1
|
Andresen J. Short-term disuse shapes the adaptive response to resistance training. J Physiol 2025. [PMID: 40198856 DOI: 10.1113/jp288530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Affiliation(s)
- Jacob Andresen
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Childs CJ, Poling HM, Chen K, Tsai YH, Wu A, Vallie A, Eiken MK, Huang S, Sweet CW, Schreiner R, Xiao Z, Spencer RC, Paris SA, Conchola AS, Villanueva JW, Anderman MF, Holloway EM, Singh A, Giger RJ, Mahe MM, Loebel C, Helmrath MA, Walton KD, Rafii S, Spence JR. Coordinated differentiation of human intestinal organoids with functional enteric neurons and vasculature. Cell Stem Cell 2025; 32:640-651.e9. [PMID: 40043706 PMCID: PMC11973701 DOI: 10.1016/j.stem.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 11/11/2024] [Accepted: 02/07/2025] [Indexed: 03/20/2025]
Abstract
Human intestinal organoids (HIOs) derived from human pluripotent stem cells co-differentiate both epithelial and mesenchymal lineages in vitro but lack important cell types such as neurons, endothelial cells, and smooth muscle, which limits translational potential. Here, we demonstrate that the intestinal stem cell niche factor, EPIREGULIN (EREG), enhances HIO differentiation with epithelium, mesenchyme, enteric neuroglial populations, endothelial cells, and organized smooth muscle in a single differentiation, without the need for co-culture. When transplanted into a murine host, HIOs mature and demonstrate enteric nervous system function, undergoing peristaltic-like contractions indicative of a functional neuromuscular unit. HIOs also form functional vasculature, demonstrated in vitro using microfluidic devices and in vivo following transplantation, where HIO endothelial cells anastomose with host vasculature. These complex HIOs represent a transformative tool for translational research in the human gut and can be used to interrogate complex diseases as well as for testing therapeutic interventions with high fidelity to human pathophysiology.
Collapse
Affiliation(s)
- Charlie J Childs
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Holly M Poling
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Biomedical Engineering, University of Cincinnati College of Engineering and Applied Science, Cincinnati, OH, USA
| | - Kevin Chen
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yu-Hwai Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Angeline Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Abigail Vallie
- Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Madeline K Eiken
- Department of Biomedical Engineering, University of Michigan and University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Sha Huang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Caden W Sweet
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ryan Schreiner
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Zhiwei Xiao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ryan C Spencer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samantha A Paris
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ansley S Conchola
- Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jonathan W Villanueva
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Meghan F Anderman
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Akaljot Singh
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maxime M Mahe
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Université de Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan and University of Michigan College of Engineering, Ann Arbor, MI, USA; Department of Materials Science and Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katherine D Walton
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shahin Rafii
- Hartman Institute for Therapeutic Organ Regeneration, Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan and University of Michigan College of Engineering, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Shin Y, Yang J, Jeong H, Kim J, Lee B, Kim J, Lee S, Kwak J, Son Y, Kim K, Yang Y, Kim C, Kwon K, Lee K. ExermiR-129-3p Enhances Muscle Function by Improving Mitochondrial Activity Through PARP1 Inhibition. J Cachexia Sarcopenia Muscle 2025; 16:e13823. [PMID: 40254925 PMCID: PMC12010049 DOI: 10.1002/jcsm.13823] [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: 08/24/2024] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Physical exercise has beneficial effects on various organs, including skeletal muscle. However, not all patients are capable of engaging in exercise to maintain muscle function, which underscores the importance of identifying molecular mechanisms of physical training that could lead to the discovery of exercise-mimicking molecules. METHODS This study sought to identify molecular mediators of exercise that could improve muscle function. We focused on the exercise-induced microRNA (miR)-129-3p, investigating its role and effects on mitochondrial activity both in vivo and in vitro. The expression of miR-129-3p was analysed in skeletal muscle following exercise, and its downstream effects on the poly (ADP-ribose) polymerase-1 (Parp1)-SIRT1-PGC1α signalling pathway were elucidated. Functional studies were conducted using muscle-specific overexpression of miR-129-3p in adult mice and intramuscular injection of AAV9-miR-129-3p in obese mice to assess exercise capacity and muscle strength. RESULTS Exercise was found to upregulate miR-129-3p in skeletal muscle (p < 0.05), which directly inhibits Parp1, a major NAD+-consuming enzyme. This inhibition leads to increased NAD+ levels (p < 0.05), activating SIRT1 and subsequently reducing the acetylation of PGC1α, thereby enhancing mitochondrial function. Muscle-specific overexpression of miR-129-3p in adult mice significantly enhanced exercise capacity (> 130%, p < 0.0001), while AAV9-miR-129-3p injections ameliorated muscle weakness (twitch force, > 140%, p < 0.05; tetanic force, > 160%, p < 0.01) in obese mice. In human skeletal muscle myoblasts, miR-129-3p improved mitochondrial function via the PARP1-SIRT1-PGC1α signalling pathway. CONCLUSION Our findings suggest that miR-129-3p, induced by exercise, can mimic the beneficial effects of physical exercise. This highlights miR-129-3p as a potential therapeutic target for improving muscle health, especially in individuals unable to exercise.
Collapse
Affiliation(s)
- Yeo Jin Shin
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
| | - Jae Won Yang
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioscienceKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| | - Heeyeon Jeong
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioscienceKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| | - Joyeong Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioinformaticsKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| | - Bora Lee
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
| | - Ji‐Won Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
| | - Seung‐Min Lee
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
| | - Ju Yeon Kwak
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University College of MedicineDaejeonRepublic of Korea
| | - Young Hoon Son
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Biohybrid Systems Group, Coulter Department of Biomedical EngineeringGeorgia Institute of Technology & Emory University School of MedicineAtlantaGeorgiaUSA
| | - Kap Jung Kim
- Department of Orthopedic SurgeryEulji University College of MedicineDaejeonRepublic of Korea
| | - Yong Ryoul Yang
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioscienceKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| | - Chuna Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioinformaticsKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| | - Ki‐Sun Kwon
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Aventi Inc.DaejeonRepublic of Korea
| | - Kwang‐Pyo Lee
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of BioscienceKRIBB School, Korea University of Science and Technology (UST)DaejeonRepublic of Korea
| |
Collapse
|
4
|
Yang H, Zhang C, Kim W, Shi M, Kiliclioglu M, Bayram C, Bolar I, Tozlu ÖÖ, Baba C, Yuksel N, Yildirim S, Iqbal S, Sebhaoui J, Hacımuftuoglu A, Uhlen M, Boren J, Turkez H, Mardinoglu A. Multi-tissue network analysis reveals the effect of JNK inhibition on dietary sucrose-induced metabolic dysfunction in rats. eLife 2025; 13:RP98427. [PMID: 39932177 PMCID: PMC11813226 DOI: 10.7554/elife.98427] [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] [Indexed: 02/13/2025] Open
Abstract
Excessive consumption of sucrose, in the form of sugar-sweetened beverages, has been implicated in the pathogenesis of metabolic dysfunction-associated fatty liver disease (MAFLD) and other related metabolic syndromes. The c-Jun N-terminal kinase (JNK) pathway plays a crucial role in response to dietary stressors, and it was demonstrated that the inhibition of the JNK pathway could potentially be used in the treatment of MAFLD. However, the intricate mechanisms underlying these interventions remain incompletely understood given their multifaceted effects across multiple tissues. In this study, we challenged rats with sucrose-sweetened water and investigated the potential effects of JNK inhibition by employing network analysis based on the transcriptome profiling obtained from hepatic and extrahepatic tissues, including visceral white adipose tissue, skeletal muscle, and brain. Our data demonstrate that JNK inhibition by JNK-IN-5A effectively reduces the circulating triglyceride accumulation and inflammation in rats subjected to sucrose consumption. Coexpression analysis and genome-scale metabolic modeling reveal that sucrose overconsumption primarily induces transcriptional dysfunction related to fatty acid and oxidative metabolism in the liver and adipose tissues, which are largely rectified after JNK inhibition at a clinically relevant dose. Skeletal muscle exhibited minimal transcriptional changes to sucrose overconsumption but underwent substantial metabolic adaptation following the JNK inhibition. Overall, our data provides novel insights into the molecular basis by which JNK inhibition exerts its metabolic effect in the metabolically active tissues. Furthermore, our findings underpin the critical role of extrahepatic metabolism in the development of diet-induced steatosis, offering valuable guidance for future studies focused on JNK-targeting for effective treatment of MAFLD.
Collapse
Affiliation(s)
- Hong Yang
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Mengnan Shi
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Metin Kiliclioglu
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Ismail Bolar
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
| | - Özlem Özdemir Tozlu
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Cem Baba
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Nursena Yuksel
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical UniversityErzurumTurkiye
| | - Serkan Yildirim
- Department of Pathology, Veterinary Faculty, Atatürk UniversityErzurumTurkiye
- Department of Pathology, Faculty of Veterinary Medicine, Kyrgyz-Türkish Manas ÜniversityBishkekKyrgyzstan
| | - Shazia Iqbal
- Trustlife Labs Drug Research and Development CenterIstanbulTurkiye
| | - Jihad Sebhaoui
- Trustlife Labs Drug Research and Development CenterIstanbulTurkiye
| | - Ahmet Hacımuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Matthias Uhlen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk UniversityErzurumTurkiye
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholmSweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College LondonLondonUnited Kingdom
| |
Collapse
|
5
|
Shi Y, Zhou D, Wang H, Huang L, Gao X, Maitiabula G, Zhang L, Wang X. Succinate Regulates Exercise-Induced Muscle Remodelling by Boosting Satellite Cell Differentiation Through Succinate Receptor 1. J Cachexia Sarcopenia Muscle 2025; 16:e13670. [PMID: 39723719 DOI: 10.1002/jcsm.13670] [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: 06/11/2024] [Revised: 09/19/2024] [Accepted: 11/16/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Skeletal muscle remodelling can cause clinically important changes in muscle phenotypes. Satellite cells (SCs) myogenic potential underlies the maintenance of muscle plasticity. Accumulating evidence shows the importance of succinate in muscle metabolism and function. However, whether succinate can affect SC function and subsequently coordinate muscle remodelling to exercise remains unexplored. METHODS A mouse model of high-intensity interval training (HIIT) was used to investigate the effects of succinate on muscle remodelling and SC function by exercise capacity test and biochemical methods. Mice with succinate receptor 1 (SUCNR1)-specific knockout in SCs were generated as an in vivo model to explore the underlying mechanisms. RNA sequencing of isolated SCs was performed to identify molecular changes responding to succinate-SUCNR1 signalling. The effects of identified key molecules on the myogenic capacity of SCs were investigated using gain- and loss-of-function assays in vitro. To support the translational application, the clinical efficacy of succinate was explored in muscle-wasting mice. RESULTS After 21 days of HIIT, mice supplemented with 1.5% succinate exhibited striking gains in grip strength (+0.38 ± 0.04 vs. 0.26 ± 0.03 N, p < 0.001) and endurance (+276.70 ± 55.80 vs. 201.70 ± 45.31 s, p < 0.05), accompanied by enhanced muscle hypertrophy and neuromuscular junction regeneration (p < 0.001). The myogenic capacity of SCs was significantly increased in gastrocnemius muscle of mice supplemented with 1% and 1.5% succinate (+16.48% vs. control, p = 0.008; +47.25% vs. control, p < 0.001, respectively). SUCNR1-specific deletion in SCs abolished the modulatory influence of succinate on muscle adaptation in response to exercise, revealing that SCs respond to succinate-SUCNR1 signalling, thereby facilitating muscle remodelling. SUCNR1 signalling markedly upregulated genes associated with stem cell differentiation and phosphorylation pathways within SCs, of which p38α mitogen-activated protein kinase (MAPK; fold change = 6.7, p < 0.001) and protein kinase C eta (PKCη; fold change = 12.5, p < 0.001) expressions were the most enriched, respectively. Mechanistically, succinate enhanced the myogenic capacity of isolated SCs by activating the SUCNR1-PKCη-p38α MAPK pathway. Finally, succinate promoted SC differentiation (1.5-fold, p < 0.001), ameliorating dexamethasone-induced muscle atrophy in mice (p < 0.001). CONCLUSIONS Our findings reveal a novel function of succinate in enhancing SC myogenic capacity via SUCNR1, leading to enhanced muscle adaptation in response to exercise. These findings provide new insights for developing pharmacological strategies to overcome muscle atrophy-related diseases.
