1
|
Collao N, Johannsen EB, Just J, De Lisio M. Single-cell transcriptomic analysis reveals alterations to cellular dynamics and paracrine signaling in radiation-induced muscle pathology. Am J Physiol Cell Physiol 2025; 328:C1995-C2012. [PMID: 40316295 DOI: 10.1152/ajpcell.00115.2025] [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: 02/09/2025] [Revised: 02/27/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Radiation therapy causes long-term skeletal muscle atrophy and fibrosis in juvenile cancer survivors. The mechanisms responsible for the skeletal muscle late effects of radiation therapy are not well-understood and have prevented the development of effective treatments. Using single-cell RNA sequencing (scRNA-seq), we characterize cellular dynamics and communication in a murine model of therapeutic radiation at 24 h and 56 days post-irradiation (post-IR). We detected changes in muscle stem (satellite) cells (MuSCs) characterized by an acute preservation of committed MuSCs and long-term relative depletion of deep quiescent MuSCs. A conserved senescence Cdkn1a signature was observed in all muscle-resident cells post-IR. Genes related to fibroblast proliferation were upregulated and a fibrotic and senescent transcriptome persisted in fibro-adipogenic progenitors (FAPs) post-IR. Intercellular communication analysis revealed FAPs as the primary contributor of extracellular matrix (ECM) and target of monocyte/macrophage-derived transforming growth factor (TGF)-β signaling post-IR through TGF-βR2 on FAPs. Together, our findings provide insights into the potential mechanisms and intercellular communication responsible for radiation-induced muscle atrophy and fibrosis.NEW & NOTEWORTHY This work describes, for the first time, the transcriptional changes occurring following radiation exposure in the skeletal muscle microenvironment using scRNA-seq technology. We revelated that FAPs exhibited a profibrotic and senescent transcriptome. Radiation exposure led to a conserved and persistent Cdkn1a gene signature and impairs intercellular communication, increasing TGF-βR2 signaling in FAPs. These findings uncover potential mechanisms and intercellular communication responsible for long-term muscle impairments post-radiation, offering new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Nicolás Collao
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, Ontario, Canada
- Éric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Regenerative Medicine Program, University of Ottawa, Ottawa, Ontario, Canada
| | - Emma B Johannsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Science, University of Ottawa, Ottawa, Ontario, Canada
- Éric Poulin Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Regenerative Medicine Program, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
2
|
Cuijpers I, Katsburg J, van Loon LJC, Troost FJ, Sthijns MMJPE. Nutritional strategies targeting age-related skeletal muscle fibrosis: underlying mechanisms. Crit Rev Food Sci Nutr 2025:1-21. [PMID: 40336331 DOI: 10.1080/10408398.2025.2498676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Aging is associated with a reduced number and function of muscle stem cells (MuSC). This results in a decreased muscle regenerative capacity and increased formation of fibrotic tissue, impairing skeletal muscle function. This review provides an overview of in vitro and in vivo animal studies investigating nutritional interventions with the potential to inhibit pathophysiological mechanisms involved in the development of skeletal muscle fibrosis. Mechanism targets include 1) MuSC function and myogenic differentiation, 2) M1 to M2 macrophage polarization, 3) myofibroblast activity or extracellular matrix (ECM) deposition, and 4) reactive oxygen species (ROS) mediated pathways, such as NOX2/4 activity. Most promising nutrients described in this review are phytonutrients, vitamins and amino acids. Quercetin targets multiple pathways (showing decreased inflammation, ECM expression and NOX2/4 activity) in various cell types and tissues (kidney, aorta, liver and (heart) muscle) of rodents and rabbits, which could contribute to fibrosis development. Additionally, sulforaphane is a promising candidate as it inhibits inflammation, ECM expression, and ROS production in mouse skeletal muscle. After validation of the effects in human skeletal muscle, supplementation with these nutrients could be implemented in a multifaceted intervention (including exercise and adequate protein intake) targeting age-related skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Iris Cuijpers
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Joey Katsburg
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Freddy J Troost
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Mireille M J P E Sthijns
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| |
Collapse
|
3
|
Harland N, Johnen L, Avula KT, Buzanich-Ladinig A, Schwarz L, Knoll J, Stenzl A, Aicher WK. The Outcome of Cell Therapy Treating Urinary Incontinence Correlates with Precise Cell Localization in the Sphincter Complex. Biomedicines 2025; 13:917. [PMID: 40299502 PMCID: PMC12024915 DOI: 10.3390/biomedicines13040917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Urethral sphincter muscle deficiency is the leading cause of stress urinary incontinence. Preclinical and clinical studies suggested that cell therapy may improve the situation. However, the overall efficacy of cell therapies did often not satisfy the patient's needs. We, therefore, investigated in a large animal model of incontinence if the localization of injected regenerative cells in the deficient urethral sphincter muscle correlated with the outcome. Methods: Urethral sphincter insufficiency was induced in three cohorts of pigs and confirmed by urodynamics. Then, either myogenic progenitor cells (MPCs) or adipose tissue-derived stromal cells (ADSCs) were injected into the injured sphincter complex by Williams needle under visual using a cystoscope. Sham-treated animals served as controls. Functional sphincter muscle regeneration was monitored by urodynamics over 5 weeks of follow-up. The localization of the injected cells was investigated by histology of cryosections of the tissue targeted. Results: Injection of MPCs near the sphincter muscle yielded better functional recovery when compared to MPC injections in adjacent sides. By contrast, injection of ADSCs in the submucosal tissue adjacent to the muscle led to better regeneration when compared to ADSC injections into the sphincter muscle. After five weeks of follow-up, MPCs yielded an overall robust but not significant improvement when compared to mock-treated controls, while ADSC injections reached significance. Conclusions: This small proof-of-principle study suggests that the clinical outcome of cell therapy for urinary incontinence depends on the choice of therapeutic cells and the precise localization of the cells in the tissue targeted as well.
