F1A/ATP5A: ATP synthase F1 subunit alpha; CKM-Cre: creatine kinase, muscle-Cre; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSK: cathepsin K; CTSL: cathepsin L; CUL3: cullin 3; EDL: extensor digitorum longus; EIF4E: eukaryotic translation initiation factor 4E; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; EIF4F: eukaryotic translation initiation factor 4F complex; FBXO32/ATROGIN1/MAFbx: F-box protein 32; GFP: green fluorescent protein; IFNG/IFN-γ: interferon gamma; KEAP1: kelch-like ECH-associated protein 1; LAMP1: lysosomal-associated membrane protein 1; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; Myl1/Mlc1f-Cre: myosin, light polypeptide 1 (promoter driving Cre recombinase); mRFP: monomeric red fluorescent protein; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NFE2L1/NRF1: nuclear factor, erythroid derived 2, like 1; NFE2L2/NRF2: nuclear factor, erythroid derived 2, like 2; NFKB1/NFκB1: nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105; OXPHOS: oxidative phosphorylation; PPARGC1A/PGC1α: peroxisome proliferator activated receptor, gamma, coactivator 1 alpha; PSMB5: proteasome (prosome, macropain) subunit, beta type 5; PSMB6: proteasome (prosome, macropain) subunit, beta type 6; PSMB7: proteasome (prosome, macropain) subunit, beta type 7; PSMB8: proteasome (prosome, macropain) subunit, beta type 8 (large multifunctional peptidase 7); PSMB9: proteasome (prosome, macropain) subunit, beta type 9 (large multifunctional peptidase 2); PSMB10: proteasome (prosome, macropain) subunit, beta type 10; PSME1: proteasome (prosome, macropain) activator subunit 1 (PA28 alpha); PSME2: proteasome (prosome, macropain) activator subunit 2 (PA28 beta); RBX1: ring-box 1; SQSTM1/p62: sequestosome 1; SREBF1/SREBP1: sterol regulatory element binding transcription factor 1; STAT3: signal transducer and activator of transcription 3; TRIM63/MURF1: tripartite motif-containing 63; ULK1: unc-51 like kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Han Dong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yifan Lyu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chien-Yung Huang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shih-Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
6
|
Horwath O, Moberg M, Hodson N, Edman S, Johansson M, Andersson E, van Hall G, Rooyackers O, Philp A, Apró W. Anabolic Sensitivity in Healthy, Lean, Older Men Is Associated With Higher Expression of Amino Acid Sensors and mTORC1 Activators Compared to Young. J Cachexia Sarcopenia Muscle 2025; 16:e13613. [PMID: 39558870 DOI: 10.1002/jcsm.13613] [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: 06/20/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Sarcopenia is thought to be underlined by age-associated anabolic resistance and dysregulation of intracellular signalling pathways. However, it is unclear whether these phenomena are driven by ageing per se or other confounding factors. METHODS Lean and healthy young (n = 10, 22 ± 3 years, BMI; 23.4 ± 0.8 kg/m2) and old men (n = 10, 70 ± 3 years, BMI; 22.7 ± 1.3 kg/m2) performed unilateral resistance exercise followed by intake of essential amino acids (EAA). Muscle biopsies were collected from the rested and the exercised leg before, immediately after and 60 and 180 min after EAA intake. Muscle samples were analysed for amino acid concentrations, muscle protein synthesis (MPS) and associated anabolic signalling. RESULTS Following exercise, peak plasma levels of EAA and leucine were similar between groups, but the area under the curve was ~11% and ~28% lower in Young (p < 0.01). Absolute levels of muscle EAA and leucine peaked 60 min after exercise, with ~15 and ~21% higher concentrations in the exercising leg (p < 0.01) but with no difference between groups. MPS increased in both the resting (~0.035%·h-1 to 0.056%·h-1, p < 0.05) and exercising leg (~0.035%·h-1 to 0.083%·h-1, p < 0.05) with no difference between groups. Phosphorylation of S6K1Thr389 increased to a similar extent in the exercising leg in both groups but was 2.8-fold higher in the resting leg of Old at the 60 min timepoint (p < 0.001). Phosphorylation of 4E-BP1Ser65 increased following EAA intake and exercise, but differences between legs were statistically different only at 180 min (p < 0.001). However, phosphorylation of this site was on average 78% greater across all timepoints in Old (p < 0.01). Phosphorylation of eEF2Thr56 was reduced (~66% and 39%) in the exercising leg at both timepoints after EAA intake and exercise, with no group differences (p < 0.05). However, phosphorylation at this site was reduced by ~27% also in the resting leg at 60 min, an effect that was only seen in Old (p < 0.01). Total levels of Rheb (~45%), LAT1 (~31%) and Rag B (~31%) were higher in Old (p < 0.001). CONCLUSION Lean and healthy old men do not manifest AR as evidenced by potent increases in MPS and mTORC1 signalling following EAA intake and exercise. Maintained anabolic sensitivity with age appears to be a function of a compensatory increase in basal levels of proteins involved in anabolic signalling. Therefore, our results suggest that age per se does not appear to cause AR in human skeletal muscle.
Collapse
Affiliation(s)
- Oscar Horwath
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Marcus Moberg
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Nathan Hodson
- Department of Exercise Sciences, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
- Department of Sport and Exercise Sciences, Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Sebastian Edman
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Mats Johansson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Eva Andersson
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Gerrit van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Olav Rooyackers
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Andrew Philp
- Centre for Healthy Ageing, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - William Apró
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Crişan D, Avram L, Morariu-Barb A, Grapa C, Hirişcau I, Crăciun R, Donca V, Nemeş A. Sarcopenia in MASLD-Eat to Beat Steatosis, Move to Prove Strength. Nutrients 2025; 17:178. [PMID: 39796612 PMCID: PMC11722590 DOI: 10.3390/nu17010178] [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: 11/23/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The connections between sarcopenia and various chronic conditions, including type 2 diabetes (T2DM), metabolic syndrome (MetS), and liver disease have been highlighted recently. There is also a high occurrence of sarcopenia in metabolic dysfunction-associated steatotic liver disease (MASLD) patients, who are often disregarded. Both experimental and clinical findings suggest a complex, bidirectional relationship between MASLD and sarcopenia. While vitamin D, testosterone, and specific drug therapies show promise in mitigating sarcopenia, consensus on effective treatments is lacking. Recent focus on lifestyle interventions emphasizes dietary therapy and exercise for sarcopenic obesity in MASLD. Challenges arise as weight loss, a primary MASLD treatment, may lead to muscle mass reduction. The therapeutic approach to sarcopenia in morbidly obese MASLD patients also includes bariatric surgery (BS). BS induces weight loss and stabilizes metabolic imbalances, but its impact on sarcopenia is nuanced, underscoring the need for further research. Our aim is to provide a comprehensive review of the interplay between sarcopenia and MASLD and offer insight into the most recent therapeutic challenges and discoveries, as sarcopenia is often overlooked or unrecognized and poses significant challenges for managing these patients.
Collapse
Affiliation(s)
- Dana Crişan
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Clinical Municipal Hospital, 400139 Cluj-Napoca, Romania
| | - Lucreţia Avram
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Clinical Municipal Hospital, 400139 Cluj-Napoca, Romania
| | - Andreea Morariu-Barb
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400162 Cluj-Napoca, Romania
| | - Cristiana Grapa
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400162 Cluj-Napoca, Romania
| | - Ioana Hirişcau
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
| | - Rareş Crăciun
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400162 Cluj-Napoca, Romania
| | - Valer Donca
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Clinical Municipal Hospital, 400139 Cluj-Napoca, Romania
| | - Andrada Nemeş
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.C.); (L.A.); (I.H.); (R.C.); (V.D.); (A.N.)
- Clinical Municipal Hospital, 400139 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Ye M, Chao X, Ye C, Guo L, Fan Z, Ma X, Liu A, Liang W, Chen S, Fang C, Zhang X, Luo Q. EGR1 mRNA expression levels and polymorphisms are associated with slaughter performance in chickens. Poult Sci 2025; 104:104533. [PMID: 39603185 PMCID: PMC11635649 DOI: 10.1016/j.psj.2024.104533] [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: 06/24/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
With the implementation of the policy of "centralized slaughtering and chilled to market" and the development of the livestock processing industry, numerous researchers have begun to focus on the selection and breeding of broilers bred for slaughter. The selection of breeds with excellent slaughtering performance and high meat production performance has become one of the most important selective breeding goals. In our previous study, we conducted transcriptome sequencing on chicken breast tissues with high and low breast muscle rates and found higher early growth response protein 1 (EGR1) expression in breast tissues with a low breast muscle ratio, thus hypothesizing that the EGR1 gene is involved in the growth and development process of chicken muscle tissues. Therefore, we analyzed the gene functions and polymorphisms of EGR1 to investigate its association with slaughter traits. We used various experimental methods, including RT-qPCR, Cell Counting Kit 8, 5-ethynyl-2'-deoxyuridine, western blot, flow cytometry, and immunofluorescence, to validate EGR1's role in chicken primary myoblasts. The results of our functional validation experiments indicate that EGR1 is highly expressed in breast tissues with a low breast muscle content and plays a key role in regulating of muscle growth and development by promoting proliferation and inhibiting the differentiation of chicken primary myoblasts. In addition, we explored the relationship between the EGR1 gene polymorphisms and slaughter traits using mixed linear models for the first time. In a population of Jiangfeng M3 lineage partridge chickens, we identified 4 EGR1 single-nucleotide polymorphisms, 2 of which were significantly associated with slaughter traits, including live weight, slaughter weight, semi-eviscerated weight, eviscerated weight, leg weight, wing weight, and breast muscle rate. In summary, ectopic expression of EGR1 promotes the proliferation and differentiation of chicken primary myoblasts. In addition, polymorphisms in EGR1 were associated with slaughter performance, providing a potential basis for further utilization of EGR1 as a breeding marker.
Collapse
Affiliation(s)
- Mao Ye
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Xiaohuan Chao
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Chutian Ye
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Lijin Guo
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Zhexia Fan
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Xuerong Ma
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Aijun Liu
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Weiming Liang
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Shuya Chen
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Cheng Fang
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Xiquan Zhang
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China
| | - Qingbin Luo
- College of Animal Science, South China Agricultural University, Guangzhou, PR China; State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, PR China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, PR China.
| |
Collapse
|
9
|
Kusano T, Sotani Y, Takeda R, Hatano A, Kawata K, Kano R, Matsumoto M, Kano Y, Hoshino D. Time-series transcriptomics reveals distinctive mRNA expression dynamics associated with gene ontology specificity and protein expression in skeletal muscle after electrical stimulation-induced resistance exercise. FASEB J 2024; 38:e70153. [PMID: 39545720 PMCID: PMC11698011 DOI: 10.1096/fj.202401420rr] [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: 07/22/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Resistance exercise upregulates and downregulates the expression of a wide range of genes in skeletal muscle. However, detailed analysis of mRNA dynamics such as response rates and temporal patterns of the transcriptome after resistance exercise has not been performed. We aimed to clarify the dynamics of time-series transcriptomics after resistance exercise. We used electrical stimulation-induced muscle contraction as a resistance exercise model (5 sets × 10 times of 3 s of 100-Hz electrical stimulation) on the tibialis anterior muscle of rats and measured the transcriptome in the muscle before and at 0, 1, 3, 6, and 12 h after muscle contractions by RNA sequencing. We also examined the relationship between the parameters of mRNA dynamics and the increase in protein expression at 12 h after muscle contractions. We found that the function of the upregulated genes differed after muscle contractions depending on their response rate. Genes related to muscle differentiation and response to mechanical stimulus were enriched in the sustainedly upregulated genes. Furthermore, there was a positive correlation between the magnitude of upregulated mRNA expression and the corresponding protein expression level at 12 h after muscle contractions. Although it has been theoretically suggested, this study experimentally demonstrated that the magnitude of the mRNA response after electrical stimulation-induced resistance exercise contributes to skeletal muscle adaptation via increases in protein expression. These findings suggest that mRNA expression dynamics such as response rate, a sustained upregulated expression pattern, and the magnitude of the response contribute to mechanisms underlying adaptation to resistance exercise.