Collapse
Affiliation(s)
- Yifan Shi
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Da Zhou
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Haoyang Wang
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Longchang Huang
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xuejin Gao
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Gulisudumu Maitiabula
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Li Zhang
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xinying Wang
- Clinical Nutrition Service Center, Department of General Surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Dorry S, Perla S, Bennett AM. Mitogen-Activated Protein Kinase Phosphatase-5 is Required for TGF-β Signaling Through a JNK-Dependent Pathway. Mol Cell Biol 2025; 45:17-31. [PMID: 39607740 PMCID: PMC11693473 DOI: 10.1080/10985549.2024.2426665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in tissue fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Bahadoran Z, Mirmiran P, Ghasemi A. Type 2 diabetes-related sarcopenia: role of nitric oxide. Nutr Metab (Lond) 2024; 21:107. [PMID: 39695784 DOI: 10.1186/s12986-024-00883-z] [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: 09/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Sarcopenia, characterized by progressive and generalized loss of skeletal muscle (SkM) mass, strength, and physical performance, is a prevalent complication in type 2 diabetes (T2D). Nitric oxide (NO), a multifunctional gasotransmitter involved in whole-body glucose and insulin homeostasis, plays key roles in normal SkM physiology and function. Here, we highlight the role of NO in SkM mass maintenance and its potential contribution to the development of T2D-related sarcopenia. Physiologic NO level, primarily produced by sarcolemmal neuronal nitric oxide synthase (nNOSμ isoform), is involved in protein synthesis in muscle fibers and maintenance of SkM mass. The observed effect of nNOSμ on SkM mass is muscle-type specific and sex-dependent. Impaired NO homeostasis [due to a diminished nNOSμ-NO availability and excessive NO production through inducible NOS (iNOS) in response to atrophic stimuli, e.g., inflammatory cytokines] in SkM occurred during the development and progression of T2D, may cause sarcopenia. Theoretically, restoration of NO through nNOS overexpression, supplying NOS substrates (e.g., L-arginine and L-citrulline), phosphodiesterase (PDE) inhibition, and supplementation with NO donors (e.g., inorganic nitrate) may be potential therapeutic approaches to preserve SkM mass and prevents sarcopenia in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, P.O. Box 19395-4763, Tehran, Iran.
| |
Collapse
|
8
|
Alissa N, Fang WB, Medrano M, Bergeron N, Kozai Y, Hu Q, Redding C, Thyfault J, Hamilton-Reeves J, Berkland C, Cheng N. CCL2 signaling promotes skeletal muscle wasting in non-tumor and breast tumor models. Dis Model Mech 2024; 17:dmm050398. [PMID: 38973385 DOI: 10.1242/dmm.050398] [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: 07/17/2023] [Accepted: 05/15/2024] [Indexed: 07/09/2024] Open
Abstract
Despite advancements in treatment, approximately 25% of patients with breast cancer experience long-term skeletal muscle wasting (SMW), which limits mobility, reduces drug tolerance and adversely impacts survival. By understanding the underlying molecular mechanisms of SMW, we may be able to develop new strategies to alleviate this condition and improve the lives of patients with breast cancer. Chemokines are small soluble factors that regulate homing of immune cells to tissues during inflammation. In breast cancers, overexpression of C-C chemokine ligand 2 (CCL2) correlates with unfavorable prognosis. Elevated levels of CCL2 in peripheral blood indicate possible systemic effects of this chemokine in patients with breast cancer. Here, we investigated the role of CCL2 signaling on SMW in tumor and non-tumor contexts. In vitro, increasing concentrations of CCL2 inhibited myoblast and myotube function through C-C chemokine receptor 2 (CCR2)-dependent mechanisms involving JNK, SMAD3 and AMPK signaling. In healthy mice, delivery of recombinant CCL2 protein promoted SMW in a dose-dependent manner. In vivo knockdown of breast tumor-derived CCL2 partially protected against SMW. Overall, chronic, upregulated CCL2-CCR2 signaling positively regulates SMW, with implications for therapeutic targeting.
Collapse
Affiliation(s)
- Nadia Alissa
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Bin Fang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Marcela Medrano
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nick Bergeron
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yuuka Kozai
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Qingting Hu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Chloe Redding
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John Thyfault
- Department of Cell Biology and Physiology and Internal Medicine-Division of Endocrinology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Nikki Cheng
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Cancer Center, Kansas City, KS 66160, USA
| |
Collapse
|
9
|
Dorry S, Perla S, Bennett AM. MAPK Phosphatase-5 is required for TGF-β signaling through a JNK-dependent pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600976. [PMID: 38979264 PMCID: PMC11230413 DOI: 10.1101/2024.06.27.600976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Fowler A, Knaus KR, Khuu S, Khalilimeybodi A, Schenk S, Ward SR, Fry AC, Rangamani P, McCulloch AD. Network model of skeletal muscle cell signalling predicts differential responses to endurance and resistance exercise training. Exp Physiol 2024; 109:939-955. [PMID: 38643471 PMCID: PMC11140181 DOI: 10.1113/ep091712] [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: 12/06/2023] [Accepted: 03/20/2024] [Indexed: 04/22/2024]
Abstract
Exercise-induced muscle adaptations vary based on exercise modality and intensity. We constructed a signalling network model from 87 published studies of human or rodent skeletal muscle cell responses to endurance or resistance exercise in vivo or simulated exercise in vitro. The network comprises 259 signalling interactions between 120 nodes, representing eight membrane receptors and eight canonical signalling pathways regulating 14 transcriptional regulators, 28 target genes and 12 exercise-induced phenotypes. Using this network, we formulated a logic-based ordinary differential equation model predicting time-dependent molecular and phenotypic alterations following acute endurance and resistance exercises. Compared with nine independent studies, the model accurately predicted 18/21 (85%) acute responses to resistance exercise and 12/16 (75%) acute responses to endurance exercise. Detailed sensitivity analysis of differential phenotypic responses to resistance and endurance training showed that, in the model, exercise regulates cell growth and protein synthesis primarily by signalling via mechanistic target of rapamycin, which is activated by Akt and inhibited in endurance exercise by AMP-activated protein kinase. Endurance exercise preferentially activates inflammation via reactive oxygen species and nuclear factor κB signalling. Furthermore, the expected preferential activation of mitochondrial biogenesis by endurance exercise was counterbalanced in the model by protein kinase C in response to resistance training. This model provides a new tool for investigating cross-talk between skeletal muscle signalling pathways activated by endurance and resistance exercise, and the mechanisms of interactions such as the interference effects of endurance training on resistance exercise outcomes.
Collapse
Affiliation(s)
- Annabelle Fowler
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Katherine R. Knaus
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Stephanie Khuu
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Simon Schenk
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Samuel R. Ward
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew C. Fry
- Department of Health, Sport and Exercise SciencesUniversity of KansasLawrenceKansasUSA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew D. McCulloch
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
11
|
Millward DJ. Post-natal muscle growth and protein turnover: a narrative review of current understanding. Nutr Res Rev 2024; 37:141-168. [PMID: 37395180 DOI: 10.1017/s0954422423000124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A model explaining the dietary-protein-driven post-natal skeletal muscle growth and protein turnover in the rat is updated, and the mechanisms involved are described, in this narrative review. Dietary protein controls both bone length and muscle growth, which are interrelated through mechanotransduction mechanisms with muscle growth induced both from stretching subsequent to bone length growth and from internal work against gravity. This induces satellite cell activation, myogenesis and remodelling of the extracellular matrix, establishing a growth capacity for myofibre length and cross-sectional area. Protein deposition within this capacity is enabled by adequate dietary protein and other key nutrients. After briefly reviewing the experimental animal origins of the growth model, key concepts and processes important for growth are reviewed. These include the growth in number and size of the myonuclear domain, satellite cell activity during post-natal development and the autocrine/paracrine action of IGF-1. Regulatory and signalling pathways reviewed include developmental mechanotransduction, signalling through the insulin/IGF-1-PI3K-Akt and the Ras-MAPK pathways in the myofibre and during mechanotransduction of satellite cells. Likely pathways activated by maximal-intensity muscle contractions are highlighted and the regulation of the capacity for protein synthesis in terms of ribosome assembly and the translational regulation of 5-TOPmRNA classes by mTORC1 and LARP1 are discussed. Evidence for and potential mechanisms by which volume limitation of muscle growth can occur which would limit protein deposition within the myofibre are reviewed. An understanding of how muscle growth is achieved allows better nutritional management of its growth in health and disease.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, School of Biosciences & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
12
|
Barai P, Chen J. Beyond protein synthesis: non-translational functions of threonyl-tRNA synthetases. Biochem Soc Trans 2024; 52:661-670. [PMID: 38477373 PMCID: PMC11088916 DOI: 10.1042/bst20230506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
Aminoacyl-tRNA synthetases (AARSs) play an indispensable role in the translation of mRNAs into proteins. It has become amply clear that AARSs also have non-canonical or non-translational, yet essential, functions in a myriad of cellular and developmental processes. In this mini-review we discuss the current understanding of the roles of threonyl-tRNA synthetase (TARS) beyond protein synthesis and the underlying mechanisms. The two proteins in eukaryotes - cytoplasmic TARS1 and mitochondrial TARS2 - exert their non-canonical functions in the regulation of gene expression, cell signaling, angiogenesis, inflammatory responses, and tumorigenesis. The TARS proteins utilize a range of biochemical mechanisms, including assembly of a translation initiation complex, unexpected protein-protein interactions that lead to activation or inhibition of intracellular signaling pathways, and cytokine-like signaling through cell surface receptors in inflammation and angiogenesis. It is likely that new functions and novel mechanisms will continue to emerge for these multi-talented proteins.
Collapse
Affiliation(s)
- Pallob Barai
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jie Chen
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
13
|
Soares RN, Lessard SJ. Low Response to Aerobic Training in Metabolic Disease: Role of Skeletal Muscle. Exerc Sport Sci Rev 2024; 52:47-53. [PMID: 38112622 PMCID: PMC10963145 DOI: 10.1249/jes.0000000000000331] [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] [Indexed: 12/21/2023]
Abstract
Aerobic exercise is established to increase cardiorespiratory fitness (CRF), which is linked to reduced morbidity and mortality. However, people with metabolic diseases such as type 1 and type 2 diabetes may be more likely to display blunted improvements in CRF with training. Here, we present evidence supporting the hypothesis that altered skeletal muscle signaling and remodeling may contribute to low CRF with metabolic disease.