Collapse
Affiliation(s)
- Niklas Harland
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
| | - Liv Johnen
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
| | - Kamal T. Avula
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
| | - Andrea Buzanich-Ladinig
- Clinical Centre for Population Medicine in Fish, Pig and Poultry, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.B.-L.); (L.S.)
| | - Lukas Schwarz
- Clinical Centre for Population Medicine in Fish, Pig and Poultry, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.B.-L.); (L.S.)
| | - Jasmin Knoll
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
| | - Arnulf Stenzl
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
| | - Wilhelm K. Aicher
- Center for Medical Research, University of Tuebingen Hospital, 72072 Tuebingen, Germany; (N.H.); (L.J.); (K.T.A.); (A.S.)
- Department of Cardio-Thoracic Surgery, University of Tuebingen Hospital, 72076 Tuebingen, Germany
| |
Collapse
|
4
|
Shameem M, Olson SL, Marron Fernandez de Velasco E, Kumar A, Singh BN. Cardiac Fibroblasts: Helping or Hurting. Genes (Basel) 2025; 16:381. [PMID: 40282342 PMCID: PMC12026832 DOI: 10.3390/genes16040381] [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: 02/28/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Cardiac fibroblasts (CFs) are the essential cell type for heart morphogenesis and homeostasis. In addition to maintaining the structural integrity of the heart tissue, muscle fibroblasts are involved in complex signaling cascades that regulate cardiomyocyte proliferation, migration, and maturation. While CFs serve as the primary source of extracellular matrix proteins (ECM), tissue repair, and paracrine signaling, they are also responsible for adverse pathological changes associated with cardiovascular disease. Following activation, fibroblasts produce excessive ECM components that ultimately lead to fibrosis and cardiac dysfunction. Decades of research have led to a much deeper understanding of the role of CFs in cardiogenesis. Recent studies using the single-cell genomic approach have focused on advancing the role of CFs in cellular interactions, and the mechanistic implications involved during cardiovascular development and disease. Arguably, the unique role of fibroblasts in development, tissue repair, and disease progression categorizes them into the friend or foe category. This brief review summarizes the current understanding of cardiac fibroblast biology and discusses the key findings in the context of development and pathophysiological conditions.
Collapse
Affiliation(s)
- Mohammad Shameem
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Shelby L. Olson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - Akhilesh Kumar
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bhairab N. Singh
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
LaRocca TJ, Lark DS. Mapping Organism-wide Single Cell mRNA Expression Linked to Extracellular Vesicle Biogenesis, Secretion, and Cargo. FUNCTION 2025; 6:zqaf005. [PMID: 39863422 PMCID: PMC11931722 DOI: 10.1093/function/zqaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/23/2025] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are functional lipid-bound nanoparticles trafficked between cells and found in every biofluid. It is widely claimed that EVs can be secreted by every cell, but the quantity and composition of these EVs can differ greatly among cell types and tissues. Defining this heterogeneity has broad implications for EV-based communication in health and disease. Recent discoveries have linked single-cell EV secretion to the expression of genes encoding EV machinery and cargo. To gain insight at single-cell resolution across an entire organism, we compared the abundance, variance, and co-expression of 67 genes involved in EV biogenesis and secretion, or carried as cargo, across >44 000 cells obtained from 117 cell populations in the Tabula Muris. Our analysis provides both novel holistic and cell population-specific insight into EV biology. The highest overall expression of EV genes occurs in secretory cells of the pancreas and perhaps more surprisingly, multiple non-neuronal cell populations of the brain. We find that the most abundant EV genes encode the most abundant EV cargo proteins (tetraspanins and syndecans), but these genes are highly differentially expressed across functionally distinct cell populations. Expression variance identifies dynamic and constitutively expressed EV genes while co-expression analysis reveals novel insights into cell population-specific coordination of expression. Results of our analysis illustrate the diverse transcriptional regulation of EV genes which could be useful for predicting how individual cell populations might communicate via EVs to influence health and disease.