Collapse
Affiliation(s)
- Tatsuya Kusano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Yuta Sotani
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Reo Takeda
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Atsushi Hatano
- Department of Omics and Systems Biology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Kentaro Kawata
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaIbarakiJapan
| | - Ryotaro Kano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Yutaka Kano
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| | - Daisuke Hoshino
- Bioscience and Technology Program, Department of Engineering ScienceThe University of Electro‐CommunicationsChofuTokyoJapan
| |
Collapse
|
10
|
Zhou X, Zhu S, Li J, Mateus A, Williams C, Gilthorpe J, Backman LJ. Mechanical Loading Modulates AMPK and mTOR Signaling in Muscle Cells. J Proteome Res 2024; 23:4286-4295. [PMID: 39213513 PMCID: PMC11459513 DOI: 10.1021/acs.jproteome.4c00242] [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: 04/03/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Skeletal muscle adaptation to exercise involves various phenotypic changes that enhance the metabolic and contractile functions. One key regulator of these adaptive responses is the activation of AMPK, which is influenced by exercise intensity. However, the mechanistic understanding of AMPK activation during exercise remains incomplete. In this study, we utilized an in vitro model to investigate the effects of mechanical loading on AMPK activation and its interaction with the mTOR signaling pathway. Proteomic analysis of muscle cells subjected to static loading (SL) revealed distinct quantitative protein alterations associated with RNA metabolism, with 10% SL inducing the most pronounced response compared to lower intensities of 5% and 2% as well as the control. Additionally, 10% SL suppressed RNA and protein synthesis while activating AMPK and inhibiting the mTOR pathway. We also found that SRSF2, necessary for pre-mRNA splicing, is regulated by AMPK and mTOR signaling, which, in turn, is regulated in an intensity-dependent manner by SL with the highest expression in 2% SL. Further examination showed that the ADP/ATP ratio was increased after 10% SL compared to the control and that SL induced changes in mitochondrial biogenesis. Furthermore, Seahorse assay results indicate that 10% SL enhances mitochondrial respiration. These findings provide novel insights into the cellular responses to mechanical loading and shed light on the intricate AMPK-mTOR regulatory network in muscle cells.
Collapse
Affiliation(s)
- Xin Zhou
- Department
of Medical and Translational Biology, Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
| | - Shaochun Zhu
- Department
of Chemistry, Faculty of Medicine, Umeå
University, 90187 Umeå, Sweden
| | - Junhong Li
- Department
of Medical and Translational Biology, Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
- Section
of Physiotherapy, Department of Community Medicine and Rehabilitation,
Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
| | - Andre Mateus
- Department
of Chemistry, Faculty of Medicine, Umeå
University, 90187 Umeå, Sweden
| | - Chloe Williams
- Department
of Medical and Translational Biology, Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
| | - Jonathan Gilthorpe
- Department
of Medical and Translational Biology, Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
| | - Ludvig J. Backman
- Department
of Medical and Translational Biology, Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
- Section
of Physiotherapy, Department of Community Medicine and Rehabilitation,
Faculty of Medicine, Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
11
|
Zhao YC, Gao BH. Integrative effects of resistance training and endurance training on mitochondrial remodeling in skeletal muscle. Eur J Appl Physiol 2024; 124:2851-2865. [PMID: 38981937 DOI: 10.1007/s00421-024-05549-5] [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: 01/06/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Resistance training activates mammalian target of rapamycin (mTOR) pathway of hypertrophy for strength gain, while endurance training increases peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway of mitochondrial biogenesis benefiting oxidative phosphorylation. The conventional view suggests that resistance training-induced hypertrophy signaling interferes with endurance training-induced mitochondrial remodeling. However, this idea has been challenged because acute leg press and knee extension in humans enhance both muscle hypertrophy and mitochondrial remodeling signals. Thus, we first examined the muscle mitochondrial remodeling and hypertrophy signals with endurance training and resistance training, respectively. In addition, we discussed the influence of resistance training on muscle mitochondria, demonstrating that the PGC-1α-mediated muscle mitochondrial adaptation and hypertrophy occur simultaneously. The second aim was to discuss the integrative effects of concurrent training, which consists of endurance and resistance training sessions on mitochondrial remodeling. The study found that the resistance training component does not reduce muscle mitochondrial remodeling signals in concurrent training. On the contrary, concurrent training has the potential to amplify skeletal muscle mitochondrial biogenesis compared to a single exercise model. Concurrent training involving differential sequences of resistance and endurance training may result in varied mitochondrial biogenesis signals, which should be linked to the pre-activation of mTOR or PGC-1α signaling. Our review proposed a mechanism for mTOR signaling that promotes PGC-1α signaling through unidentified pathways. This mechanism may be account for the superior muscle mitochondrial remodeling change following the concurrent training. Our review suggested an interaction between resistance training and endurance training in skeletal muscle mitochondrial adaptation.
Collapse
Affiliation(s)
- Yong-Cai Zhao
- College of Exercise and Health, Tianjin University of Sport, No. 16 Donghai Road, Jinghai District, Tianjin, 301617, China.
| | - Bing-Hong Gao
- School of Athletic Performance, Shanghai University of Sport, No. 399 Changhai Road, Yangpu District, Shanghai, 200438, China
| |
Collapse
|
12
|
Thomas ACQ, Stead CA, Burniston JG, Phillips SM. Exercise-specific adaptations in human skeletal muscle: Molecular mechanisms of making muscles fit and mighty. Free Radic Biol Med 2024; 223:341-356. [PMID: 39147070 DOI: 10.1016/j.freeradbiomed.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The mechanisms leading to a predominantly hypertrophied phenotype versus a predominantly oxidative phenotype, the hallmarks of resistance training (RT) or aerobic training (AT), respectively, are being unraveled. In humans, exposure of naïve persons to either AT or RT results in their skeletal muscle exhibiting generic 'exercise stress-related' signaling, transcription, and translation responses. However, with increasing engagement in AT or RT, the responses become refined, and the phenotype typically associated with each form of exercise emerges. Here, we review some of the mechanisms underpinning the adaptations of how muscles become, through AT, 'fit' and RT, 'mighty.' Much of our understanding of molecular exercise physiology has arisen from targeted analysis of post-translational modifications and measures of protein synthesis. Phosphorylation of specific residue sites has been a dominant focus, with canonical signaling pathways (AMPK and mTOR) studied extensively in the context of AT and RT, respectively. These alone, along with protein synthesis, have only begun to elucidate key differences in AT and RT signaling. Still, key yet uncharacterized differences exist in signaling and regulation of protein synthesis that drive unique adaptation to AT and RT. Omic studies are required to better understand the divergent relationship between exercise and phenotypic outcomes of training.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada; Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stuart M Phillips
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
13
|
Ramos-Jiménez A, Hernández-Torres RP, Hernández-Ontiveros DA, Ortiz-Ortiz M, López-Fregoso RJ, Martínez-Sanz JM, Rodríguez-Uribe G, Hernández-Lepe MA. An Update of the Promise of Glycine Supplementation for Enhancing Physical Performance and Recovery. Sports (Basel) 2024; 12:265. [PMID: 39453231 PMCID: PMC11510825 DOI: 10.3390/sports12100265] [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] [Received: 07/09/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Glycine, the simple amino acid, is a key component of muscle metabolism with proven cytoprotective effects and hypothetical benefits as a therapeutic nutrient. Cell, in vitro, and animal studies suggest that glycine enhances protection against muscle wasting by activating anabolic pathways and inhibiting proteolytic gene expression. Some evidence indicates that glycine supplementation may enhance peak power output, reduce lactic acid accumulation during high-intensity exercise, and improve sleep quality and recovery. This literature review critically explores glycine's potential as an ergogenic aid and its relevance to muscle regeneration, muscle strength, endurance exercise performance, and sleep quality. It also underscores key areas for future research. It is concluded that more randomized controlled clinical trials in humans are needed to confirm glycine's potential as a dietary supplement to support muscle function, recovery, and overall athletic performance as an ergogenic aid and to establish nutritional recommendations for athletic performance. Also, it is essential to consider that high doses (>500 mg/kg of body mass) could induce cytotoxic effects and contribute to acute glutamate toxicity.
Collapse
Affiliation(s)
- Arnulfo Ramos-Jiménez
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Department of Health Sciences, Biomedical Sciences Institute, Autonomous University of Ciudad Juarez, Chihuahua 32310, Mexico
| | - Rosa Patricia Hernández-Torres
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Faculty of Physical Culture Sciences, Autonomous University of Chihuahua, Chihuahua 31000, Mexico
| | - David Alfredo Hernández-Ontiveros
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
| | - Melinna Ortiz-Ortiz
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
| | - Reymond Josué López-Fregoso
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
| | - José Miguel Martínez-Sanz
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Nursing Department, Faculty of Health Sciences, University of Alicante, San Vicente del Raspeig, 03690 Alicante, Spain
| | - Genaro Rodríguez-Uribe
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
- Academic Body “Salud Personalizada (UABC-CA-336)”, Autonomous University of Baja California, Tijuana 22390, Mexico
| | - Marco Antonio Hernández-Lepe
- Conahcyt National Laboratory of Body Composition and Energetic Metabolism (LaNCoCoME), Tijuana 22390, Mexico; (A.R.-J.); (R.P.H.-T.); (D.A.H.-O.); (M.O.-O.); (R.J.L.-F.); (J.M.M.-S.); (G.R.-U.)