Collapse
|
14
|
O'Leary MF, Jackman SR, Bowtell JL. Shatavari supplementation in postmenopausal women alters the skeletal muscle proteome and pathways involved in training adaptation. Eur J Nutr 2024; 63:869-879. [PMID: 38214710 PMCID: PMC10948523 DOI: 10.1007/s00394-023-03310-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 12/10/2023] [Indexed: 01/13/2024]
Abstract
PURPOSE Shatavari is an understudied, widely available herbal supplement. It contains steroidal saponins and phytoestrogens. We previously showed that six weeks of shatavari supplementation improved handgrip strength and increased markers of myosin contractile function. Mechanistic insights into shatavari's actions are limited. Therefore, we performed proteomics on vastus lateralis (VL) samples that remained from our original study. METHODS In a randomised double-blind trial, women (68.5 ± 6 years) ingested either placebo or shatavari (equivalent to 26,500 mg/d fresh weight) for six weeks. Tandem mass tag global proteomic analysis of VL samples was conducted (N = 7 shatavari, N = 5 placebo). Data were normalized to total peptides and scaled using a reference sample. Data were filtered using a 5% FDR. For each protein, the pre to post supplementation difference was expressed as log2 fold change. Welch's t tests with Benjamini-Hochberg corrections were performed for each protein. Pathway enrichment (PADOG, CAMERA) was interrogated in Reactome (v85). RESULTS No individual protein was significantly different between supplementation conditions. Both PADOG and CAMERA indicated that pathways related to (1) Integrin/MAPK signalling, (2) metabolism/insulin secretion; (3) cell proliferation/senescence/DNA repair/cell death; (4) haemostasis/platelets/fibrin; (5) signal transduction; (6) neutrophil degranulation and (7) chemical synapse function were significantly upregulated. CAMERA indicated pathways related to translation/amino acid metabolism, viral infection, and muscle contraction were downregulated. CONCLUSION Our analyses indicate that shatavari may support muscle adaptation responses to exercise. These data provide useful signposts for future investigation of shatavari's utility in conserving and enhancing musculoskeletal function in older age. TRIAL REGISTRATION NCT05025917 30/08/21, retrospectively registered.
Collapse
Affiliation(s)
- Mary F O'Leary
- Department of Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| | - Sarah R Jackman
- Department of Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Joanna L Bowtell
- Department of Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
15
|
Hesketh SJ. Advancing cancer cachexia diagnosis with -omics technology and exercise as molecular medicine. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:1-15. [PMID: 38463663 PMCID: PMC10918365 DOI: 10.1016/j.smhs.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 03/12/2024] Open
Abstract
Muscle atrophy exacerbates disease outcomes and increases mortality, whereas the preservation of skeletal muscle mass and function play pivotal roles in ensuring long-term health and overall quality-of-life. Muscle atrophy represents a significant clinical challenge, involving the continued loss of muscle mass and strength, which frequently accompany the development of numerous types of cancer. Cancer cachexia is a highly prevalent multifactorial syndrome, and although cachexia is one of the main causes of cancer-related deaths, there are still no approved management strategies for the disease. The etiology of this condition is based on the upregulation of systemic inflammation factors and catabolic stimuli, resulting in the inhibition of protein synthesis and enhancement of protein degradation. Numerous necessary cellular processes are disrupted by cachectic pathology, which mediate intracellular signalling pathways resulting in the net loss of muscle and organelles. However, the exact underpinning molecular mechanisms of how these changes are orchestrated are incompletely understood. Much work is still required, but structured exercise has the capacity to counteract numerous detrimental effects linked to cancer cachexia. Primarily through the stimulation of muscle protein synthesis, enhancement of mitochondrial function, and the release of myokines. As a result, muscle mass and strength increase, leading to improved mobility, and quality-of-life. This review summarises existing knowledge of the complex molecular networks that regulate cancer cachexia and exercise, highlighting the molecular interplay between the two for potential therapeutic intervention. Finally, the utility of mass spectrometry-based proteomics is considered as a way of establishing early diagnostic biomarkers of cachectic patients.
Collapse
|
16
|
Tan Q, He Q, Peng Z, Zeng X, Liu Y, Li D, Wang S, Wang J. Topical rhubarb charcoal-crosslinked chitosan/silk fibroin sponge scaffold for the repair of diabetic ulcers improves hepatic lipid deposition in db/db mice via the AMPK signalling pathway. Lipids Health Dis 2024; 23:52. [PMID: 38378566 PMCID: PMC10877747 DOI: 10.1186/s12944-024-02041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/04/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is closely linked to metabolic syndrome, characterised by insulin resistance, hyperglycaemia, abnormal lipid metabolism, and chronic inflammation. Diabetic ulcers (DUs) comprise consequential complications that arise as a result of T2DM. To investigate, db/db mice were used for the disease model. The findings demonstrated that a scaffold made from a combination of rhubarb charcoal-crosslinked chitosan and silk fibroin, designated as RCS/SF, was able to improve the healing process of diabetic wounds in db/db mice. However, previous studies have primarily concentrated on investigating the impacts of the RSC/SF scaffold on wound healing only, while its influence on the entire body has not been fully elucidated. MATERIAL AND METHODS The silk fibroin/chitosan sponge scaffold containing rhubarb charcoal was fabricated in the present study using a freeze-drying approach. Subsequently, an incision with a diameter of 8 mm was made on the dorsal skin of the mice, and the RCS/SF scaffold was applied directly to the wound for 14 days. Subsequently, the impact of RCS/SF scaffold therapy on hepatic lipid metabolism was assessed through analysis of serum and liver biochemistry, histopathology, quantitative real-time PCR (qRT-PCR), immunohistochemistry, and Western blotting. RESULTS The use of the RCS/SF scaffold led to an enhancement in the conditions associated with serum glucolipid metabolism in db/db mice. An assessment of hepatic histopathology further confirmed this enhancement. Additionally, the qRT-PCR analysis revealed that treatment with RCS/SF scaffold resulted in the downregulation of genes associated with fatty acid synthesis, fatty acid uptake, triglyceride (TG) synthesis, gluconeogenesis, and inflammatory factors. Moreover, the beneficial effect of the RCS/SF scaffold on oxidative stress was shown by assessing antioxidant enzymes and lipid peroxidation. Additionally, the network pharmacology analysis verified that the adenosine monophosphate-activated protein kinase (AMPK) signalling pathway had a vital function in mitigating non-alcoholic fatty liver disease (NAFLD) by utilizing R. officinale. The measurement of AMPK, sterol regulatory element binding protein 1 (SREBP1), fatty acid synthase (FASN), and acetyl CoA carboxylase (ACC) gene and protein expression provided support for this discovery. Furthermore, the molecular docking investigations revealed a robust affinity between the active components of rhubarb and the downstream targets of AMPK (SREBP1 and FASN). CONCLUSION By regulating the AMPK signalling pathway, the RCS/SF scaffold applied topically effectively mitigated hepatic lipid accumulation, decreased inflammation, and attenuated oxidative stress. The present study, therefore, emphasises the crucial role of the topical RCS/SF scaffold in regulating hepatic lipid metabolism, thereby confirming the concept of "external and internal reshaping".
Collapse
Affiliation(s)
- Qi Tan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qifeng He
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Ze Peng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xin Zeng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yuzhe Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Dong Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China.
| |
Collapse
|
17
|
Radak Z, Pan L, Zhou L, Mozaffaritabar S, Gu Y, A Pinho R, Zheng X, Ba X, Boldogh I. Epigenetic and "redoxogenetic" adaptation to physical exercise. Free Radic Biol Med 2024; 210:65-74. [PMID: 37977212 DOI: 10.1016/j.freeradbiomed.2023.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Exercise-induced adaptation is achieved by altering the epigenetic landscape of the entire genome leading to the expression of genes involved in various processes including regulatory, metabolic, adaptive, immune, and myogenic functions. Clinical and experimental data suggest that the methylation pattern/levels of promoter/enhancer is not linearly correlated with gene expression and proteome levels during physical activity implying a level of complexity and interplay with other regulatory modulators. It has been shown that a higher level of physical fitness is associated with a slower DNA methylation-based aging clock. There is strong evidence supporting exercise-induced ROS being a key regulatory mediator through overlapping events, both as signaling entities and through oxidative modifications to various protein mediators and DNA molecules. ROS generated by physical activity shapes epigenome both directly and indirectly, a complexity we are beginning to unravel within the epigenetic arrangement. Oxidative modification of guanine to 8-oxoguanine is a non-genotoxic alteration, does not distort DNA helix and serves as an epigenetic-like mark. The reader and eraser of oxidized guanine is the 8-oxoguanine DNA glycosylase 1, contributing to changes in gene expression. In fact, it can modulate methylation patterns of promoters/enhancers consequently leading to multiple phenotypic changes. Here, we provide evidence and discuss the potential roles of exercise-induced ROS in altering cytosine methylation patterns during muscle adaptation processes.
Collapse
Affiliation(s)
- Zsolt Radak
- Research Center for Molecular Exercise Science, Hungarian University of Sport Science, 1123, Budapest, Hungary; Faculty of Sport Sciences, Waseda University, Tokorozawa, 359-1192, Japan.
| | - Lang Pan
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX77555, USA
| | - Lei Zhou
- Research Center for Molecular Exercise Science, Hungarian University of Sport Science, 1123, Budapest, Hungary
| | - Soroosh Mozaffaritabar
- Research Center for Molecular Exercise Science, Hungarian University of Sport Science, 1123, Budapest, Hungary
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Ricardo A Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Xu Zheng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, School of Life Science, Northeast Normal University, Changchun, Jilin, China; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX77555, USA
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of Ministry of Education, School of Life Science, Northeast Normal University, Changchun, Jilin, China; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX77555, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX77555, USA
| |
Collapse
|
18
|
Afsar B, Afsar RE. Sodium-glucose co-transporter 2 inhibitors and Sarcopenia: A controversy that must be solved. Clin Nutr 2023; 42:2338-2352. [PMID: 37862820 DOI: 10.1016/j.clnu.2023.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/22/2023]
Abstract
Diabetes mellitus is a risk factor for muscle loss and sarcopenia. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) or "gliflozins" are one of the newest anti-hyperglycemic drugs. They reduce blood glucose levels by inhibiting renal glucose reabsorption in the early proximal convoluted tubule. Various randomized trials showed that SGLT2i have cardio-protective and reno-protective action. SGLT2i also affect body composition. They usually decrease body fat percentage, visceral and subcutaneous adipose tissue. However, regarding the muscle mass, there are conflicting findings some studies showing detrimental effects and others showed neutral or beneficial effects. This issue is extremely important not only because of the wide use of SGLT2i around globe; but also skeletal muscle mass consumes large amounts of calories during exercise and is an important determinant of resting metabolic rate and skeletal muscle loss hinders energy consumption leading to obesity. In this systematic review, we extensively reviewed the experimental and clinical studies regarding the impact of SGLT2i on muscle mass and related metabolic alterations. Importantly, studies are heterogeneous and there is unmet need to highlight the alterations in muscle during SGLT2i use.
Collapse
Affiliation(s)
- Baris Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey.
| | - Rengin Elsurer Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey
| |
Collapse
|
19
|
Vived C, Lee-Papastavros A, Aparecida da Silva Pereira J, Yi P, MacDonald TL. β Cell Stress and Endocrine Function During T1D: What Is Next to Discover? Endocrinology 2023; 165:bqad162. [PMID: 37947352 DOI: 10.1210/endocr/bqad162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Canonically, type 1 diabetes (T1D) is a disease characterized by autoreactive T cells as perpetrators of endocrine dysfunction and β cell death in the spiral toward loss of β cell mass, hyperglycemia, and insulin dependence. β Cells have mostly been considered as bystanders in a flurry of autoimmune processes. More recently, our framework for understanding and investigating T1D has evolved. It appears increasingly likely that intracellular β cell stress is an important component of T1D etiology/pathology that perpetuates autoimmunity during the progression to T1D. Here we discuss the emerging and complex role of β cell stress in initiating, provoking, and catalyzing T1D. We outline the bridges between hyperglycemia, endoplasmic reticulum stress, oxidative stress, and autoimmunity from the viewpoint of intrinsic β cell (dys)function, and we extend this discussion to the potential role for a therapeutic β cell stress-metabolism axis in T1D. Lastly, we mention research angles that may be pursued to improve β cell endocrine function during T1D. Biology gleaned from studying T1D will certainly overlap to innovate therapeutic strategies for T2D, and also enhance the pursuit of creating optimized stem cell-derived β cells as endocrine therapy.
Collapse
Affiliation(s)
- Celia Vived
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jéssica Aparecida da Silva Pereira
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Tara L MacDonald
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
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: 72] [Impact Index Per Article: 36.0] [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
|
21
|
Smith JAB, Murach KA, Dyar KA, Zierath JR. Exercise metabolism and adaptation in skeletal muscle. Nat Rev Mol Cell Biol 2023; 24:607-632. [PMID: 37225892 PMCID: PMC10527431 DOI: 10.1038/s41580-023-00606-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/26/2023]
Abstract
Viewing metabolism through the lens of exercise biology has proven an accessible and practical strategy to gain new insights into local and systemic metabolic regulation. Recent methodological developments have advanced understanding of the central role of skeletal muscle in many exercise-associated health benefits and have uncovered the molecular underpinnings driving adaptive responses to training regimens. In this Review, we provide a contemporary view of the metabolic flexibility and functional plasticity of skeletal muscle in response to exercise. First, we provide background on the macrostructure and ultrastructure of skeletal muscle fibres, highlighting the current understanding of sarcomeric networks and mitochondrial subpopulations. Next, we discuss acute exercise skeletal muscle metabolism and the signalling, transcriptional and epigenetic regulation of adaptations to exercise training. We address knowledge gaps throughout and propose future directions for the field. This Review contextualizes recent research of skeletal muscle exercise metabolism, framing further advances and translation into practice.
Collapse
Affiliation(s)
- Jonathon A B Smith
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kevin A Murach
- Molecular Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Kenneth A Dyar
- Metabolic Physiology, Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
22
|
Zheng X, Zhang W, Hu Y, Zhao Z, Wu J, Zhang X, Hao F, Han J, Xu J, Hao W, Wang R, Tian M, Radak Z, Nakabeppu Y, Boldogh I, Ba X. DNA repair byproduct 8-oxoguanine base promotes myoblast differentiation. Redox Biol 2023; 61:102634. [PMID: 36827746 PMCID: PMC9982643 DOI: 10.1016/j.redox.2023.102634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Muscle contraction increases the level of reactive oxygen species (ROS), which has been acknowledged as key signaling entities in muscle remodeling and to underlie the healthy adaptation of skeletal muscle. ROS inevitably endows damage to various cellular molecules including DNA. DNA damage ought to be repaired to ensure genome integrity; yet, how DNA repair byproducts affect muscle adaptation remains elusive. Here, we showed that exercise elicited the generation of 8-oxo-7,8-dihydroguanine (8-oxoG), that was primarily found in mitochondrial genome of myofibers. Upon exercise, TA muscle's 8-oxoG excision capacity markedly enhanced, and in the interstitial fluid of TA muscle from the post-exercise mice, the level of free 8-oxoG base was significantly increased. Addition of 8-oxoG to myoblasts triggered myogenic differentiation via activating Ras-MEK-MyoD signal axis. 8-Oxoguanine DNA glycosylase1 (OGG1) silencing from cells or Ogg1 KO from mice decreased Ras activation, ERK phosphorylation, MyoD transcriptional activation, myogenic regulatory factors gene (MRFs) expression. In reconstruction experiments, exogenously added 8-oxoG base enhanced the expression of MRFs and accelerated the recovery of the injured skeletal muscle. Collectively, these data not only suggest that DNA repair metabolite 8-oxoG function as a signal entity for muscle remodeling and contribute to exercise-induced adaptation of skeletal muscle, but also raised the potential for utilizing 8-oxoG in clinical treatment to skeletal muscle damage-related disorders.
Collapse
Affiliation(s)
- Xu Zheng
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Wenhe Zhang
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yinchao Hu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Zhexuan Zhao
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Jiaxin Wu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Xiaoqing Zhang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Fengqi Hao
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Jinling Han
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Jing Xu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Wenjing Hao
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ruoxi Wang
- Institute of Biomedical Sciences, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, Shandong, 250014, China
| | - Meihong Tian
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, H-1123, Budapest, Hungary
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX77555, USA
| | - Xueqing Ba
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, China; School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China.
| |
Collapse
|
23
|
Li B, Wang J, Raza SHA, Wang S, Liang C, Zhang W, Yu S, Shah MA, Al Abdulmonem W, Alharbi YM, Aljohani ASM, Pant SD, Zan L. MAPK family genes' influences on myogenesis in cattle: Genome-wide analysis and identification. Res Vet Sci 2023; 159:198-212. [PMID: 37148739 DOI: 10.1016/j.rvsc.2023.04.024] [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: 12/05/2022] [Revised: 02/11/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
The mitogen-activated protein kinase (MAPK) family is highly conserved in mammals, and is involved in a variety of physiological phenomena like regeneration, development, cell proliferation, and differentiation. In this study, 13 MAPK genes were identified in cattle and their corresponding protein properties were characterized using genome-wide identification and analysis. Phylogenetic analysis showed that the 13 BtMAPKs were cluster grouped into eight major evolutionary branches, which were segmented into three large subfamilies: ERK, p38 and JNK MAPK. BtMAPKs from the same subfamily had similar protein motif compositions, but considerably different exon-intron patterns. The heatmap analysis of transcriptome sequencing data showed that the expression of BtMAPKs was tissue-specific, with BtMAPK6 and BtMAPK12 highly expressed in muscle tissues. Furthermore, knockdown of BtMAPK6 and BtMAPK12 revealed that BtMAPK6 had no effect on myogenic cell proliferation, but negatively affected the differentiation of myogenic cells. In contrast, BtMAPK12 improved both the cell proliferation and differentiation. Taken together, these results provide novel insights into the functions of MAPK families in cattle, which could serve as a basis for further studies on the specific mechanisms of the genes in myogenesis.
Collapse
Affiliation(s)
- Bingzhi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Jianfang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China; Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642 China
| | - Sihu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Chengcheng Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Wenzheng Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Shengchen Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Mujahid Ali Shah
- Faculty of Fisheries and Protection of Water, University of South Bohemia in Ceske Budejovice, Czech Republic
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah 51452, Kingdom of Saudi Arabia
| | - Yousef Mesfer Alharbi
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Sameer D Pant
- Gulbali Institute, Charles Sturt University, Boorooma Street, Wagga Wagga, NSW 2678, Australia
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 Shaanxi, China; National Beef Cattle Improvement Center, Northwest A&F University, Yangling, 712100 Shaanxi, China.
| |
Collapse
|
24
|
Mendes S, Leal DV, Baker LA, Ferreira A, Smith AC, Viana JL. The Potential Modulatory Effects of Exercise on Skeletal Muscle Redox Status in Chronic Kidney Disease. Int J Mol Sci 2023; 24:ijms24076017. [PMID: 37046990 PMCID: PMC10094245 DOI: 10.3390/ijms24076017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic Kidney Disease (CKD) is a global health burden with high mortality and health costs. CKD patients exhibit lower cardiorespiratory and muscular fitness, strongly associated with morbidity/mortality, which is exacerbated when they reach the need for renal replacement therapies (RRT). Muscle wasting in CKD has been associated with an inflammatory/oxidative status affecting the resident cells' microenvironment, decreasing repair capacity and leading to atrophy. Exercise may help counteracting such effects; however, the molecular mechanisms remain uncertain. Thus, trying to pinpoint and understand these mechanisms is of particular interest. This review will start with a general background about myogenesis, followed by an overview of the impact of redox imbalance as a mechanism of muscle wasting in CKD, with focus on the modulatory effect of exercise on the skeletal muscle microenvironment.
Collapse
Affiliation(s)
- Sara Mendes
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia, 4475-690 Maia, Portugal
| | - Diogo V Leal
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia, 4475-690 Maia, Portugal
| | - Luke A Baker
- Leicester Kidney Lifestyle Team, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Aníbal Ferreira
- Nova Medical School, 1169-056 Lisbon, Portugal
- NephroCare Portugal SA, 1750-233 Lisbon, Portugal
| | - Alice C Smith
- Leicester Kidney Lifestyle Team, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - João L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia, 4475-690 Maia, Portugal
| |
Collapse
|
25
|
McIntosh MC, Sexton CL, Godwin JS, Ruple BA, Michel JM, Plotkin DL, Ziegenfuss TN, Lopez HL, Smith R, Dwaraka VB, Sharples AP, Dalbo VJ, Mobley CB, Vann CG, Roberts MD. Different Resistance Exercise Loading Paradigms Similarly Affect Skeletal Muscle Gene Expression Patterns of Myostatin-Related Targets and mTORC1 Signaling Markers. Cells 2023; 12:898. [PMID: 36980239 PMCID: PMC10047349 DOI: 10.3390/cells12060898] [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: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Although transcriptome profiling has been used in several resistance training studies, the associated analytical approaches seldom provide in-depth information on individual genes linked to skeletal muscle hypertrophy. Therefore, a secondary analysis was performed herein on a muscle transcriptomic dataset we previously published involving trained college-aged men (n = 11) performing two resistance exercise bouts in a randomized and crossover fashion. The lower-load bout (30 Fail) consisted of 8 sets of lower body exercises to volitional fatigue using 30% one-repetition maximum (1 RM) loads, whereas the higher-load bout (80 Fail) consisted of the same exercises using 80% 1 RM loads. Vastus lateralis muscle biopsies were collected prior to (PRE), 3 h, and 6 h after each exercise bout, and 58 genes associated with skeletal muscle hypertrophy were manually interrogated from our prior microarray data. Select targets were further interrogated for associated protein expression and phosphorylation induced-signaling events. Although none of the 58 gene targets demonstrated significant bout x time interactions, ~57% (32 genes) showed a significant main effect of time from PRE to 3 h (15↑ and 17↓, p < 0.01), and ~26% (17 genes) showed a significant main effect of time from PRE to 6 h (8↑ and 9↓, p < 0.01). Notably, genes associated with the myostatin (9 genes) and mammalian target of rapamycin complex 1 (mTORC1) (9 genes) signaling pathways were most represented. Compared to mTORC1 signaling mRNAs, more MSTN signaling-related mRNAs (7 of 9) were altered post-exercise, regardless of the bout, and RHEB was the only mTORC1-associated mRNA that was upregulated following exercise. Phosphorylated (phospho-) p70S6K (Thr389) (p = 0.001; PRE to 3 h) and follistatin protein levels (p = 0.021; PRE to 6 h) increased post-exercise, regardless of the bout, whereas phospho-AKT (Thr389), phospho-mTOR (Ser2448), and myostatin protein levels remained unaltered. These data continue to suggest that performing resistance exercise to volitional fatigue, regardless of load selection, elicits similar transient mRNA and signaling responses in skeletal muscle. Moreover, these data provide further evidence that the transcriptional regulation of myostatin signaling is an involved mechanism in response to resistance exercise.