- Medical and Psychology School, Autonomous University of Baja California, Tijuana 22390, Mexico
- Academic Body “Salud Personalizada (UABC-CA-336)”, Autonomous University of Baja California, Tijuana 22390, Mexico
| |
Collapse
|
14
|
Chen H, Xiong R, Cheng J, Ye J, Qiu Y, Huang S, Li M, Liu Z, Pang J, Zhang X, Guo S, Li H, Zhu H. Effects and Mechanisms of Polyunsaturated Fatty Acids on Age-Related Musculoskeletal Diseases: Sarcopenia, Osteoporosis, and Osteoarthritis-A Narrative Review. Nutrients 2024; 16:3130. [PMID: 39339730 PMCID: PMC11434726 DOI: 10.3390/nu16183130] [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: 08/22/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The process of the globally aging population has been accelerating, leading to an increasing social burden. As people age, the musculoskeletal system will gradually go through a series of degenerative and loss of function and eventually develop age-related musculoskeletal diseases, like sarcopenia, osteoporosis, and osteoarthritis. On the other hand, several studies have shown that polyunsaturated fatty acids (PUFAs) possess various important physiological functions on the health of muscles, bones, and joints. Objective: This narrative review paper provides a summary of the literature about the effects and mechanisms of PUFAs on age-related musculoskeletal diseases for the prevention and management of these diseases. Methods: Web of Science, PubMed, Science Direct, and Scopus databases have been searched to select the relevant literature on epidemiological, cellular, and animal experiments and clinical evidence in recent decades with keywords "polyunsaturated fatty acids", "PUFAs", "omega-3", "omega-6", "musculoskeletal diseases", "sarcopenia", "osteoporosis", "osteoarthritis", and so on. Results: PUFAs could prevent and treat age-related musculoskeletal diseases (sarcopenia, osteoporosis, and osteoarthritis) by reducing oxidative stress and inflammation and controlling the growth, differentiation, apoptosis, and autophagy of cells. This review paper provides comprehensive evidence of PUFAs on age-related musculoskeletal diseases, which will be helpful for exploitation into functional foods and drugs for their prevention and treatment. Conclusions: PUFAs could play an important role in the prevention and treatment of sarcopenia, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Haoqi Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ruogu Xiong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jin Cheng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jialu Ye
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yingzhen Qiu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Siyu Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Mengchu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhaoyan Liu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinzhu Pang
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011050, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011050, China
- Sun Yat-sen University-Mengniu Joint Research Center of Nutrition and Health for Middle-Aged and Elderly, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shanshan Guo
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011050, China
| | - Huabin Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huilian Zhu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Sun Yat-sen University-Mengniu Joint Research Center of Nutrition and Health for Middle-Aged and Elderly, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
15
|
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: 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: 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
|
16
|
Huang S, Jiang Y, Li J, Mao L, Qiu Z, Zhang S, Jiang Y, Liu Y, Liu W, Xiong Z, Zhang W, Liu X, Zhang Y, Bai X, Guo B. Osteocytes/Osteoblasts Produce SAA3 to Regulate Hepatic Metabolism of Cholesterol. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307818. [PMID: 38613835 PMCID: PMC11199997 DOI: 10.1002/advs.202307818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Hypercholesterolaemia is a systemic metabolic disease, but the role of organs other than liver in cholesterol metabolism is unappreciated. The phenotypic characterization of the Tsc1Dmp1 mice reveal that genetic depletion of tuberous sclerosis complex 1 (TSC1) in osteocytes/osteoblasts (Dmp1-Cre) triggers progressive increase in serum cholesterol level. The resulting cholesterol metabolic dysregulation is shown to be associated with upregulation and elevation of serum amyloid A3 (SAA3), a lipid metabolism related factor, in the bone and serum respectively. SAA3, elicited from the bone, bound to toll-like receptor 4 (TLR4) on hepatocytes to phosphorylate c-Jun, and caused impeded conversion of cholesterol to bile acids via suppression on cholesterol 7 α-hydroxylase (Cyp7a1) expression. Ablation of Saa3 in Tsc1Dmp1 mice prevented the CYP7A1 reduction in liver and cholesterol elevation in serum. These results expand the understanding of bone function and hepatic regulation of cholesterol metabolism and uncover a potential therapeutic use of pharmacological modulation of SAA3 in hypercholesterolaemia.
Collapse
Affiliation(s)
- Shijiang Huang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuanjun Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jing Li
- Department of Obstetrics and GynecologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Linlin Mao
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zeyou Qiu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Equipment Material DepartmentWest China Xiamen Hospital of Sichuan UniversityXiamenFujian361000China
| | - Sheng Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuhui Jiang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yong Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wen Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhi Xiong
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Wuju Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Central LaboratoryThe Fifth Affiliated HospitalSouthern Medical UniversityGuangzhouGuangdong510900China
| | - Xiaolin Liu
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yue Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdong510630China
| | - Bin Guo
- State Key Laboratory of Organ Failure ResearchDepartment of Cell BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- The Tenth Affiliated HospitalSouthern Medical UniversityDongguanGuangdong523018China
| |
Collapse
|
17
|
Tufail M, Wan WD, Jiang C, Li N. Targeting PI3K/AKT/mTOR signaling to overcome drug resistance in cancer. Chem Biol Interact 2024; 396:111055. [PMID: 38763348 DOI: 10.1016/j.cbi.2024.111055] [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: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
This review comprehensively explores the challenge of drug resistance in cancer by focusing on the pivotal PI3K/AKT/mTOR pathway, elucidating its role in oncogenesis and resistance mechanisms across various cancer types. It meticulously examines the diverse mechanisms underlying resistance, including genetic mutations, feedback loops, and microenvironmental factors, while also discussing the associated resistance patterns. Evaluating current therapeutic strategies targeting this pathway, the article highlights the hurdles encountered in drug development and clinical trials. Innovative approaches to overcome resistance, such as combination therapies and precision medicine, are critically analyzed, alongside discussions on emerging therapies like immunotherapy and molecularly targeted agents. Overall, this comprehensive review not only sheds light on the complexities of resistance in cancer but also provides a roadmap for advancing cancer treatment.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
18
|
Suszyńska-Zajczyk J, Witucki Ł, Perła-Kaján J, Jakubowski H. Diet-induced hyperhomocysteinemia causes sex-dependent deficiencies in offspring musculature and brain function. Front Cell Dev Biol 2024; 12:1322844. [PMID: 38559811 PMCID: PMC10979824 DOI: 10.3389/fcell.2024.1322844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Hyperhomocysteinemia (HHcy), characterized by elevated homocysteine (Hcy) levels, is a known risk factor for cardiovascular, renal, and neurological diseases, as well as pregnancy complications. Our study aimed to investigate whether HHcy induced by a high-methionine (high-Met) diet exacerbates cognitive and behavioral deficits in offspring and leads to other breeding problems. Dietary HHcy was induced four weeks before mating and continued throughout gestation and post-delivery. A battery of behavioral tests was conducted on offspring between postnatal days (PNDs) 5 and 30 to assess motor function/activity and cognition. The results were correlated with brain morphometric measurements and quantitative analysis of mammalian target of rapamycin (mTOR)/autophagy markers. The high-Met diet significantly increased parental and offspring urinary tHcy levels and influenced offspring behavior in a sex-dependent manner. Female offspring exhibited impaired cognition, potentially related to morphometric changes observed exclusively in HHcy females. Male HHcy pups demonstrated muscle weakness, evidenced by slower surface righting, reduced hind limb suspension (HLS) hanging time, weaker grip strength, and decreased activity in the beaker test. Western blot analyses indicated the downregulation of autophagy and the upregulation of mTOR activity in HHcy cortexes. HHcy also led to breeding impairments, including reduced breeding rate, in-utero fetal death, lower pups' body weight, and increased mortality, likely attributed to placental dysfunction associated with HHcy. In conclusion, a high-Met diet impairs memory and cognition in female juveniles and weakens muscle strength in male pups. These effects may stem from abnormal placental function affecting early neurogenesis, the dysregulation of autophagy-related pathways in the cortex, or epigenetic mechanisms of gene regulation triggered by HHcy during embryonic development.
Collapse
Affiliation(s)
- Joanna Suszyńska-Zajczyk
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Łukasz Witucki
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Perła-Kaján
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers University, New Jersey Medical School, International Center for Public Health, Newark, NJ, United States
| |
Collapse
|
19
|
Blitsman Y, Hollander E, Benafsha C, Yegodayev KM, Hadad U, Goldbart R, Traitel T, Rudich A, Elkabets M, Kost J. The Potential of PIP3 in Enhancing Wound Healing. Int J Mol Sci 2024; 25:1780. [PMID: 38339058 PMCID: PMC10855400 DOI: 10.3390/ijms25031780] [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: 12/14/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Given the role of phosphatidylinositol 3,4,5-trisphosphate (PIP3) in modulating cellular processes such as proliferation, survival, and migration, we hypothesized its potential as a novel therapeutic agent for wound closure enhancement. In this study, PIP3 was examined in its free form or as a complex with cationic starch (Q-starch) as a carrier. The intracellular bioactivity and localization of free PIP3 and the Q-starch/PIP3 complexes were examined. Our results present the capability of Q-starch to form complexes with PIP3, facilitate its cellular membrane internalization, and activate intracellular paths leading to enhanced wound healing. Both free PIP3 and Q-starch/PIP3 complexes enhanced monolayer gap closure in scratch assays and induced amplified collagen production within HaCAT and BJ fibroblast cells. Western blot presented enhanced AKT activation by free or complexed PIP3 in BJ fibroblasts in which endogenous PIP3 production was pharmacologically inhibited. Furthermore, both free PIP3 and Q-starch/PIP3 complexes expedited wound closure in mice, after single or daily dermal injections into the wound margins. Free PIP3 and the Q-starch/PIP3 complexes inherently activated the AKT signaling pathway, which is responsible for crucial wound healing processes such as migration; this was also observed in wound assays in mice. PIP3 was identified as a promising molecule for enhancing wound healing, and its ability to circumvent PI3K inhibition suggests possible implications for chronic wound healing.
Collapse
Affiliation(s)
- Yossi Blitsman
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| | - Etili Hollander
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| | - Chen Benafsha
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| | - Ksenia M. Yegodayev
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.M.Y.); (M.E.)
| | - Uzi Hadad
- The Ilse Katz Institute for Nanoscale Science and Technology, Marcus Campus, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
| | - Riki Goldbart
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| | - Tamar Traitel
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (K.M.Y.); (M.E.)
| | - Joseph Kost
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (Y.B.); (C.B.); (R.G.); (T.T.)
| |
Collapse
|
20
|
McColl TJ, Clarke DC. Kinetic modeling of leucine-mediated signaling and protein metabolism in human skeletal muscle. iScience 2024; 27:108634. [PMID: 38188514 PMCID: PMC10767222 DOI: 10.1016/j.isci.2023.108634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Skeletal muscle protein levels are governed by the relative rates of muscle protein synthesis (MPS) and breakdown (MPB). The mechanisms controlling these rates are complex, and their integrated behaviors are challenging to study through experiments alone. The purpose of this study was to develop and analyze a kinetic model of leucine-mediated mTOR signaling and protein metabolism in the skeletal muscle of young adults. Our model amalgamates published cellular-level models of the IRS1-PI3K-Akt-mTORC1 signaling system and of skeletal-muscle leucine kinetics with physiological-level models of leucine digestion and transport and insulin dynamics. The model satisfactorily predicts experimental data from diverse leucine feeding protocols. Model analysis revealed that total levels of p70S6K are a primary determinant of MPS, insulin signaling substantially affects muscle net protein balance via its effects on MPB, and p70S6K-mediated feedback of mTORC1 signaling reduces MPS in a dose-dependent manner.