Collapse
Affiliation(s)
| | - Casey L. Sexton
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| | | | | | - J. Max Michel
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| | | | | | | | | | | | - Adam P. Sharples
- Institute for Physical Performance, Norwegian School of Sport Sciences, 0164 Oslo, Norway
| | - Vincent J. Dalbo
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton 4700, Australia
| | | | - Christopher G. Vann
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 03824, USA
| | | |
Collapse
|
26
|
Nicoll JX, Fry AC, Mosier EM. The effects of a caffeine containing pre-workout supplement on β 2-adrenergic and MAPK signaling during resistance exercise. Eur J Appl Physiol 2023; 123:585-599. [PMID: 36383249 DOI: 10.1007/s00421-022-05085-0] [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: 07/18/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022]
Abstract
AIM The acute myocellular responses of caffeine supplementation during resistance exercise (RE) have not been investigated. β2-Adrenergic receptors (β2AR) may be a target of the stimulatory effects of caffeine and stimulate bioenergetic pathways including protein kinase A (PKA), and mitogen-activated protein kinases (MAPK). PURPOSE Elucidate the effects of pre-workout supplementation on signaling responses to an acute RE bout. METHODS In a randomized, counter-balanced, double-blind, placebo-controlled, within-subject crossover study, ten resistance-trained males (mean ± SD; age = 22 ± 2.4 years, height = 175 ± 7 cm, body mass = 84.1 ± 11.8 kg) consumed a caffeine containing multi-ingredient pre-workout supplement (SUPP) or color and flavor matched placebo (PL) 60 min prior to an acute RE bout of barbell back squats. Pre- and post-exercise muscle biopsies were analyzed for the phosphorylation (p-) of β2AR, PKA, and MAPK (ERK, JNK, p38). Epinephrine was determined prior to supplementation (baseline; BL), after supplementation but prior to RE (PRE), and immediately after RE (POST). RESULTS Epinephrine increased at PRE in SUPP (mean ± SE: 323 ± 34 vs 457 ± 68 pmol/l; p = 0.028), and was greatest at POST in the SUPP condition compared to PL (5140 ± 852 vs 2862 ± 498 pmol/l; p = 0.006). p-β2AR and p-MAPK increased post-exercise (p < 0.05) with no differences between conditions (p > 0.05). Pearson correlations indicated there was a relationship between epinephrine and p-β2AR in PL (r = - 0.810; p = 0.008), and p-β2AR and ERK in SUPP (r = 0.941; p < 0.001). CONCLUSION Consumption of a caffeine containing pre-workout supplement improves performance, possibly through increases in pre-exercise catecholamines. However, the acute myocellular signaling responses were largely similar post-exercise.
Collapse
Affiliation(s)
- Justin X Nicoll
- Department of Kinesiology, California State University, Northridge, 18111 Nordhoff St, Northridge, CA, 91330-8287, USA.
| | - Andrew C Fry
- Jayhawk Athletic Performance Laboratory-Wu Tsai Human Performance Alliance, Department of Health, Sport, and Exercise Sciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Eric M Mosier
- Department of Kinesiology, Washburn University, Topeka, KS, 66621, USA
| |
Collapse
|
27
|
Lee CJ, Nicoll JX. Time Course Evaluation of Mitogen-Activated Protein Kinase Phosphorylation to Resistance Exercise: A Systematic Review. J Strength Cond Res 2023; 37:710-725. [PMID: 36727997 DOI: 10.1519/jsc.0000000000004409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
ABSTRACT Lee, CJ and Nicoll, JX. Time course evaluation of mitogen-activated protein kinase phosphorylation to resistance exercise: a systematic review. J Strength Cond Res 37(3): 710-725, 2023-Resistance exercise (RE) can increase the signaling activities of mitogen-activated protein kinases (MAPKs), specifically extracellular signal-regulated kinases 1/2 (ERK1/2), p90 ribosomal S6 kinases (p90RSK), c-Jun NH2-terminal kinases (JNK), and p38-MAPK. These RE-induced responses contribute to various intracellular processes modulating growth and development in skeletal muscles, playing an essential role in resistance training adaptations. The time course of MAPK phosphorylation to different RE conditions, such as training experience and varying loads, remains ambiguous. A systematic review was conducted to determine the effects of different post-RE recovery time points on the MAPK signaling cascade. In addition, the effects of loading and training statuses on MAPK responses were also investigated. The review was performed according to the preferred reporting items for systematic reviews and meta-analyses guidelines with a literature search incorporating 3 electronic databases. A modified version of the Downs and Black checklist was used to evaluate the methodological quality of the studies. The signaling responses were measured within a time range between immediately post-RE and >6 hours post-RE. Forty-four studies met the inclusion criteria, and all were classified as good-to-moderate methodological quality. Mitogen-activated protein kinase phosphorylation increased to different levels after RE, with the highest near the cessation of exercise. Although overall signaling was attenuated among trained individuals likely because of training adaptations, greater MAPK responses can be attributed to moderate loads of 65-85% 1RM regardless of the training experience. However, specific training-induced responses remain equivocal, and further investigations are required to determine the ideal training parameters to optimize anabolic intramuscular signaling, which may likely optimize resistance training adaptations.
Collapse
Affiliation(s)
- Christopher J Lee
- Department of Kinesiology, California State University, Northridge, Northridge, California
| | | |
Collapse
|
28
|
Mechanotransduction for Muscle Protein Synthesis via Mechanically Activated Ion Channels. Life (Basel) 2023; 13:life13020341. [PMID: 36836698 PMCID: PMC9962945 DOI: 10.3390/life13020341] [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: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Cell mechanotransduction, the ability to detect physical forces and convert them into a series of biochemical events, is important for a wide range of physiological processes. Cells express an array of mechanosensors transducing physical forces into intracellular signaling cascades, including ion channels. Ion channels that can be directly activated by mechanical cues are known as mechanically activated (MA), or stretch-activated (SA), channels. In response to repeated exposures to mechanical stimulation in the form of resistance training, enhanced protein synthesis and fiber hypertrophy are elicited in skeletal muscle, whereas a lack of mechanical stimuli due to inactivity/mechanical unloading leads to reduced muscle protein synthesis and fiber atrophy. To date, the role of MA channels in the transduction of mechanical load to intracellular signaling pathways regulating muscle protein synthesis is poorly described. This review article will discuss MA channels in striated muscle, their regulation, and putative roles in the anabolic processes in muscle cells/fibers in response to mechanical stimuli.
Collapse
|
29
|
Abstract
The HER2+ subtype of human breast cancer is associated with the malignant transformation of luminal ductal cells of the mammary epithelium. The sequence analysis of tumor DNA identifies loss of function mutations and deletions of the MAP2K4 and MAP2K7 genes that encode direct activators of the JUN NH2-terminal kinase (JNK). We report that in vitro studies of human mammary epithelial cells with CRISPR-induced mutations in the MAPK and MAP2K components of the JNK pathway caused no change in growth in 2D culture, but these mutations promoted epithelial cell proliferation in 3D culture. Analysis of gene expression signatures in 3D culture demonstrated similar changes caused by HER2 activation and JNK pathway loss. The mechanism of signal transduction cross-talk may be mediated, in part, by JNK-suppressed expression of integrin α6β4 that binds HER2 and amplifies HER2 signaling. These data suggest that HER2 activation and JNK pathway loss may synergize to promote breast cancer. To test this hypothesis, we performed in vivo studies using a mouse model of HER2+ breast cancer with Cre/loxP-mediated ablation of genes encoding JNK (Mapk8 and Mapk9) and the MAP2K (Map2k4 and Map2k7) that activate JNK in mammary epithelial cells. Kaplan-Meier analysis of tumor development demonstrated that JNK pathway deficiency promotes HER2+-driven breast cancer. Collectively, these data identify JNK pathway genes as potential suppressors for HER2+ breast cancer.
Collapse
|
30
|
Myostatin Mutation Enhances Bovine Myogenic Differentiation through PI3K/AKT/mTOR Signalling via Removing DNA Methylation of RACK1. Cells 2022; 12:cells12010059. [PMID: 36611855 PMCID: PMC9818849 DOI: 10.3390/cells12010059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/26/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Myostatin (MSTN) is a negative regulator of skeletal muscle development and plays an important role in muscle development. Fluctuations in gene expression influenced by DNA methylation are critical for homeostatic responses in muscle. However, little is known about the mechanisms underlying this fluctuation regulation and myogenic differentiation of skeletal muscle. Here we report a genome-wide analysis of DNA methylation dynamics in bovine skeletal muscle myogenesis after myostatin editing. We show that, after myostatin editing, an increase in TETs (DNA demethylases) and a concomitant increase in the receptor for activated C kinase 1 (RACK1) control the myogenic development of skeletal muscle. Interestingly, enhancement of PI3K/AKT/mTOR signaling by RACK1 appears to be an essential driver of myogenic differentiation, as it was associated with an increase in myogenic differentiation marker factors (MyHC and MyoG) during muscle differentiation. Overall, our results suggest that loss of myostatin promotes the myogenic differentiation response in skeletal muscle by decreasing DNA methylation of RACK1.
Collapse
|
31
|
Willoughby DS, Cardaci TD, Machek SB, Wilburn DT, Heileson JL. Resistance Exercise-Induced Increases in Muscle Myostatin mRNA and Protein Expression Are Subsequently Decreased in Circulation in the Presence of Increased Levels of the Extracellular Matrix Stabilizing Protein Decorin. J Sports Sci Med 2022; 21:616-624. [PMID: 36523894 PMCID: PMC9741719 DOI: 10.52082/jssm.2022.616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Resistance exercise (RE) activates cell signaling pathways associated with myostatin. Decorin is located in the extracellular matrix (ECM) and can block the inhibitory effect of myostatin. This study sought to determine the impact of low-load (LL) and high-load (HL) RE on myostatin mRNA and protein expression along with changes in muscle decorin and circulating follistatin. Ten resistance-trained men performed a LL (50% 1RM) and HL (80% 1RM) RE session using the angled leg press and leg extension with load and volume equated. Venous blood samples and muscle biopsies were obtained prior to and at 3h and 24h following each RE session. Muscle myostatin mRNA expression was increased at 24h post-exercise (p = 0.032) in LL and at 3h (p = 0.044) and 24h (p = 0.003) post-exercise in HL. Muscle decorin was increased at 24h post-exercise (p < 0.001) in LL and HL; however, muscle myostatin was increased at 24h post-exercise (p < 0.001) only in HL. For muscle Smad 2/3, no significant differences were observed (p > 0.05). Serum follistatin was increased and myostatin decreased at 24h post-exercise (p < 0.001) in LL and HL. Muscle myostatin gene and protein expression increased in response to HL RE. However, serum myostatin was decreased in the presence of increases in decorin in muscle and follistatin in circulation. Therefore, our data suggest a possible mechanism may exist where decorin within the ECM is able to bind to, and decrease, myostatin that might otherwise enter the circulation for activin IIB (ACTIIB) receptor binding and subsequent canonical signaling through Smad 2/3.
Collapse
Affiliation(s)
- Darryn S. Willoughby
- School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA, Department of Health and Human Performance, Baylor University, Waco, TX, USA, School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Thomas D. Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Steven B. Machek
- Department of Kinesiology, California State University Monterey Bay, Seaside, CA, USA
| | - Dylan T. Wilburn
- Department of Health and Human Performance, Baylor University, Waco, TX, USA
| | - Jeffery L. Heileson
- Nutrition Services Division, Walter Reed National Military Medical Center, Bethesda, MD, USA
| |
Collapse
|
32
|
Barros D, Marques EA, Magalhães J, Carvalho J. Energy metabolism and frailty: The potential role of exercise-induced myokines - A narrative review. Ageing Res Rev 2022; 82:101780. [PMID: 36334911 DOI: 10.1016/j.arr.2022.101780] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Frailty is a complex condition that emerges from dysregulation in multiple physiological systems. Increasing evidence suggests the potential role of age-related energy dysregulation as a key driver of frailty. Exercise is considered the most efficacious intervention to prevent and even ameliorate frailty as it up-tunes and improves the function of several related systems. However, the mechanisms and molecules responsible for these intersystem benefits are not fully understood. The skeletal muscle is considered a secretory organ with endocrine functions that can produce and secrete exercise-related molecules such as myokines. These molecules are cytokines and other peptides released by muscle fibers in response to acute and/or chronic exercise. The available evidence supports that several myokines can elicit autocrine, paracrine, or endocrine effects, partly mediating inter-organ crosstalk and also having a critical role in improving cardiovascular, metabolic, immune, and neurological health. This review describes the current evidence about the potential link between energy metabolism dysregulation and frailty and provides a theoretical framework for the potential role of myokines (via exercise) in counteracting frailty. It also summarizes the physiological role of selected myokines and their response to different acute and chronic exercise protocols in older adults.
Collapse
Affiliation(s)
- Duarte Barros
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Elisa A Marques
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia (ISMAI), Portugal; School of Sport and Exercise Sciences, Loughborough University, Loughborough, UK
| | - José Magalhães
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Joana Carvalho
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| |
Collapse
|
33
|
Ahsan M, Garneau L, Aguer C. The bidirectional relationship between AMPK pathway activation and myokine secretion in skeletal muscle: How it affects energy metabolism. Front Physiol 2022; 13:1040809. [PMID: 36479347 PMCID: PMC9721351 DOI: 10.3389/fphys.2022.1040809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2023] Open
Abstract
Myokines are peptides and proteins secreted by skeletal muscle cells, into the interstitium, or in the blood. Their regulation may be dependent or independent of muscle contraction to induce a variety of metabolic effects. Numerous myokines have been implicated in influencing energy metabolism via AMP-activated protein kinase (AMPK) signalling. As AMPK is centrally involved in glucose and lipid metabolism, it is important to understand how myokines influence its signalling, and vice versa. Such insight will better elucidate the mechanism of metabolic regulation during exercise and at rest. This review encompasses the latest research conducted on the relationship between AMPK signalling and myokines within skeletal muscles via autocrine or paracrine signalling.
Collapse
Affiliation(s)
- Mahdi Ahsan
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Léa Garneau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort –Recherche, Ottawa, ON, Canada
| | - Céline Aguer
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort –Recherche, Ottawa, ON, Canada
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University—Campus Outaouais, Gatineau, QC, Canada
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
34
|
Mowry KC, Thomson-Parker TL, Morales C, Fikes KK, Stutts KJ, Leatherwood JL, Anderson MJ, Smith RX, Suagee-Bedore JK. Effects of Crude Rice Bran Oil and a Flaxseed Oil Blend in Young Horses Engaged in a Training Program. Animals (Basel) 2022; 12:ani12213006. [PMID: 36359130 PMCID: PMC9653641 DOI: 10.3390/ani12213006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Rice bran oil and flaxseed oil contain omega-3 fatty acids with the potential to reduce post-exercise inflammation and muscle damage. This study measures plasma interleukin-1β and creatine kinase and fatty acid profiles in lightly worked, young horses (Equus caballus) undergoing an exercise test after 60 days (d) of oil consumption, where the oil replaced 25% of concentrate calories. Treatments consisted of CON (no oil), FLAX (flaxseed oil blend), and RICE (crude rice bran oil). Blood was collected pre-exercise, and again at 1 min, 30 min, 24 h, 48 h, and 72 h post-IET. Data were analyzed by repeated measures ANOVA. Plasma creatine kinase activity was not different in CON during the study, greater (p < 0.05) in RICE from pre-exercise to 30 min post-exercise across all exercise tests, and lesser (p < 0.05) in FLAX at 30 min post-exercise on d 30 compared to d 0. Plasma interleukin-1β was greater (p < 0.01) in CON on d 60, but no differences were observed in FLAX and RICE throughout the study. Plasma alpha-linolenic and linoleic acids were greatest (p < 0.05) in FLAX after 30 d of inclusion, while CON horses had greater (p < 0.05) EPA across all exercise tests and DHA after 60 d. These results indicate that 60 d of inclusion of crude rice bran oil or a flaxseed oil blend may benefit lightly worked, young horses by reducing training-program-related increases in interleukin-1β, while a flaxseed oil blend may reduce exercise-induced increases in creatine kinase. Additionally, the flaxseed oil blend has the potential to increase plasma omega-3 and omega-6 fatty acids. Replacing 25% of concentrate calories with flaxseed or rice bran oil has potential benefits for young horses in training.
Collapse
Affiliation(s)
- Kayla C. Mowry
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Timber L. Thomson-Parker
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Cruz Morales
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Kalley K. Fikes
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Kyle J. Stutts
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Jessica L. Leatherwood
- Department of Animal Science, College of Agriculture and Life Sciences, Texas A&M University, College State, TX 77843, USA
| | - Mark J. Anderson
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
| | - Rachelle X. Smith
- Sam Houston State University Analytical Laboratory, Sam Houston State University, Huntsville, TX 77341, USA
| | - Jessica K. Suagee-Bedore
- School of Agricultural Sciences, College of Science and Engineering Technology, Sam Houston State University, Huntsville, TX 77340, USA
- Correspondence: ; Tel.: +1-540-231-2232
| |
Collapse
|
35
|
Brennan CM, Hill AS, St. Andre M, Li X, Madeti V, Breitkopf S, Garren S, Xue L, Gilbert T, Hadjipanayis A, Monetti M, Emerson CP, Moccia R, Owens J, Christoforou N. DUX4 expression activates JNK and p38 MAP kinases in myoblasts. Dis Model Mech 2022; 15:dmm049516. [PMID: 36196640 PMCID: PMC10655719 DOI: 10.1242/dmm.049516] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by misexpression of the DUX4 transcription factor in skeletal muscle that results in transcriptional alterations, abnormal phenotypes and cell death. To gain insight into the kinetics of DUX4-induced stresses, we activated DUX4 expression in myoblasts and performed longitudinal RNA sequencing paired with proteomics and phosphoproteomics. This analysis revealed changes in cellular physiology upon DUX4 activation, including DNA damage and altered mRNA splicing. Phosphoproteomic analysis uncovered rapid widespread changes in protein phosphorylation following DUX4 induction, indicating that alterations in kinase signaling might play a role in DUX4-mediated stress and cell death. Indeed, we demonstrate that two stress-responsive MAP kinase pathways, JNK and p38, are activated in response to DUX4 expression. Inhibition of each of these pathways ameliorated DUX4-mediated cell death in myoblasts. These findings uncover that the JNK pathway is involved in DUX4-mediated cell death and provide additional insights into the role of the p38 pathway, a clinical target for the treatment of FSHD.
Collapse
Affiliation(s)
- Christopher M. Brennan
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
- WRDM Postdoctoral Program, Pfizer Inc., Cambridge, MA 02139, USA
| | - Abby S. Hill
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
| | | | - Xianfeng Li
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
| | - Vijaya Madeti
- NGS Technology Center, Inflammation and Immunology Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Susanne Breitkopf
- Proteomics Technology Center, Internal Medicine Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Seth Garren
- NGS Technology Center, Inflammation and Immunology Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Liang Xue
- Machine Learning and Computational Science, Pfizer Inc., Cambridge, MA 02139, USA
| | - Tamara Gilbert
- High Content Imaging Technology Center, Internal Medicine Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Angela Hadjipanayis
- NGS Technology Center, Inflammation and Immunology Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Mara Monetti
- Proteomics Technology Center, Internal Medicine Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Charles P. Emerson
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert Moccia
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
| | - Jane Owens
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA 02139, USA
| | | |
Collapse
|
36
|
Nordgaard C, Vind AC, Stonadge A, Kjøbsted R, Snieckute G, Antas P, Blasius M, Reinert MS, Del Val AM, Bekker-Jensen DB, Haahr P, Miroshnikova YA, Mazouzi A, Falk S, Perrier-Groult E, Tiedje C, Li X, Jakobsen JR, Jørgensen NO, Wojtaszewski JF, Mallein-Gerin F, Andersen JL, Pennisi CP, Clemmensen C, Kassem M, Jafari A, Brummelkamp T, Li VS, Wickström SA, Olsen JV, Blanco G, Bekker-Jensen S. ZAKβ is activated by cellular compression and mediates contraction-induced MAP kinase signaling in skeletal muscle. EMBO J 2022; 41:e111650. [PMID: 35899396 PMCID: PMC9434084 DOI: 10.15252/embj.2022111650] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 06/22/2022] [Indexed: 12/31/2022] Open
Abstract
Mechanical inputs give rise to p38 and JNK activation, which mediate adaptive physiological responses in various tissues. In skeletal muscle, contraction‐induced p38 and JNK signaling ensure adaptation to exercise, muscle repair, and hypertrophy. However, the mechanisms by which muscle fibers sense mechanical load to activate this signaling have remained elusive. Here, we show that the upstream MAP3K ZAKβ is activated by cellular compression induced by osmotic shock and cyclic compression in vitro, and muscle contraction in vivo. This function relies on ZAKβ's ability to recognize stress fibers in cells and Z‐discs in muscle fibers when mechanically perturbed. Consequently, ZAK‐deficient mice present with skeletal muscle defects characterized by fibers with centralized nuclei and progressive adaptation towards a slower myosin profile. Our results highlight how cells in general respond to mechanical compressive load and how mechanical forces generated during muscle contraction are translated into MAP kinase signaling.
Collapse
Affiliation(s)
- Cathrine Nordgaard
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anna Constance Vind
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Amy Stonadge
- Department of Biology, University of York, York, UK
| | - Rasmus Kjøbsted
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Goda Snieckute
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Pedro Antas
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Melanie Blasius
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Marie Sofie Reinert
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ana Martinez Del Val
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, Faculty of Health and Medical Sciences, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Dorte Breinholdt Bekker-Jensen
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, Faculty of Health and Medical Sciences, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Peter Haahr
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yekaterina A Miroshnikova
- Stem Cells and Metabolism Research Program, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Abdelghani Mazouzi
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sarah Falk
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Christopher Tiedje
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Xiang Li
- Department of Biology, University of York, York, UK
| | - Jens Rithamer Jakobsen
- Department of Orthopedic Surgery M, Institute of Sports Medicine Copenhagen, Bispebjerg Hospital, Copenhagen, Denmark
| | | | - Jørgen Fp Wojtaszewski
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | | | - Jesper Løvind Andersen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Orthopedic Surgery M, Institute of Sports Medicine Copenhagen, Bispebjerg Hospital, Copenhagen, Denmark
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology and Metabolism, University Hospital of Odense and University of Southern Denmark, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Thijn Brummelkamp
- Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Cancer Genomics Center, Amsterdam, The Netherlands
| | - Vivian Sw Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, UK
| | - Sara A Wickström
- Stem Cells and Metabolism Research Program, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jesper Velgaard Olsen
- Mass Spectrometry for Quantitative Proteomics, Proteomics Program, Faculty of Health and Medical Sciences, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
MacDonald TL, Pattamaprapanont P, Cooney EM, Nava RC, Mitri J, Hafida S, Lessard SJ. Canagliflozin Prevents Hyperglycemia-Associated Muscle Extracellular Matrix Accumulation and Improves the Adaptive Response to Aerobic Exercise. Diabetes 2022; 71:881-893. [PMID: 35108373 PMCID: PMC9044131 DOI: 10.2337/db21-0934] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/27/2022] [Indexed: 02/03/2023]
Abstract
Chronic hyperglycemia is associated with low response to aerobic exercise training in rodent models and humans, including reduced aerobic exercise capacity and impaired oxidative remodeling in skeletal muscle. Here, we investigated whether glucose lowering with the sodium-glucose cotransporter 2 inhibitor (SGLT2i), canagliflozin (Cana; 30 mg/kg/day), could restore exercise training response in a model of hyperglycemia (low-dose streptozotocin [STZ]). Cana effectively prevented increased blood glucose in STZ-treated mice. After 6 weeks of voluntary wheel running, Cana-treated mice displayed improvements in aerobic exercise capacity, higher capillary density in striated muscle, and a more oxidative fiber-type in skeletal muscle. In contrast, these responses were blunted or absent in STZ-treated mice. Recent work implicates glucose-induced accumulation of skeletal muscle extracellular matrix (ECM) and hyperactivation of c-Jun N-terminal kinase (JNK)/SMAD2 mechanical signaling as potential mechanisms underlying poor exercise response. In line with this, muscle ECM accretion was prevented by Cana in STZ-treated mice. JNK/SMAD2 signaling with acute exercise was twofold higher in STZ compared with control but was normalized by Cana. In human participants, ECM accumulation was associated with increased JNK signaling, low VO2peak, and impaired metabolic health (oral glucose tolerance test-derived insulin sensitivity). These data demonstrate that hyperglycemia-associated impairments in exercise adaptation can be ameliorated by cotherapy with SGLT2i.