Collapse
Affiliation(s)
- Taylor J. McColl
- Department of Biomedical Physiology and KinesiologySimon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - David C. Clarke
- Department of Biomedical Physiology and KinesiologySimon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
21
|
Stadhouders LEM, Smith JAB, Gabriel BM, Verbrugge SAJ, Hammersen TD, Kolijn D, Vogel ISP, Mohamed AD, de Wit GMJ, Offringa C, Hoogaars WMH, Gehlert S, Wackerhage H, Jaspers RT. Myotube growth is associated with cancer-like metabolic reprogramming and is limited by phosphoglycerate dehydrogenase. Exp Cell Res 2023; 433:113820. [PMID: 37879549 DOI: 10.1016/j.yexcr.2023.113820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/27/2023]
Abstract
The Warburg effect links growth and glycolysis in cancer. A key purpose of the Warburg effect is to generate glycolytic intermediates for anabolic reactions, such as nucleotides → RNA/DNA and amino acids → protein synthesis. The aim of this study was to investigate whether a similar 'glycolysis-for-anabolism' metabolic reprogramming also occurs in hypertrophying skeletal muscle. To interrogate this, we first induced C2C12 myotube hypertrophy with IGF-1. We then added 14C glucose to the differentiation medium and measured radioactivity in isolated protein and RNA to establish whether 14C had entered anabolism. We found that especially protein became radioactive, suggesting a glucose → glycolytic intermediates → non-essential amino acid(s) → protein series of reactions, the rate of which was increased by IGF-1. Next, to investigate the importance of glycolytic flux and non-essential amino acid synthesis for myotube hypertrophy, we exposed C2C12 and primary mouse myotubes to the glycolysis inhibitor 2-Deoxy-d-glucose (2DG). We found that inhibiting glycolysis lowered C2C12 and primary myotube size. Similarly, siRNA silencing of PHGDH, the key enzyme of the serine biosynthesis pathway, decreased C2C12 and primary myotube size; whereas retroviral PHGDH overexpression increased C2C12 myotube size. Together these results suggest that glycolysis is important for hypertrophying myotubes, which reprogram their metabolism to facilitate anabolism, similar to cancer cells.
Collapse
Affiliation(s)
- Lian E M Stadhouders
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Jonathon A B Smith
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Department of Physiology and Pharmacology (FYFA), Group of Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Brendan M Gabriel
- Aberdeen Cardiovascular & Diabetes Centre, The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Sander A J Verbrugge
- Exercise Biology, Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992, München/Munich, Germany
| | - Tim D Hammersen
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Detmar Kolijn
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands; Department of Clinical Pharmacology and Molecular Cardiology, Ruhr University Bochum, Bochum, Germany
| | - Ilse S P Vogel
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Abdalla D Mohamed
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; Cancer Therapeutics Unit, Target Genomic and Chromosomal Instability, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London, SM2 5NG, UK
| | - Gerard M J de Wit
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Carla Offringa
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Willem M H Hoogaars
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Sebastian Gehlert
- Department for the Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Universitätsplatz 1, 31141, Hildesheim, Germany; Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933, Cologne, Germany
| | - Henning Wackerhage
- Exercise Biology, Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992, München/Munich, Germany
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Furrer R, Heim B, Schmid S, Dilbaz S, Adak V, Nordström KJV, Ritz D, Steurer SA, Walter J, Handschin C. Molecular control of endurance training adaptation in male mouse skeletal muscle. Nat Metab 2023; 5:2020-2035. [PMID: 37697056 PMCID: PMC10663156 DOI: 10.1038/s42255-023-00891-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/11/2023] [Indexed: 09/13/2023]
Abstract
Skeletal muscle has an enormous plastic potential to adapt to various external and internal perturbations. Although morphological changes in endurance-trained muscles are well described, the molecular underpinnings of training adaptation are poorly understood. We therefore aimed to elucidate the molecular signature of muscles of trained male mice and unravel the training status-dependent responses to an acute bout of exercise. Our results reveal that, even though at baseline an unexpectedly low number of genes define the trained muscle, training status substantially affects the transcriptional response to an acute challenge, both quantitatively and qualitatively, in part associated with epigenetic modifications. Finally, transiently activated factors such as the peroxisome proliferator-activated receptor-γ coactivator 1α are indispensable for normal training adaptation. Together, these results provide a molecular framework of the temporal and training status-dependent exercise response that underpins muscle plasticity in training.
Collapse
Affiliation(s)
| | - Barbara Heim
- Biozentrum, University of Basel, Basel, Switzerland
- University Hospital Basel, Basel, Switzerland
| | - Svenia Schmid
- Biozentrum, University of Basel, Basel, Switzerland
- University Hospital Basel, Basel, Switzerland
| | - Sedat Dilbaz
- Biozentrum, University of Basel, Basel, Switzerland
| | - Volkan Adak
- Biozentrum, University of Basel, Basel, Switzerland
| | - Karl J V Nordström
- Laboratory of EpiGenetics, Saarland University, Saarbrücken, Germany
- AstraZeneca, Mölndal, Sweden
| | - Danilo Ritz
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Jörn Walter
- Laboratory of EpiGenetics, Saarland University, Saarbrücken, Germany
| | | |
Collapse
|
23
|
Cree T, Gomez TR, Timpani CA, Rybalka E, Price JT, Goodman CA. FKBP25 regulates myoblast viability and migration and is differentially expressed in in vivo models of muscle adaptation. FEBS J 2023; 290:4660-4678. [PMID: 37345229 DOI: 10.1111/febs.16894] [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: 09/01/2022] [Revised: 04/18/2023] [Accepted: 06/21/2023] [Indexed: 06/23/2023]
Abstract
FKBP25 (FKBP3 gene) is a dual-domain PPIase protein that consists of a C-terminal PPIase domain and an N-terminal basic tilted helix bundle (BTHB). The PPIase domain of FKBP25 has been shown to bind to microtubules, which has impacts upon microtubule polymerisation and cell cycle progression. Using quantitative proteomics, it was recently found that FKBP25 was expressed in the top 10% of the mouse skeletal muscle proteome. However, to date there have been few studies investigating the role of FKBP25 in non-transformed systems. As such, this study aimed to investigate potential roles for FKBP25 in myoblast viability, migration and differentiation and in adaptation of mature skeletal muscle. Doxycycline-inducible FKBP25 knockdown in C2C12 myoblasts revealed an increase in cell accumulation/viability and migration in vitro that was independent of alterations in tubulin dynamics; however, FKBP25 knockdown had no discernible impact on myoblast differentiation into myotubes. Finally, a series of in vivo models of muscle adaptation were assessed, where it was observed that FKBP25 protein expression was increased in hypertrophy and regeneration conditions (chronic mechanical overload and the mdx model of Duchenne muscular dystrophy) but decreased in an atrophy model (denervation). Overall, the findings of this study establish FKBP25 as a regulator of myoblast viability and migration, with possible implications for satellite cell proliferation and migration and muscle regeneration, and as a potential regulator of in vivo skeletal muscle adaptation.
Collapse
Affiliation(s)
- Tabitha Cree
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - Tania Ruz Gomez
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
| | - Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
| | - John T Price
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
- Monash Biomedicine Discovery Institute, Clayton, Australia
| | - Craig A Goodman
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Australia
- Department of Physiology, Centre for Muscle Research (CMR), The University of Melbourne, Parkville, Australia
| |
Collapse
|
24
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
25
|
Mirzoev TM. The emerging role of Piezo1 channels in skeletal muscle physiology. Biophys Rev 2023; 15:1171-1184. [PMID: 37975010 PMCID: PMC10643716 DOI: 10.1007/s12551-023-01154-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Piezo1 channels are mechanically activated (MA) cation channels that are involved in sensing of various mechanical perturbations, such as membrane stretch and shear stress, and play a crucial role in cell mechanotransduction. In response to mechanical stimuli, these channels open up and allow cations to travel into the cell and induce biochemical reactions that can change the cell's metabolism and function. Skeletal muscle cells/fibers inherently depend upon mechanical cues in the form of fluid shear stress and contractions (physical exercise). For example, an exposure of skeletal muscles to chronic mechanical loading leads to increased anabolism and fiber hypertrophy, while prolonged mechanical unloading results in muscle atrophy. MA Piezo1 channels have recently emerged as key mechanosensors that are capable of linking mechanical signals and intramuscular signaling in skeletal muscle cells/fibers. This review will summarize the emerging role of Piezo1 channels in the development and regeneration of skeletal muscle tissue as well as in the regulation of skeletal muscle atrophy. In addition, an overview of potential Piezo1-related signaling pathways underlying anabolic and catabolic processes will be provided. A better understanding of Piezo1's role in skeletal muscle mechanotransduction may represent an important basis for the development of therapeutic strategies for maintaining muscle functions under disuse conditions and in some disease states.
Collapse
Affiliation(s)
- Timur M. Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow, Russia
| |
Collapse
|
26
|
Furrer R, Hawley JA, Handschin C. The molecular athlete: exercise physiology from mechanisms to medals. Physiol Rev 2023; 103:1693-1787. [PMID: 36603158 PMCID: PMC10110736 DOI: 10.1152/physrev.00017.2022] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Human skeletal muscle demonstrates remarkable plasticity, adapting to numerous external stimuli including the habitual level of contractile loading. Accordingly, muscle function and exercise capacity encompass a broad spectrum, from inactive individuals with low levels of endurance and strength to elite athletes who produce prodigious performances underpinned by pleiotropic training-induced muscular adaptations. Our current understanding of the signal integration, interpretation, and output coordination of the cellular and molecular mechanisms that govern muscle plasticity across this continuum is incomplete. As such, training methods and their application to elite athletes largely rely on a "trial-and-error" approach, with the experience and practices of successful coaches and athletes often providing the bases for "post hoc" scientific enquiry and research. This review provides a synopsis of the morphological and functional changes along with the molecular mechanisms underlying exercise adaptation to endurance- and resistance-based training. These traits are placed in the context of innate genetic and interindividual differences in exercise capacity and performance, with special consideration given to aging athletes. Collectively, we provide a comprehensive overview of skeletal muscle plasticity in response to different modes of exercise and how such adaptations translate from "molecules to medals."
Collapse
Affiliation(s)
| | - John A Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | | |
Collapse
|
27
|
Mirzoev TM, Paramonova II, Rozhkov SV, Kalashnikova EP, Belova SP, Tyganov SA, Vilchinskaya NA, Shenkman BS. Metformin Pre-Treatment as a Means of Mitigating Disuse-Induced Rat Soleus Muscle Wasting. Curr Issues Mol Biol 2023; 45:3068-3086. [PMID: 37185725 PMCID: PMC10136829 DOI: 10.3390/cimb45040201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023] Open
Abstract
Currently, no ideal treatment exists to combat skeletal muscle disuse-induced atrophy and loss of strength. Because the activity of AMP-activated protein kinase (AMPK) in rat soleus muscle is suppressed at the early stages of disuse, we hypothesized that pre-treatment of rats with metformin (an AMPK activator) would exert beneficial effects on skeletal muscle during disuse. Muscle disuse was performed via hindlimb suspension (HS). Wistar rats were divided into four groups: (1) control (C), (2) control + metformin for 10 days (C+Met), (3) HS for 7 days (HS), (4) metformin treatment for 7 days before HS and during the first 3 days of 1-week HS (HS+Met). Anabolic and catabolic markers were assessed using WB and RT-PCR. Treatment with metformin partly prevented an HS-induced decrease in rat soleus weight and size of slow-twitch fibers. Metformin prevented HS-related slow-to-fast fiber transformation. Absolute soleus muscle force in the HS+Met group was increased vs. the HS group. GSK-3β (Ser9) phosphorylation was significantly increased in the HS+Met group vs. the HS group. Metformin pre-treatment partly prevented HS-induced decrease in 18S+28S rRNA content and attenuated upregulation of calpain-1 and ubiquitin. Thus, pre-treatment of rats with metformin can ameliorate disuse-induced reductions in soleus muscle weight, the diameter of slow-type fibers, and absolute muscle strength.