Collapse
Affiliation(s)
- Tara L. MacDonald
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | | | | | - Roberto C. Nava
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Joanna Mitri
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Samar Hafida
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Sarah J. Lessard
- Research Division, Joslin Diabetes Center, Boston, MA
- Harvard Medical School, Boston, MA
- Corresponding author: Sarah J. Lessard,
| |
Collapse
|
38
|
Lu J, Yu L, Shi J. Low-dose paclitaxel modulates the cross talk between the JNK and Smad signaling in primary biliary fibroblasts. Rev Assoc Med Bras (1992) 2022; 68:159-164. [PMID: 35239875 DOI: 10.1590/1806-9282.20210777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE The objective of this study was to explore the molecular mechanism underlying the occurrence of benign bile duct stricture and the target of low-dose paclitaxel in the prevention of benign bile duct stricture. METHODS Under the stimulation of transforming growth factor beta 1, the expression of collagen type I and connective tissue growth factor were detected on isolated primary fibroblasts. The phosphorylation levels of JNK and Smad2L were detected using Western blot. The effect of low-dose paclitaxel on the transforming growth factor beta 1-induced inhibition of type I collagen and connective tissue growth factor expression and JNK and Smad2L phosphorylation was also observed. RESULTS Transforming growth factor beta 1 induced the secretion of type I collagen and connective tissue growth factor as well as JNK phosphorylation in biliary fibroblasts. The JNK inhibitor or siRNA-Smad2 inhibited the transforming growth factor beta 1-induced secretion of type I collagen and connective tissue growth factor. Low-dose paclitaxel inhibited the expression of type I collagen induced by transforming growth factor beta 1 and may inhibit the secretion of collagen in biliary fibroblasts. CONCLUSION The activation of JNK/Smad2L induced by transforming growth factor beta 1 is involved in the occurrence of benign bile duct stricture that is mediated by the overexpression of type I collagen and connective tissue growth factor, and low-dose paclitaxel may inhibit the phosphorylation of JNK/Smad2L.
Collapse
Affiliation(s)
- Jiamei Lu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Department of Nephrology - Xi'an, China
| | - Liang Yu
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Hepatobiliary Surgery - Xi'an, China
| | - Jianhua Shi
- The First Affiliated Hospital of Xi'an Jiaotong University, Department of Hepatobiliary Surgery - Xi'an, China
| |
Collapse
|
39
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
40
|
Alizadeh Pahlavani H. Exercise Therapy for People With Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front Endocrinol (Lausanne) 2022; 13:811751. [PMID: 35250869 PMCID: PMC8892203 DOI: 10.3389/fendo.2022.811751] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenic obesity is defined as a multifactorial disease in aging with decreased body muscle, decreased muscle strength, decreased independence, increased fat mass, due to decreased physical activity, changes in adipokines and myokines, and decreased satellite cells. People with sarcopenic obesity cause harmful changes in myokines and adipokines. These changes are due to a decrease interleukin-10 (IL-10), interleukin-15 (IL-15), insulin-like growth factor hormone (IGF-1), irisin, leukemia inhibitory factor (LIF), fibroblast growth factor-21 (FGF-21), adiponectin, and apelin. While factors such as myostatin, leptin, interleukin-6 (IL-6), interleukin-8 (IL-8), and resistin increase. The consequences of these changes are an increase in inflammatory factors, increased degradation of muscle proteins, increased fat mass, and decreased muscle tissue, which exacerbates sarcopenia obesity. In contrast, exercise, especially strength training, reverses this process, which includes increasing muscle protein synthesis, increasing myogenesis, increasing mitochondrial biogenesis, increasing brown fat, reducing white fat, reducing inflammatory factors, and reducing muscle atrophy. Since some people with chronic diseases are not able to do high-intensity strength training, exercises with blood flow restriction (BFR) are newly recommended. Numerous studies have shown that low-intensity BFR training produces the same increase in hypertrophy and muscle strength such as high-intensity strength training. Therefore, it seems that exercise interventions with BFR can be an effective way to prevent the exacerbation of sarcopenia obesity. However, due to limited studies on adipokines and exercises with BFR in people with sarcopenic obesity, more research is needed.
Collapse
|
41
|
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
|
42
|
Tan Z, Zhao M, Li J, Li S, Zhu S, Yao X, Gao X, Yang S. Myostatin is involved in skeletal muscle dysfunction in chronic obstructive pulmonary disease via Drp-1 mediated abnormal mitochondrial division. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:162. [PMID: 35280400 PMCID: PMC8908114 DOI: 10.21037/atm-22-377] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/18/2022] [Indexed: 11/06/2022]
Abstract
Background Skeletal muscle dysfunction (SMD) is one of the most prominent extrapulmonary effects of chronic obstructive pulmonary disease (COPD). Myostatin negatively regulates the growth of skeletal muscle. We confirmed that myostatin expression is significantly increased in the quadriceps femoris muscle tissue of rats with COPD and is involved in the development of SMD in COPD, but the mechanism by which this occurs has yet to be uncovered. Dynamin-related protein 1 (Drp-1) has been shown to promote apoptosis and affect cellular energy metabolism by mediating enhanced mitochondrial division. Preliminary findings from our group illustrated that mitochondrial division and Drp-1 expression were increased in COPD quadriceps femoris cells. However, it is not yet clear whether mitochondrial dynamics are affected by myostatin in COPD quadriceps myocytes. Methods The study sought to explore the effects and potential mechanisms of myostatin on skeletal muscle atrophy, mitochondrial dynamics, apoptosis, and the links between related processes in COPD. Results Our findings showed that cigarette smoke exposure stimulated an increase in myostatin, increased superoxide production, decreased mitochondrial membrane potential, significantly promoted Drp-1-mediated mitochondrial fission, and promoted apoptosis. Conclusions In summary, our study demonstrated that cigarette smoke led to increased Drp-1 expression and enhanced mitochondrial division by upregulating myostatin, which in turn promoted apoptosis and affected cellular energy metabolism, leading to the development of SMD in COPD. This study extends understandings of skeletal muscle function in COPD and provides a basis for the use of myostatin and Drp-1 as novel therapeutic targets for SMD in COPD.
Collapse
Affiliation(s)
- Zhidan Tan
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Manzhi Zhao
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Siqi Li
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Su Zhu
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Xiaoxuan Yao
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Xinglin Gao
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| | - Shifang Yang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, Guangzhou, China
| |
Collapse
|
43
|
Teixeira da Silva R, Machado IF, Teodoro JS, Panisello-Roselló A, Roselló-Catafau J, Rolo AP, Palmeira CM. PEG35 as a Preconditioning Agent against Hypoxia/Reoxygenation Injury. Int J Mol Sci 2022; 23:1156. [PMID: 35163080 PMCID: PMC8834864 DOI: 10.3390/ijms23031156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/22/2022] Open
Abstract
Pharmacological conditioning is a protective strategy against ischemia/reperfusion injury, which occurs during liver resection and transplantation. Polyethylene glycols have shown multiple benefits in cell and organ preservation, including antioxidant capacity, edema prevention and membrane stabilization. Recently, polyethylene glycol 35 kDa (PEG35) preconditioning resulted in decreased hepatic injury and protected the mitochondria in a rat model of cold ischemia. Thus, the study aimed to decipher the mechanisms underlying PEG35 preconditioning-induced protection against ischemia/reperfusion injury. A hypoxia/reoxygenation model using HepG2 cells was established to evaluate the effects of PEG35 preconditioning. Several parameters were assessed, including cell viability, mitochondrial membrane potential, ROS production, ATP levels, protein content and gene expression to investigate autophagy, mitochondrial biogenesis and dynamics. PEG35 preconditioning preserved the mitochondrial function by decreasing the excessive production of ROS and subsequent ATP depletion, as well as by recovering the membrane potential. Furthermore, PEG35 increased levels of autophagy-related proteins and the expression of genes involved in mitochondrial biogenesis and fusion. In conclusion, PEG35 preconditioning effectively ameliorates hepatic hypoxia/reoxygenation injury through the enhancement of autophagy and mitochondrial quality control. Therefore, PEG35 could be useful as a potential pharmacological tool for attenuating hepatic ischemia/reperfusion injury in clinical practice.
Collapse
Affiliation(s)
- Rui Teixeira da Silva
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- Experimental Pathology Department, Institute of Biomedical Research of Barcelona (IIBB), CSIC-IDIBAPS, 08036 Barcelona, Spain; (A.P.-R.); (J.R.-C.)
| | - Ivo F. Machado
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- IIIUC—Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - João S. Teodoro
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
| | - Arnau Panisello-Roselló
- Experimental Pathology Department, Institute of Biomedical Research of Barcelona (IIBB), CSIC-IDIBAPS, 08036 Barcelona, Spain; (A.P.-R.); (J.R.-C.)
| | - Joan Roselló-Catafau
- Experimental Pathology Department, Institute of Biomedical Research of Barcelona (IIBB), CSIC-IDIBAPS, 08036 Barcelona, Spain; (A.P.-R.); (J.R.-C.)
| | - Anabela P. Rolo
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
| | - Carlos M. Palmeira
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal; (J.S.T.); (A.P.R.); (C.M.P.)
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
| |
Collapse
|
44
|
Tan Z, Li S, Zhu S, Yao X, Li J, Gao X, Yang S. Effect of cigarette smoke extract on mitochondrial division in mouse quadriceps femoris cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1699. [PMID: 34988208 PMCID: PMC8667143 DOI: 10.21037/atm-21-5891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND To observe the effect of cigarette smoke extract (CSE) on mitochondrial division in mouse quadriceps femoris cells and to explore the potential molecular mechanism of skeletal muscle dysfunction (SMD) in patients with chronic obstructive pulmonary disease (COPD). METHODS Quadriceps femoris were cultured, passaged, and stimulated with different concentrations of CSE. We divided cells into four groups (Control, 2.5%, 5%, 10%). The growth of cells, the expression of Dynamin related protein 1 (Drp-1), and apoptosis were observed and evaluated by fluorescence microscopy, RT-PCR, Western blot, and flow cytometry. RESULTS The longer the intervention time, the more obvious the decrease in cell number. In the 5% and 10% groups, the cells became round with gaps. Under an inverted fluorescence microscope, the green fluorescence of cells in 5% and 10% stained with Mito-Tracker Green was significantly less than that of the Control and 2.5%. Red fluorescence was reduced and the green fluorescence was increased in the 5% and 10% stained with JC-1. Flow cytometry analysis showed that reactive oxygen species (ROS) and apoptosis were increased in the CSE intervention groups. In the Control, 2.5%, 5%, and 10%, the levels of ROS were 0.052±0.015, 0.170±0.030, 5.340±0.500, and 24.400±1.900, respectively. The apoptotic rates (%) were 0.270±0.009, 2.650±0.060, 11.850±0.020, and 31.820±1.260, respectively. The relative expression levels were, 0.900±0.093, 1.141±0.099, 1.361±0.034, 2.155±0.092 for DNM1L mRNA, and 0.509±0.008, 0.569±0.028, 0.792±0.048, 0.940±0.062 for Drp-1. There were significant differences in the apoptotic rate, and Drp-1 expression between 5% and 10% compared with the Control and 2.5% (P<0.05). CONCLUSIONS CSE may enhance mitochondrial division of quadriceps femoris cells by up-regulating the expression of Drp-1, affecting cellular energy metabolism and promoting quadriceps femoris apoptosis, ultimately leading to the occurrence and development of skeletal muscle dysfunction in COPD.