Collapse
Affiliation(s)
- Timur M Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| | - Inna I Paramonova
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| | - Sergey V Rozhkov
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| | | | - Svetlana P Belova
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| | - Sergey A Tyganov
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| | | | - Boris S Shenkman
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| |
Collapse
|
28
|
Lyu W, Kousaka M, Jia H, Kato H. Effects of Turmeric Extract on Age-Related Skeletal Muscle Atrophy in Senescence-Accelerated Mice. Life (Basel) 2023; 13:life13040941. [PMID: 37109470 PMCID: PMC10141758 DOI: 10.3390/life13040941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Muscle atrophy is one of the main causes of sarcopenia—the age-related loss of skeletal muscle. In this study, we investigated the effect of turmeric (Curcuma longa) extract (TE) supplementation on age-related muscle atrophy in a senescence-accelerated mouse model and explored the underlying mechanisms. Twenty-six-week-old male, senescence-accelerated mouse resistant (SAMR) mice received the AIN-93G basal diet, while twenty-six-week-old male, senescence-accelerated mouse prone 8 (SAMP8) mice received the AIN-93G basal diet or a 2% TE powder-supplemented diet for ten weeks. Our findings revealed that TE supplementation showed certain effects on ameliorating the decrease in body weight, tibialis anterior weight, and mesenteric fat tissue weight in SAMP8 mice. TE improved gene expression in the glucocorticoid receptor-FoxO signaling pathway in skeletal muscle, including redd1, klf15, foxo1, murf1, and mafbx. Furthermore, TE might have the certain potential on improving the dynamic balance between anabolic and catabolic processes by inhibiting the binding of glucocorticoid receptor or FoxO1 to the glucocorticoid response element or FoxO-binding element in the MuRF1 promoter in skeletal muscle, thereby promoting muscle mass and strength, and preventing muscle atrophy and sarcopenia prevention. Moreover, TE may have reduced mitochondrial damage and maintained cell growth and division by downregulating the mRNA expression of the genes mfn2 and tsc2. Thus, the results indicated TE’s potential for preventing age-related muscle atrophy and sarcopenia.
Collapse
Affiliation(s)
- Weida Lyu
- Health Nutrition, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0032, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Marika Kousaka
- Health Nutrition, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Huijuan Jia
- Health Nutrition, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hisanori Kato
- Health Nutrition, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
29
|
Santo André HC, Esteves GP, Barreto GHC, Longhini F, Dolan E, Benatti FB. The Influence of n-3PUFA Supplementation on Muscle Strength, Mass, and Function: A Systematic Review and Meta-Analysis. Adv Nutr 2023; 14:115-127. [PMID: 36811583 PMCID: PMC10103001 DOI: 10.1016/j.advnut.2022.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 12/25/2022] Open
Abstract
The effects of omega 3 polyunsaturated fatty acids (n-3PUFA) supplementation on skeletal muscle are currently unclear. The purpose of this systematic review was to synthesize all available evidence regarding the influence of n-3PUFA supplementation on muscle mass, strength, and function in healthy young and older adults. Four databases were searched (Medline, Embase, Cochrane CENTRAL, and SportDiscus). Predefined eligibility criteria were determined according to Population, Intervention, Comparator, Outcomes, and Study Design. Only peer-reviewed studies were included. The Cochrane RoB2 Tool and the NutriGrade approach were used to access risk of bias and certainty in evidence. Effect sizes were calculated using pre-post scores and analyzed using a three-level, random-effects meta-analysis. When sufficient studies were available, subanalyses were performed in the muscle mass, strength, and function outcomes according to participant's age (<60 or ≥60 years), supplementation dosage (<2 or ≥2 g/day), and training intervention ("resistance training" vs. "none or other"). Overall, 14 individual studies were included, total 1443 participants (913 females; 520 males) and 52 outcomes measures. Studies had high overall risk of bias and consideration of all NutriGrade elements resulted in a certainty assessment of moderate meta-evidence for all outcomes. n-3PUFA supplementation had no significant effect on muscle mass (standard mean difference [SMD] = 0.07 [95% CI: -0.02, 0.17], P = 0.11) and muscle function (SMD = 0.03 [95% CI: -0.09, 0.15], P = 0.58), but it showed a very small albeit significant positive effect on muscle strength (SMD = 0.12 [95% CI: 0.006, 0.24], P = 0.04) in participants when compared with placebo. Subgroup analyses showed that age, supplementation dose, or cosupplementation alongside resistance training did not influence these responses. In conclusion, our analyses indicated that n-3PUFA supplementation may lead to very small increases in muscle strength but did not impact muscle mass and function in healthy young and older adults. To our knowledge, this is the first review and meta-analysis investigating whether n-3PUFA supplementation can lead to increases in muscle strength, mass, and function in healthy adults. Registered protocol: doi.org/10.17605/OSF.IO/2FWQT.
Collapse
Affiliation(s)
| | - Gabriel P Esteves
- Applied Physiology & Nutrition Research Group, Rheumatology Division, Faculdade de Medicina FMUSP, University of São Paulo, São Paulo, SP, Brazil
| | - Gabriel H C Barreto
- Applied Physiology & Nutrition Research Group, Rheumatology Division, Faculdade de Medicina FMUSP, University of São Paulo, São Paulo, SP, Brazil
| | - Fernando Longhini
- School of Applied Science (FCA), State University of Campinas, Limeira, SP, Brazil
| | - Eimear Dolan
- Applied Physiology & Nutrition Research Group, Rheumatology Division, Faculdade de Medicina FMUSP, University of São Paulo, São Paulo, SP, Brazil
| | - Fabiana B Benatti
- School of Applied Science (FCA), State University of Campinas, Limeira, SP, Brazil; Applied Physiology & Nutrition Research Group, Rheumatology Division, Faculdade de Medicina FMUSP, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
30
|
Functional Nutrients to Ameliorate Neurogenic Muscle Atrophy. Metabolites 2022; 12:metabo12111149. [DOI: 10.3390/metabo12111149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Neurogenic muscle atrophy is a debilitating condition that occurs from nerve trauma in association with diseases or during aging, leading to reduced interaction between motoneurons and skeletal fibers. Current therapeutic approaches aiming at preserving muscle mass in a scenario of decreased nervous input include physical activity and employment of drugs that slow down the progression of the condition yet provide no concrete resolution. Nutritional support appears as a precious tool, adding to the success of personalized medicine, and could thus play a relevant part in mitigating neurogenic muscle atrophy. We herein summarize the molecular pathways triggered by denervation of the skeletal muscle that could be affected by functional nutrients. In this narrative review, we examine and discuss studies pertaining to the use of functional ingredients to counteract neurogenic muscle atrophy, focusing on their preventive or curative means of action within the skeletal muscle. We reviewed experimental models of denervation in rodents and in amyotrophic lateral sclerosis, as well as that caused by aging, considering the knowledge generated with use of animal experimental models and, also, from human studies.
Collapse
|
31
|
Abstract
Skeletal muscle mass is a very plastic characteristic of skeletal muscle and is regulated by signaling pathways that control the balance between anabolic and catabolic processes. The serine/threonine kinase mechanistic/mammalian target of rapamycin (mTOR) has been shown to be critically important in the regulation of skeletal muscle mass through its regulation of protein synthesis and degradation pathways. In this commentary, recent advances in the understanding of the role of mTORC1 in the regulation of muscle mass under conditions that induce hypertrophy and atrophy will be highlighted.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| |
Collapse
|
32
|
Gomisin G improves muscle strength by enhancing mitochondrial biogenesis and function in disuse muscle atrophic mice. Biomed Pharmacother 2022; 153:113406. [DOI: 10.1016/j.biopha.2022.113406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
|
33
|
Salucci S, Bartoletti-Stella A, Bavelloni A, Aramini B, Blalock WL, Fabbri F, Vannini I, Sambri V, Stella F, Faenza I. Extra Virgin Olive Oil (EVOO), a Mediterranean Diet Component, in the Management of Muscle Mass and Function Preservation. Nutrients 2022; 14:nu14173567. [PMID: 36079827 PMCID: PMC9459997 DOI: 10.3390/nu14173567] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 12/25/2022] Open
Abstract
Aging results in a progressive decline in skeletal muscle mass, strength and function, a condition known as sarcopenia. This pathological condition is due to multifactorial processes including physical inactivity, inflammation, oxidative stress, hormonal changes, and nutritional intake. Physical therapy remains the standard approach to treat sarcopenia, although some interventions based on dietary supplementation are in clinical development. In this context, thanks to its known anti-inflammatory and antioxidative properties, there is great interest in using extra virgin olive oil (EVOO) supplementation to promote muscle mass and health in sarcopenic patients. To date, the molecular mechanisms responsible for the pathological changes associated with sarcopenia remain undefined; however, a complete understanding of the signaling pathways that regulate skeletal muscle protein synthesis and their behavior during sarcopenia appears vital for defining how EVOO might attenuate muscle wasting during aging. This review highlights the main molecular players that control skeletal muscle mass, with particular regard to sarcopenia, and discusses, based on the more recent findings, the potential of EVOO in delaying/preventing loss of muscle mass and function, with the aim of stimulating further research to assess dietary supplementation with EVOO as an approach to prevent or delay sarcopenia in aging individuals.
Collapse
Affiliation(s)
- Sara Salucci
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy
- Correspondence:
| | - Anna Bartoletti-Stella
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Alberto Bavelloni
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine-DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy
| | - William L. Blalock
- “Luigi Luca Cavalli-Sforza” Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), 40136 Bologna, Italy
- IRCCS, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Vittorio Sambri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
- Unit of Microbiology, Greater Romagna Hub Laboratory, 47522 Pievesestina, Italy
| | - Franco Stella
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine-DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy
| | - Irene Faenza
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
34
|
Loquat Leaf Extract Enhances Muscle Contraction-Induced Activation of Protein Synthesis Signaling in Rat Skeletal Muscle. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2234118. [PMID: 35783523 PMCID: PMC9249488 DOI: 10.1155/2022/2234118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/22/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
Loquat (Eriobotrya japonica (Thunb.) Lindl.) leaves are traditionally used to improve muscle weakness, but their effects on muscle protein synthesis require further research. Therefore, we aimed to investigate whether loquat leaf extract (LLE) enhances muscle contraction-induced activation of muscle protein synthesis signaling. Male Wistar rats (12 weeks old, n = 6/group) were categorized into water treatment (CON) and LLE treatment (LLE) groups. The rats were administered distilled water or LLE (1.5 g/kg/day) once a day by oral gavage for 7 days. On day 7, at 3 h post-LLE administration, the gastrocnemius muscle in the right leg of each rat was stimulated by electrical muscle stimulation (EMS) (100 Hz, 30 V) through five sets of 10 isometric contractions (7 s contraction, 3 s rest) with 3 min interset intervals. The rats were then sacrificed, and the gastrocnemius muscles of both legs were excised at 3 h post-EMS. The phosphorylation levels of mammalian target of rapamycin complex 1 (mTORC1) signaling pathway molecules (Akt, mTOR, and p70S6K) were determined by Western blotting. Regarding the muscle contraction-induced protein synthesis signaling pathway, Akt phosphorylation at Ser473 was not significantly different between the CON and LLE groups. mTOR phosphorylation at Ser2448 was increased by EMS but did not show a significant difference between the CON and LLE groups. p70S6K phosphorylation at Thr389 was significantly increased in response to EMS, whereas the LLE group showed significantly higher p70S6K phosphorylation at Thr389 than that in the CON group. This suggests that LLE enhances muscle contraction-induced activation of p70S6K phosphorylation in rat skeletal muscles.