Collapse
Affiliation(s)
- Zhidan Tan
- The Second School of Clinical Medicine, Southern Medical University, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Siqi Li
- The Second School of Clinical Medicine, Southern Medical University, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Su Zhu
- Shantou University Medical College, Shantou, China
| | - Xiaoxuan Yao
- Shantou University Medical College, Shantou, China
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xinglin Gao
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shifang Yang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Geriatrics Institute, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Chenodeoxycholic Acid Has Non-Thermogenic, Mitodynamic Anti-Obesity Effects in an In Vitro CRISPR/Cas9 Model of Bile Acid Receptor TGR5 Knockdown. Int J Mol Sci 2021; 22:ijms222111738. [PMID: 34769169 PMCID: PMC8584144 DOI: 10.3390/ijms222111738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 01/14/2023] Open
Abstract
Bile acids (BA) have shown promising effects in animal models of obesity. However, the said effects are thought to rely on a thermogenic effect, which is questionably present in humans. A previous work has shown that the BA chenodeoxycholic acid (CDCA) can revert obesity and accelerate metabolism in animal and cell culture models. Thus, the aim of this study was to understand if this obesity reduction is indeed thermogenically-dependent. A CRISPR/Cas9 model of TGR5 (BA receptor) knockdown in 3T3-L1 adipocytes was developed to diminish thermogenic effects. Various parameters were assessed, including mitochondrial bioenergetics by Seahorse flux analysis, oxidative stress and membrane potential by fluorometry, intermediary metabolism by NMR, protein content assessment by Western Blot, gene expression by qPCR, and confocal microscopy evaluation of mitophagy. CDCA was still capable, for the most part, of reversing the harmful effects of cellular obesity, elevating mitophagy and leading to the reduction of harmed mitochondria within the cells, boosting mitochondrial activity, and thus energy consumption. In summary, CDCA has a non-thermogenic, obesity reducing capacity that hinges on a healthy mitochondrial population, explaining at least some of these effects and opening avenues of human treatment for metabolic diseases.
Collapse
|
46
|
Weng K, Huo W, Li Y, Zhang Y, Zhang Y, Chen G, Xu Q. Fiber characteristics and meat quality of different muscular tissues from slow- and fast-growing broilers. Poult Sci 2021; 101:101537. [PMID: 34788716 PMCID: PMC8591497 DOI: 10.1016/j.psj.2021.101537] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022] Open
Abstract
Chicken meat is an important source of high-quality animal protein. Its consumption continues to grow in both developed and developing countries. Muscle fiber characteristics are key determinants of meat quality and quantity. Skeletal muscle is a highly plastic tissue that is affected by breed differences and muscular tissues. However, studies regarding the effects of different breeds and muscular tissues on the fibers and meat quality traits in broilers are lacking. In this study, Ross 308 chickens (fast-growing [FG] broilers) and Xueshan chickens (slow-growing [SG] broilers) were selected, and their fiber and meat quality traits were characterized. The results showed that the breast muscle primarily comprised glycolytic fibers, whereas the leg muscle comprised glycolytic and a few oxidative fibers, regardless of the breed. The highest percentage of oxidative fibers (26.51%) appeared in the soleus muscle (SOL) of SG broilers. In addition, higher shear force, lower pressing loss, and thicker muscle fibers with less extracellular space were observed for SG meat than for FG meat. When comparing the different muscular tissues, a higher oxidative fiber percentage, ultimate pH, redness, and intramuscular fat (IMF) content were detected in the leg muscle than in the breast muscle in the 2 breeds. In summary, these data indicated that SG broilers had thicker muscle fibers than the FG broilers and that the leg muscle had more oxidative fibers than the breast muscle. Thicker fibers may contribute to increased firmness and more oxidative fibers lead to higher redness value and IMF content.
Collapse
Affiliation(s)
- Kaiqi Weng
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weiran Huo
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yi Li
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yu Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guohong Chen
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qi Xu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
47
|
Dai C, Reyes-Ordoñez A, You JS, Chen J. A non-translational role of threonyl-tRNA synthetase in regulating JNK signaling during myogenic differentiation. FASEB J 2021; 35:e21948. [PMID: 34569098 DOI: 10.1096/fj.202101094r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 11/11/2022]
Abstract
Aminoacyl-tRNA synthetases (aaRSs) are house-keeping enzymes that are essential for protein synthesis. However, it has become increasingly evident that some aaRSs also have non-translational functions. Here we report the identification of a non-translational function of threonyl-tRNA synthetase (ThrRS) in myogenic differentiation. We find that ThrRS negatively regulates myoblast differentiation in vitro and injury-induced skeletal muscle regeneration in vivo. This function is independent of amino acid binding or aminoacylation activity of ThrRS, and knockdown of ThrRS leads to enhanced differentiation without affecting the global protein synthesis rate. Furthermore, we show that the non-catalytic new domains (UNE-T and TGS) of ThrRS are both necessary and sufficient for the myogenic function. In searching for a molecular mechanism of this new function, we find the kinase JNK to be a downstream target of ThrRS. Our data further reveal MEKK4 and MKK4 as upstream regulators of JNK in myogenesis and the MEKK4-MKK4-JNK pathway to be a mediator of the myogenic function of ThrRS. Finally, we show that ThrRS physically interacts with Axin1, disrupts Axin1-MEKK4 interaction and consequently inhibits JNK signaling. In conclusion, we uncover a non-translational function for ThrRS in the maintenance of homeostasis of skeletal myogenesis and identify the Axin1-MEKK4-MKK4-JNK signaling axis to be an immediate target of ThrRS action.
Collapse
Affiliation(s)
- Chong Dai
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Adriana Reyes-Ordoñez
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jae-Sung You
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jie Chen
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
48
|
Eftestøl E, Franchi MV, Kasper S, Flück M. JNK activation in TA and EDL muscle is load-dependent in rats receiving identical excitation patterns. Sci Rep 2021; 11:16405. [PMID: 34385505 PMCID: PMC8361015 DOI: 10.1038/s41598-021-94930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/12/2021] [Indexed: 11/09/2022] Open
Abstract
As the excitation-contraction coupling is inseparable during voluntary exercise, the relative contribution of the mechanical and neural input on hypertrophy-related molecular signalling is still poorly understood. Herein, we use a rat in-vivo strength exercise model with an electrically-induced standardized excitation pattern, previously shown to induce a load-dependent increase in myonuclear number and hypertrophy, to study acute effects of load on molecular signalling. We assessed protein abundance and specific phosphorylation of the four protein kinases FAK, mTOR, p70S6K and JNK after 2, 10 and 28 min of a low- or high-load contraction, in order to assess the effects of load, exercise duration and muscle-type on their response to exercise. Specific phosphorylation of mTOR, p70S6K and JNK was increased after 28 min of exercise under the low- and high-load protocol. Elevated phosphorylation of mTOR and JNK was detectable already after 2 and 10 min of exercise, respectively, but greatest after 28 min of exercise, and JNK phosphorylation was highly load-dependent. The abundance of all four kinases was higher in TA compared to EDL muscle, p70S6K abundance was increased after exercise in a load-independent manner, and FAK and JNK abundance was reduced after 28 min of exercise in both the exercised and control muscles. In conclusion, the current study shows that JNK activation after a single resistance exercise is load-specific, resembling the previously reported degree of myonuclear accrual and muscle hypertrophy with repetition of the exercise stimulus.
Collapse
Affiliation(s)
- Einar Eftestøl
- Department of Biosciences, University of Oslo, Kristine Bonnevies hus, Blindernveien 31, 0371, Oslo, Norway.
| | - Martino V Franchi
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland.,Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Stephanie Kasper
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland
| |
Collapse
|
49
|
Lautaoja JH, M O'Connell T, Mäntyselkä S, Peräkylä J, Kainulainen H, Pekkala S, Permi P, Hulmi JJ. Higher glucose availability augments the metabolic responses of the C2C12 myotubes to exercise-like electrical pulse stimulation. Am J Physiol Endocrinol Metab 2021; 321:E229-E245. [PMID: 34181491 PMCID: PMC8410101 DOI: 10.1152/ajpendo.00133.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The application of exercise-like electrical pulse simulation (EL-EPS) has become a widely used exercise mimetic in vitro. EL-EPS produces similar physiological responses as in vivo exercise, while less is known about the detailed metabolic effects. Routinely, the C2C12 myotubes are cultured in high-glucose medium (4.5 g/L), which may alter EL-EPS responses. In this study, we evaluate the metabolic effects of EL-EPS under the high- and low-glucose (1.0 g/L) conditions to understand how substrate availability affects the myotube response to EL-EPS. The C2C12 myotube, media, and cell-free media metabolites were analyzed using untargeted nuclear magnetic resonance (NMR)-based metabolomics. Furthermore, translational and metabolic changes and possible exerkine effects were analyzed. EL-EPS enhanced substrate utilization as well as production and secretion of lactate, acetate, 3-hydroxybutyrate, and branched-chain fatty acids (BCFAs). The increase in BCFAs correlated with branched-chain amino acids (BCAAs) and BCFAs were strongly decreased when myotubes were cultured without BCAAs suggesting the action of acyl-CoA thioesterases on BCAA catabolites. Notably, not all EL-EPS responses were augmented by high glucose because EL-EPS increased phosphorylated c-Jun N-terminal kinase and interleukin-6 secretion independent of glucose availability. Administration of acetate and EL-EPS conditioned media on HepG2 hepatocytes had no adverse effects on lipolysis or triacylglycerol content. Our results demonstrate that unlike in cell-free media, the C2C12 myotube and media metabolites were affected by EL-EPS, particularly under high-glucose condition suggesting that media composition should be considered in future EL-EPS studies. Furthermore, acetate and BCFAs were identified as putative exerkines warranting more research.NEW & NOTEWORTHY The present study examined for the first time the metabolome of 1) C2C12 myotubes, 2) their growth media, and 3) cell-free media after exercise-like electrical pulse stimulation under distinct nutritional loads. We report that myotubes grown under high-glucose conditions had greater responsiveness to EL-EPS when compared with lower glucose availability conditions and increased media content of acetate and branched-chain fatty acids suggests they might act as putative exerkines warranting further research.
Collapse
Affiliation(s)
- Juulia H Lautaoja
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Thomas M O'Connell
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sakari Mäntyselkä
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Juuli Peräkylä
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Heikki Kainulainen
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Perttu Permi
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Juha J Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
50
|
Halle JL, Counts-Franch BR, Prince RM, Carson JA. The Effect of Mechanical Stretch on Myotube Growth Suppression by Colon-26 Tumor-Derived Factors. Front Cell Dev Biol 2021; 9:690452. [PMID: 34395422 PMCID: PMC8363303 DOI: 10.3389/fcell.2021.690452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Preclinical models and in vitro experiments have provided valuable insight into the regulation of cancer-induced muscle wasting. Colon-26 (C26) tumor cells induce cachexia in mice, and conditioned media (CM) from these cells promotes myotube atrophy and catabolic signaling. While mechanical stimuli can prevent some effects of tumor-derived factors on myotubes, the impact of mechanical signaling on tumor-derived factor regulation of myosin heavy chain (MyHC) expression is not well understood. Therefore, we examined the effects of stretch-induced mechanical signaling on C2C12 myotube growth and MyHC expression after C26 CM exposure. C26 CM was administered to myotubes on day 5 of differentiation for 48 h. During the last 4 or 24 h of C26 CM exposure, 5% static uniaxial stretch was administered. C26 CM suppressed myotube growth and MyHC protein and mRNA expression. Stretch for 24 h increased myotube size and prevented the C26 CM suppression of MyHC-Fast protein expression. Stretch did not change suppressed MyHC mRNA expression. Stretch for 24 h reduced Atrogin-1/MAFbx, MuRF-1, and LC3B II/I ratio and increased integrin β1D protein expression and the myogenin-to-MyoD protein ratio. Stretch in the last 4 h of CM increased ERK1/2 phosphorylation but did not alter the CM induction of STAT3 or p38 phosphorylation. These results provide evidence that in myotubes pre-incubated with CM, the induction of mechanical signaling can still provide a growth stimulus and preserve MyHC-Fast protein expression independent of changes in mRNA expression.
Collapse
Affiliation(s)
| | | | | | - James A. Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|