Collapse
|
35
|
Powers SK, Schrager M. Redox signaling regulates skeletal muscle remodeling in response to exercise and prolonged inactivity. Redox Biol 2022; 54:102374. [PMID: 35738088 PMCID: PMC9233275 DOI: 10.1016/j.redox.2022.102374] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
Skeletal muscle fibers are malleable and undergo rapid remodeling in response to increased contractile activity (i.e., exercise) or prolonged periods of muscle inactivity (e.g., prolonged bedrest). Exploration of the cell signaling pathways regulating these skeletal muscle adaptations reveal that redox signaling pathways play a key role in the control of muscle remodeling during both exercise and prolonged muscle inactivity. In this regard, muscular exercise results in an acute increase in the production of reactive oxygen species (ROS) in the contracting fibers; however, this contraction-induced rise in ROS production rapidly declines when contractions cease. In contrast, prolonged muscle disuse results in a chronic elevation in ROS production within the inactive fibers. This difference in the temporal pattern of ROS production in muscle during exercise and muscle inactivity stimulates divergent cell-signaling pathways that activate both genomic and nongenomic mechanisms to promote muscle remodeling. This review examines the role that redox signaling plays in skeletal muscle adaptation in response to both prolonged muscle inactivity and endurance exercise training. We begin with a summary of the sites of ROS production in muscle fibers followed by a review of the cellular antioxidants that are responsible for regulation of ROS levels in the cell. We then discuss the specific redox-sensitive signaling pathways that promote skeletal muscle adaptation in response to both prolonged muscle inactivity and exercise. To stimulate future research, we close with a discussion of unanswered questions in this exciting field.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Health Sciences, Stetson University, Deland, FL, 32723, USA.
| | - Matthew Schrager
- Department of Health Sciences, Stetson University, Deland, FL, 32723, USA
| |
Collapse
|
36
|
Nishimura Y, Chunthorng-Orn J, Lord S, Musa I, Dawson P, Holm L, Lai YC. Ubiquitin E3 ligase Atrogin-1 protein is regulated via the rapamycin-sensitive mTOR-S6K1 signaling pathway in C2C12 muscle cells. Am J Physiol Cell Physiol 2022; 323:C215-C225. [PMID: 35704697 DOI: 10.1152/ajpcell.00384.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atrogin-1 and MuRF1 are highly expressed in multiple conditions of skeletal muscle atrophy. The PI3K/Akt/FoxO signaling pathway is well known to regulate Atrogin-1 and MuRF1 gene expressions. However, Akt activation also activates the mammalian target of rapamycin complex 1 (mTORC1) which induces skeletal muscle hypertrophy. Whether mTORC1-dependent signaling has a role in regulating Atrogin-1 and/or MuRF1 gene and protein expression is currently unclear. In this study, we showed that activation of insulin-mediated Akt signaling suppresses both Atrogin-1 and MuRF1 protein contents and that inhibition of Akt increases both Atrogin-1 and MuRF1 protein contents in C2C12 myotubes. Interestingly, inhibition of mTORC1 using a specific mTORC1 inhibitor, rapamycin, increased Atrogin-1, but not MuRF1, protein content. Furthermore, activation of AMP-activated protein kinase (AMPK), a negative regulator of the mTORC1 signaling pathway, also showed distinct time-dependent changes between Atrogin-1 and MuRF1 protein contents, suggesting differential regulatory mechanisms between Atrogin-1 and MuRF1 protein content. To further explore the downstream of mTORC1 signaling, we employed a specific S6K1 inhibitor, PF-4708671. We found that Atrogin-1 protein content was dose-dependently increased with PF-4708671 treatment, whereas MuRF1 protein content was decreased at 50 μM of PF-4708671 treatment. However, MuRF1 protein content was unexpectedly increased when treated with PF-4708671 for a longer period. Overall, our results indicate that Atrogin-1 and MuRF1 protein contents are regulated by different mechanisms, the downstream of Akt, and that Atrogin-1 protein content can be regulated by rapamycin-sensitive mTOR-S6K1 dependent signaling pathway.
Collapse
Affiliation(s)
- Yusuke Nishimura
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jitpisute Chunthorng-Orn
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Samuel Lord
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ibrahim Musa
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter Dawson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Lars Holm
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
37
|
Gehlert S, Weinisch P, Römisch-Margl W, Jaspers RT, Artati A, Adamski J, Dyar KA, Aussieker T, Jacko D, Bloch W, Wackerhage H, Kastenmüller G. Effects of Acute and Chronic Resistance Exercise on the Skeletal Muscle Metabolome. Metabolites 2022; 12:445. [PMID: 35629949 PMCID: PMC9142957 DOI: 10.3390/metabo12050445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 12/18/2022] Open
Abstract
Resistance training promotes metabolic health and stimulates muscle hypertrophy, but the precise routes by which resistance exercise (RE) conveys these health benefits are largely unknown. AIM To investigate how acute RE affects human skeletal muscle metabolism. METHODS We collected vastus lateralis biopsies from six healthy male untrained volunteers at rest, before the first of 13 RE training sessions, and 45 min after the first and last bouts of RE. Biopsies were analysed using untargeted mass spectrometry-based metabolomics. RESULTS We measured 617 metabolites covering a broad range of metabolic pathways. In the untrained state RE altered 33 metabolites, including increased 3-methylhistidine and N-lactoylvaline, suggesting increased protein breakdown, as well as metabolites linked to ATP (xanthosine) and NAD (N1-methyl-2-pyridone-5-carboxamide) metabolism; the bile acid chenodeoxycholate also increased in response to RE in muscle opposing previous findings in blood. Resistance training led to muscle hypertrophy, with slow type I and fast/intermediate type II muscle fibre diameter increasing by 10.7% and 10.4%, respectively. Comparison of post-exercise metabolite levels between trained and untrained state revealed alterations of 46 metabolites, including decreased N-acetylated ketogenic amino acids and increased beta-citrylglutamate which might support growth. Only five of the metabolites that changed after acute exercise in the untrained state were altered after chronic training, indicating that training induces multiple metabolic changes not directly related to the acute exercise response. CONCLUSION The human skeletal muscle metabolome is sensitive towards acute RE in the trained and untrained states and reflects a broad range of adaptive processes in response to repeated stimulation.
Collapse
Affiliation(s)
- Sebastian Gehlert
- Department for Biosciences of Sports, Institute of Sport Science, University of Hildesheim, 31139 Hildesheim, Germany
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, 50933 Cologne, Germany; (T.A.); (D.J.); (W.B.)
| | - Patrick Weinisch
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (P.W.); (W.R.-M.)
| | - Werner Römisch-Margl
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (P.W.); (W.R.-M.)
| | - Richard T. Jaspers
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, 1081 BT Amsterdam, The Netherlands;
| | - Anna Artati
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Kenneth A. Dyar
- Metabolic Physiology, Institute of Diabetes and Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Thorben Aussieker
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, 50933 Cologne, Germany; (T.A.); (D.J.); (W.B.)
| | - Daniel Jacko
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, 50933 Cologne, Germany; (T.A.); (D.J.); (W.B.)
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, 50933 Cologne, Germany; (T.A.); (D.J.); (W.B.)
| | - Henning Wackerhage
- Department of Sport and Health Sciences, Technical University of Munich, 80809 Munich, Germany;
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (P.W.); (W.R.-M.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
38
|
Jia H, Yamashita T, Li X, Kato H. Laurel Attenuates Dexamethasone-Induced Skeletal Muscle Atrophy In Vitro and in a Rat Model. Nutrients 2022; 14:nu14102029. [PMID: 35631169 PMCID: PMC9143575 DOI: 10.3390/nu14102029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Prevention of muscle atrophy contributes to improved quality of life and life expectancy. In this study, we investigated the effects of laurel, selected from 34 spices and herbs, on dexamethasone (DEX)-induced skeletal muscle atrophy and deciphered the underlying mechanisms. Co-treatment of C2C12 myotubes with laurel for 12 h inhibited the DEX-induced expression of intracellular ubiquitin ligases—muscle atrophy F-box (atrogin-1/MAFbx) and muscle RING finger 1 (MuRF1)—and reduction in myotube diameter. Male Wistar rats were supplemented with 2% laurel for 17 days, with DEX-induced skeletal muscle atrophy occurring in the last 3 days. Laurel supplementation inhibited the mRNA expression of MuRF1, regulated DNA damage and development 1 (Redd1), and forkhead box class O 1 (Foxo1) in the muscles of rats. Mechanistically, we evaluated the effects of laurel on the cellular proteolysis machinery—namely, the ubiquitin/proteasome system and autophagy—and the mTOR signaling pathway, which regulates protein synthesis. These data indicated that the amelioration of DEX-induced skeletal muscle atrophy induced by laurel, is mainly mediated by the transcriptional inhibition of downstream factors of the ubiquitin-proteasome system. Thus, laurel may be a potential food ingredient that prevents muscle atrophy.
Collapse
|
39
|
Liu QQ, Xie WQ, Luo YX, Li YD, Huang WH, Wu YX, Li YS. High Intensity Interval Training: A Potential Method for Treating Sarcopenia. Clin Interv Aging 2022; 17:857-872. [PMID: 35656091 PMCID: PMC9152764 DOI: 10.2147/cia.s366245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sarcopenia, an age-related disease characterized by loss of muscle strength and muscle mass, has attracted the attention of medical experts due to its severe morbidity, low living quality, high expenditure of health care, and mortality. Traditionally, persistent aerobic exercise (PAE) is considered as a valid way to attenuate muscular atrophy. However, nowadays, high intensity interval training (HIIT) has emerged as a more effective and time-efficient method to replace traditional exercise modes. HIIT displays comprehensive effects on exercise capacity and skeletal muscle metabolism, and it provides a time-out for the recovery of cardiopulmonary and muscular functions without causing severe adverse effects. Studies demonstrated that compared with PAE, HIIT showed similar or even higher effects in improving muscle strength, enhancing physical performances and increasing muscle mass of elder people. Therefore, HIIT might become a promising way to cope with the age-related loss of muscle mass and muscle function. However, it is worth mentioning that no study of HIIT was conducted directly on sarcopenia patients, which is attributed to the suspicious of safety and validity. In this review, we will assess the effects of different training parameters on muscle and sarcopenia, summarize previous papers which compared the effects of HIIT and PAE in improving muscle quality and function, and evaluate the potential of HIIT to replace the status of PAE in treating old people with muscle atrophy and low modality; and point out drawbacks of temporary experiments. Our aim is to discuss the feasibility of HIIT to treat sarcopenia and provide a reference for clinical scientists who want to utilize HIIT as a new way to cope with sarcopenia.
Collapse
Affiliation(s)
- Qian-Qi Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410083, People’s Republic of China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Yu-Xuan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410083, People’s Republic of China
| | - Yi-Dan Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410083, People’s Republic of China
| | - Wei-Hong Huang
- Mobile Health Ministry of Education - China Mobile Joint Laboratory, Xiangya Hospital Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Yu-Xiang Wu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, Hubei, 430056, People’s Republic of China
- Yu-Xiang Wu, Department of Health and Kinesiology, School of Physical Education, Jianghan University, No. 8, Sanjiaohu Road, Wuhan, Hubei, 430056, People’s Republic of China, Tel +86 27 8422 6921, Email
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Correspondence: Yu-Sheng Li, Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People’s Republic of China, Tel +86-13975889696, Email
| |
Collapse
|
40
|
Long DE, Kosmac K, Dungan CM, Bamman MM, Peterson CA, Kern PA. Potential Benefits of Combined Statin and Metformin Therapy on Resistance Training Response in Older Individuals. Front Physiol 2022; 13:872745. [PMID: 35492586 PMCID: PMC9047873 DOI: 10.3389/fphys.2022.872745] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Metformin and statins are currently the focus of large clinical trials testing their ability to counter age-associated declines in health, but recent reports suggest that both may negatively affect skeletal muscle response to exercise. However, it has also been suggested that metformin may act as a possible protectant of statin-related muscle symptoms. The potential impact of combined drug use on the hypertrophic response to resistance exercise in healthy older adults has not been described. We present secondary statin analyses of data from the MASTERS trial where metformin blunted the hypertrophy response in healthy participants (>65 years) following 14 weeks of progressive resistance training (PRT) when compared to identical placebo treatment (n = 94). Approximately one-third of MASTERS participants were taking prescribed statins. Combined metformin and statin resulted in rescue of the metformin-mediated impaired growth response to PRT but did not significantly affect strength. Improved muscle fiber growth may be associated with medication-induced increased abundance of CD11b+/CD206+ M2-like macrophages. Sarcopenia is a significant problem with aging and this study identifies a potential interaction between these commonly used drugs which may help prevent metformin-related blunting of the beneficial effects of PRT.Trial Registration: ClinicalTrials.gov, NCT02308228, Registered on 25 November 2014.
Collapse
Affiliation(s)
- Douglas E. Long
- Department of Physical Therapy and Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Kate Kosmac
- Department of Physical Therapy and Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Cory M. Dungan
- Department of Physical Therapy and Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Marcas M. Bamman
- Florida Institute for Human and Machine Cognition, Pensacola, FL, United States
- Center for Exercise Medicine and Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charlotte A. Peterson
- Department of Physical Therapy and Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, United States
| | - Philip A. Kern
- Department of Internal Medicine, Division of Endocrinology, Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, United States
- *Correspondence: Philip A. Kern,
| |
Collapse
|
41
|
de Melo Madureira ÁN, de Oliveira JRS, de Menezes Lima VL. The Role of IL-6 Released During Exercise to Insulin Sensitivity and Muscle Hypertrophy. Mini Rev Med Chem 2022; 22:2419-2428. [PMID: 35264090 DOI: 10.2174/1389557522666220309161245] [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: 08/23/2021] [Revised: 11/25/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
Interleukin-6 (IL-6) influences both inflammatory response and anti-inflammatory processes. This cytokine can be released by the exercising skeletal muscle, which characterizes it as a myokine. Unlike what is observed in inflammation, IL-6 produced by skeletal muscle is not preceded by the release of other pro-inflammatory cytokines, but is seems to be dependent on the lactate produced during exercise, thus causing different effects from those of seen in inflammatory state. After binding to its receptor, myokine IL-6 activates the PI3K-Akt pathway. One consequence of this upregulation is the potentiation of insulin signaling, which enhances insulin sensitivity. IL-6 increases GLUT-4 vesicle mobilization to muscle cell periphery, increasing the glucose transport into the cell, and also glycogen synthesis. Muscle glycogen provides energy for the ATP resynthesis, and regulates Ca2+ release by the sarcoplasmic reticulum, influencing muscle contraction, and, hence, muscle function by multiple pathways. Another implication for the upregulation of PI3K-Akt pathway is the activation of mTORC1, which regulates mRNA translational efficiency by regulating translation machinery, and translational capacity by inducing ribosomal biogenesis. Thus, IL-6 may contribute for skeletal muscle hypertrophy and function by increasing contractile protein synthesis.
Collapse
Affiliation(s)
- Álvaro Nóbrega de Melo Madureira
- Laboratory of Lipids and Application of Biomolecules to Prevalent and Neglected Diseases (LAB-DPN), Department of Biochemistry, Federal University of Pernambuco (UFPE)
| | - João Ricardhis Saturnino de Oliveira
- Laboratory of Lipids and Application of Biomolecules to Prevalent and Neglected Diseases (LAB-DPN), Department of Biochemistry, Federal University of Pernambuco (UFPE)
| | - Vera Lúcia de Menezes Lima
- Laboratory of Lipids and Application of Biomolecules to Prevalent and Neglected Diseases (LAB-DPN), Department of Biochemistry, Federal University of Pernambuco (UFPE)
| |
Collapse
|
42
|
Rodgers BD, Ward CW. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev 2022; 43:329-365. [PMID: 34520530 PMCID: PMC8905337 DOI: 10.1210/endrev/bnab030] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Muscle wasting disease indications are among the most debilitating and often deadly noncommunicable disease states. As a comorbidity, muscle wasting is associated with different neuromuscular diseases and myopathies, cancer, heart failure, chronic pulmonary and renal diseases, peripheral neuropathies, inflammatory disorders, and, of course, musculoskeletal injuries. Current treatment strategies are relatively ineffective and can at best only limit the rate of muscle degeneration. This includes nutritional supplementation and appetite stimulants as well as immunosuppressants capable of exacerbating muscle loss. Arguably, the most promising treatments in development attempt to disrupt myostatin and activin receptor signaling because these circulating factors are potent inhibitors of muscle growth and regulators of muscle progenitor cell differentiation. Indeed, several studies demonstrated the clinical potential of "inhibiting the inhibitors," increasing muscle cell protein synthesis, decreasing degradation, enhancing mitochondrial biogenesis, and preserving muscle function. Such changes can prevent muscle wasting in various disease animal models yet many drugs targeting this pathway failed during clinical trials, some from serious treatment-related adverse events and off-target interactions. More often, however, failures resulted from the inability to improve muscle function despite preserving muscle mass. Drugs still in development include antibodies and gene therapeutics, all with different targets and thus, safety, efficacy, and proposed use profiles. Each is unique in design and, if successful, could revolutionize the treatment of both acute and chronic muscle wasting. They could also be used in combination with other developing therapeutics for related muscle pathologies or even metabolic diseases.
Collapse
Affiliation(s)
| | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
43
|
Teixeira FJ, Tavares N, Matias CN, Phillips SM. The effects of phosphatidic acid on performance and body composition - a scoping review. J Sports Sci 2022; 40:364-369. [PMID: 34706625 DOI: 10.1080/02640414.2021.1994769] [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] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
Phosphatidic acid (PA) is a lipid mediator proposed to increase muscle protein synthesis via direct stimulation of the mammalian target of rapamycin (mTOR) and may act as an anabolic supplemental aid. Evidence on the effectiveness of PA as an anabolic supplement is equivocal. We aimed to systematically assess the effect of PA on performance and body composition. Due to the small number of studies, this is a scoping review. A comprehensive search was performed in Pubmed, SPORTDiscus and Web of Science, from the 1 January 2010 to the 31 August 2020. Our search retrieved 2009 articles, which when filtered, resulted in six studies, published between 2012 and 2019, which were analysed further. Five studies were performed in adult male populations and one in an elderly male population. From these, three studies suggested no effect of PA on lean body mass , while the remaining showed a possible positive effect (body composition and performance improvements). In one of these, the supplement included other potentially anabolic substances, precluding an isolated effect of PA. After a thorough analysis of the studies included, the evidence does not support the supplementation with PA to increase performance or improve body composition in young or elderly men.
Collapse
Affiliation(s)
- Filipe J Teixeira
- Cbios (Research Center for Biosciences & Health Technologies), Universidade Lusófona De Humanidades E Tecnologias, Campo Grande, Lisboa, Portugal
- Atlântica, Instituto Universitário, Barcarena, Oeiras, Portugal
- Laboratory of Physiology and Biochemistry of Exercise (CIPER), Faculdade De Motricidade Humana, Universidade De Lisboa, Cruz-Quebrada, Portugal
- Innovation Direction, Bettery Lifelab, Lisbon, Portugal
| | - Nelson Tavares
- Cbios (Research Center for Biosciences & Health Technologies), Universidade Lusófona De Humanidades E Tecnologias, Campo Grande, Lisboa, Portugal
| | - Catarina N Matias
- Innovation Direction, Bettery Lifelab, Lisbon, Portugal
- CIDEFES -Universidade Lusófona, Lisboa, Portugal
| | | |
Collapse
|
44
|
Moberg M, Apró W, Horwath O, Hall G, Blackwood SJ, Katz A. Acute normobaric hypoxia blunts contraction-mediated mTORC1- and JNK-signaling in human skeletal muscle. Acta Physiol (Oxf) 2022; 234:e13771. [PMID: 34984845 PMCID: PMC9285439 DOI: 10.1111/apha.13771] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/28/2021] [Accepted: 01/01/2022] [Indexed: 12/26/2022]
Abstract
Aim Hypoxia has been shown to reduce resistance exercise‐induced stimulation of protein synthesis and long‐term gains in muscle mass. However, the mechanism whereby hypoxia exerts its effect is not clear. Here, we examine the effect of acute hypoxia on the activity of several signalling pathways involved in the regulation of muscle growth following a bout of resistance exercise. Methods Eight men performed two sessions of leg resistance exercise in normoxia or hypoxia (12% O2) in a randomized crossover fashion. Muscle biopsies were obtained at rest and 0, 90,180 minutes after exercise. Muscle analyses included levels of signalling proteins and metabolites associated with energy turnover. Results Exercise during normoxia induced a 5‐10‐fold increase of S6K1Thr389 phosphorylation throughout the recovery period, but hypoxia blunted the increases by ~50%. Phosphorylation of JNKThr183/Tyr185 and the JNK target SMAD2Ser245/250/255 was increased by 30‐ to 40‐fold immediately after the exercise in normoxia, but hypoxia blocked almost 70% of the activation. Throughout recovery, phosphorylation of JNK and SMAD2 remained elevated following the exercise in normoxia, but the effect of hypoxia was lost at 90‐180 minutes post‐exercise. Hypoxia had no effect on exercise‐induced Hippo or autophagy signalling and ubiquitin‐proteasome related protein levels. Nor did hypoxia alter the changes induced by exercise in high‐energy phosphates, glucose 6‐P, lactate or phosphorylation of AMPK or ACC. Conclusion We conclude that acute severe hypoxia inhibits resistance exercise‐induced mTORC1‐ and JNK signalling in human skeletal muscle, effects that do not appear to be mediated by changes in the degree of metabolic stress in the muscle.
Collapse
Affiliation(s)
- Marcus Moberg
- Åstrand Laboratory Department of Physiology, Nutrition and Biomechanics Swedish School of Sport and Health Sciences Stockholm Sweden
- Department of Physiology and Pharmacology Karolinska Institute Stockholm Sweden
| | - William Apró
- Åstrand Laboratory Department of Physiology, Nutrition and Biomechanics Swedish School of Sport and Health Sciences Stockholm Sweden
- Department of Clinical Science, Intervention and Technology Karolinska Institute Stockholm Sweden
| | - Oscar Horwath
- Åstrand Laboratory Department of Physiology, Nutrition and Biomechanics Swedish School of Sport and Health Sciences Stockholm Sweden
| | - Gerrit Hall
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Clinical Metabolomics Core Facility, Clinical Biochemistry Rigshospitalet Copenhagen Denmark
| | - Sarah Joan Blackwood
- Åstrand Laboratory Department of Physiology, Nutrition and Biomechanics Swedish School of Sport and Health Sciences Stockholm Sweden
| | - Abram Katz
- Åstrand Laboratory Department of Physiology, Nutrition and Biomechanics Swedish School of Sport and Health Sciences Stockholm Sweden
| |
Collapse
|
45
|
Wackerhage H, Vechetti IJ, Baumert P, Gehlert S, Becker L, Jaspers RT, de Angelis MH. Does a Hypertrophying Muscle Fibre Reprogramme its Metabolism Similar to a Cancer Cell? Sports Med 2022; 52:2569-2578. [PMID: 35460513 PMCID: PMC9584876 DOI: 10.1007/s40279-022-01676-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/01/2023]
Abstract
In 1924, Otto Warburg asked "How does the metabolism of a growing tissue differ from that of a non-growing tissue?" Currently, we know that proliferating healthy and cancer cells reprogramme their metabolism. This typically includes increased glucose uptake, glycolytic flux and lactate synthesis. A key function of this reprogramming is to channel glycolytic intermediates and other metabolites into anabolic reactions such as nucleotide-RNA/DNA synthesis, amino acid-protein synthesis and the synthesis of, for example, acetyl and methyl groups for epigenetic modification. In this review, we discuss evidence that a hypertrophying muscle similarly takes up more glucose and reprogrammes its metabolism to channel energy metabolites into anabolic pathways. We specifically discuss the functions of the cancer-associated enzymes phosphoglycerate dehydrogenase and pyruvate kinase muscle 2 in skeletal muscle. In addition, we ask whether increased glucose uptake by a hypertrophying muscle explains why muscularity is often negatively associated with type 2 diabetes mellitus and obesity.
Collapse
Affiliation(s)
- Henning Wackerhage
- Exercise Biology Group, Department of Health and Sports Sciences, Technical University of Munich, Munich, Germany
| | - Ivan J. Vechetti
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Philipp Baumert
- Exercise Biology Group, Department of Health and Sports Sciences, Technical University of Munich, Munich, Germany
| | - Sebastian Gehlert
- Department of Biosciences of Sports, Institute for Sports Science, University of Hildesheim, Hildesheim, Germany
| | - Lore Becker
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Richard T. Jaspers
- Laboratory for Myology, Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany ,Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| |
Collapse
|
46
|
Mesquita PHC, Vann CG, Phillips SM, McKendry J, Young KC, Kavazis AN, Roberts MD. Skeletal Muscle Ribosome and Mitochondrial Biogenesis in Response to Different Exercise Training Modalities. Front Physiol 2021; 12:725866. [PMID: 34646153 PMCID: PMC8504538 DOI: 10.3389/fphys.2021.725866] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle adaptations to resistance and endurance training include increased ribosome and mitochondrial biogenesis, respectively. Such adaptations are believed to contribute to the notable increases in hypertrophy and aerobic capacity observed with each exercise mode. Data from multiple studies suggest the existence of a competition between ribosome and mitochondrial biogenesis, in which the first adaptation is prioritized with resistance training while the latter is prioritized with endurance training. In addition, reports have shown an interference effect when both exercise modes are performed concurrently. This prioritization/interference may be due to the interplay between the 5’ AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) signaling cascades and/or the high skeletal muscle energy requirements for the synthesis and maintenance of cellular organelles. Negative associations between ribosomal DNA and mitochondrial DNA copy number in human blood cells also provide evidence of potential competition in skeletal muscle. However, several lines of evidence suggest that ribosome and mitochondrial biogenesis can occur simultaneously in response to different types of exercise and that the AMPK-mTORC1 interaction is more complex than initially thought. The purpose of this review is to provide in-depth discussions of these topics. We discuss whether a curious competition between mitochondrial and ribosome biogenesis exists and show the available evidence both in favor and against it. Finally, we provide future research avenues in this area of exercise physiology.
Collapse
Affiliation(s)
| | | | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - James McKendry
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Kaelin C Young
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| | | | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| |
Collapse
|
47
|
Mori T, Ato S, Knudsen JR, Henriquez-Olguin C, Li Z, Wakabayashi K, Suginohara T, Higashida K, Tamura Y, Nakazato K, Jensen TE, Ogasawara R. c-Myc overexpression increases ribosome biogenesis and protein synthesis independent of mTORC1 activation in mouse skeletal muscle. Am J Physiol Endocrinol Metab 2021; 321:E551-E559. [PMID: 34423683 DOI: 10.1152/ajpendo.00164.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
High-intensity muscle contractions (HiMCs) are known to increase c-Myc expression that is known to stimulate ribosome biogenesis and protein synthesis in most cells. However, although c-Myc mRNA transcription and c-Myc mRNA translation have been shown to be upregulated following resistance exercise concomitantly with increased ribosome biogenesis, this connection has not been tested directly. We investigated the effect of adeno-associated virus (AAV)-mediated c-Myc overexpression, with or without fasting or percutaneous electrical stimulation-induced HiMC, on ribosome biogenesis and protein synthesis in adult mouse skeletal muscles. AAV-mediated overexpression of c-Myc in mouse skeletal muscles for 2 wk increased the DNA polymerase subunit POL1 mRNA, 45S-pre-rRNA, total RNA, and muscle protein synthesis without altering mechanistic target of rapamycin complex 1 (mTORC1) signaling under both ad libitum and fasted conditions. RNA-sequencing (RNA-seq) analyses revealed that c-Myc overexpression mainly regulated ribosome biogenesis-related biological processes. The protein synthesis response to c-Myc overexpression mirrored the response with HiMC. No additional effect of combining c-Myc overexpression and HiMC was observed. Our results suggest that c-Myc overexpression is sufficient to stimulate skeletal muscle ribosome biogenesis and protein synthesis without activation of mTORC1. Therefore, the HiMC-induced increase in c-Myc may contribute to ribosome biogenesis and increased protein synthesis following HiMC.NEW & NOTEWORTHY Resistance exercise is known to increase c-Myc expression, which is known to stimulate ribosome biogenesis and protein synthesis in a variety of cells. However, whether the increase in c-Myc stimulates ribosome biogenesis and protein synthesis in skeletal muscles remains unknown. We found that c-Myc overexpression is sufficient to stimulate skeletal muscle ribosome biogenesis and protein synthesis without activation of mTORC1.
Collapse
Affiliation(s)
- Takahiro Mori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Satoru Ato
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Microsystems Laboratory 2, Institute of Microengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carlos Henriquez-Olguin
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Zhencheng Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Koki Wakabayashi
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Takeshi Suginohara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | | | - Yuki Tamura
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Koichi Nakazato
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Riki Ogasawara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Zhu WG, Hibbert JE, Lin KH, Steinert ND, Lemens JL, Jorgenson KW, Newman SM, Lamming DW, Hornberger TA. Weight Pulling: A Novel Mouse Model of Human Progressive Resistance Exercise. Cells 2021; 10:cells10092459. [PMID: 34572107 PMCID: PMC8465477 DOI: 10.3390/cells10092459] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022] Open
Abstract
This study describes a mouse model of progressive resistance exercise that utilizes a full-body/multi-joint exercise (weight pulling) along with a training protocol that mimics a traditional human paradigm (three training sessions per week, ~8–12 repetitions per set, 2 min of rest between sets, approximately two maximal-intensity sets per session, last set taken to failure, and a progressive increase in loading that is based on the individual’s performance). We demonstrate that weight pulling can induce an increase in the mass of numerous muscles throughout the body. The relative increase in muscle mass is similar to what has been observed in human studies, and is associated with the same type of long-term adaptations that occur in humans (e.g., fiber hypertrophy, myonuclear accretion, and, in some instances, a fast-to-slow transition in Type II fiber composition). Moreover, we demonstrate that weight pulling can induce the same type of acute responses that are thought to drive these long-term adaptations (e.g., the activation of signaling through mTORC1 and the induction of protein synthesis at 1 h post-exercise). Collectively, the results of this study indicate that weight pulling can serve as a highly translatable mouse model of progressive resistance exercise.
Collapse
Affiliation(s)
- Wenyuan G. Zhu
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jamie E. Hibbert
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kuan Hung Lin
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nathaniel D. Steinert
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jake L. Lemens
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kent W. Jorgenson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sarah M. Newman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (S.M.N.); (D.W.L.)
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (S.M.N.); (D.W.L.)
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Troy A. Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (W.G.Z.); (J.E.H.); (K.H.L.); (N.D.S.); (J.L.L.); (K.W.J.)
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- Correspondence:
| |
Collapse
|
49
|
Lysenko EA, Vinogradova OL, Popov DV. The Mechanisms of Muscle Mass and Strength Increase during Strength Training. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
50
|
Leuchtmann AB, Adak V, Dilbaz S, Handschin C. The Role of the Skeletal Muscle Secretome in Mediating Endurance and Resistance Training Adaptations. Front Physiol 2021; 12:709807. [PMID: 34456749 PMCID: PMC8387622 DOI: 10.3389/fphys.2021.709807] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Exercise, in the form of endurance or resistance training, leads to specific molecular and cellular adaptions not only in skeletal muscles, but also in many other organs such as the brain, liver, fat or bone. In addition to direct effects of exercise on these organs, the production and release of a plethora of different signaling molecules from skeletal muscle are a centerpiece of systemic plasticity. Most studies have so far focused on the regulation and function of such myokines in acute exercise bouts. In contrast, the secretome of long-term training adaptation remains less well understood, and the contribution of non-myokine factors, including metabolites, enzymes, microRNAs or mitochondrial DNA transported in extracellular vesicles or by other means, is underappreciated. In this review, we therefore provide an overview on the current knowledge of endurance and resistance exercise-induced factors of the skeletal muscle secretome that mediate muscular and systemic adaptations to long-term training. Targeting these factors and leveraging their functions could not only have broad implications for athletic performance, but also for the prevention and therapy in diseased and elderly populations.
Collapse
|