Collapse
Affiliation(s)
- Thomas J LaRocca
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, CO 80521, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO 80521, USA
| | - Daniel S Lark
- Department of Health and Exercise Science, College of Health and Human Sciences, Colorado State University, Fort Collins, CO 80521, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO 80521, USA
| |
Collapse
|
6
|
Murach KA, Englund DA, Chambers TL, Dungan CM, Porter HL, Wren JD, Freeman WM, Dupont‐Versteegden EE, Wen Y. A satellite cell-dependent epigenetic fingerprint in skeletal muscle identity genes after lifelong physical activity. FASEB J 2025; 39:e70435. [PMID: 40047419 PMCID: PMC11884312 DOI: 10.1096/fj.202500177r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025]
Abstract
Satellite cells comprise a small proportion of mononuclear cells in adult skeletal muscle. Despite their relative rarity, satellite cells have critical functions in muscle adaptation, particularly during prolonged exercise training. The mechanisms by which satellite cells mediate skeletal muscle responsiveness to physical activity throughout the lifespan are still being defined, but epigenetic regulation may play a role. To explore this possibility, we analyzed global DNA methylation patterns in muscle tissue from female mice that engaged in lifelong voluntary unweighted wheel running with or without satellite cells. Satellite cells were ablated in adulthood using the tamoxifen-inducible Pax7-DTA model. Compared to sedentary mice, wheel running for 13 months caused muscle DNA methylation differences in the promoter regions of numerous muscle fiber-enriched genes-Cacgn1, Dnm2, Mlip, Myl1, Myom2, Mstn, Sgca, Sgcg, Tnnc1, Tnni2, Tpm1, and Ttn-only when satellite cells were present. These genes relate to muscle fiber identity, cytoarchitecture, and size as well as overall muscle function. Epigenetic alterations to such genes are consistent with previously observed histological and in vivo impairments to running adaptation after satellite cell depletion in these same mice. Musk promoter region methylation was affected only in the absence of satellite cells with lifelong running relative to sedentary; this dovetails with work showing that satellite cells influence skeletal muscle innervation. Defining the epigenetic effects of satellite cells on identity genes in muscle fibers after lifelong physical activity provides new directions for how these rare stem cells can promote muscle adaptation and function throughout the lifespan.
Collapse
MESH Headings
- Animals
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/physiology
- Satellite Cells, Skeletal Muscle/cytology
- Epigenesis, Genetic
- Mice
- DNA Methylation
- Female
- Physical Conditioning, Animal/physiology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Mice, Inbred C57BL
- Promoter Regions, Genetic
- Mice, Transgenic
Collapse
Affiliation(s)
- Kevin A. Murach
- Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, Exercise Science Research CenterUniversity of ArkansasFayettevilleArkansasUSA
| | - Davis A. Englund
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Toby L. Chambers
- Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, Exercise Science Research CenterUniversity of ArkansasFayettevilleArkansasUSA
| | - Cory M. Dungan
- Department of Health, Human Performance, and RecreationBaylor UniversityWacoTexasUSA
| | - Hunter L. Porter
- Genes & Human Disease ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Jonathan D. Wren
- Genes & Human Disease ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Willard M. Freeman
- Genes & Human Disease ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma City Veterans Affairs Medical CenterOklahoma CityOklahomaUSA
| | - Esther E. Dupont‐Versteegden
- University of Kentucky Center for Muscle BiologyLexingtonKentuckyUSA
- Department of Physical TherapyUniversity of KentuckyLexingtonKentuckyUSA
| | - Yuan Wen
- University of Kentucky Center for Muscle BiologyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Division of Biomedical Informatics, Department of Internal MedicineUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
7
|
Murach KA, Bagley JR. A primer on global molecular responses to exercise in skeletal muscle: Omics in focus. JOURNAL OF SPORT AND HEALTH SCIENCE 2025:101029. [PMID: 39961420 DOI: 10.1016/j.jshs.2025.101029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/19/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Advances in skeletal muscle omics has expanded our understanding of exercise-induced adaptations at the molecular level. Over the past 2 decades, transcriptome studies in muscle have detailed acute and chronic responses to resistance, endurance, and concurrent exercise, focusing on variables such as training status, nutrition, age, sex, and metabolic health profile. Multi-omics approaches, such as the integration of transcriptomic and epigenetic data, along with emerging ribosomal RNA sequencing advancements, have further provided insights into how skeletal muscle adapts to exercise across the lifespan. Downstream of the transcriptome, proteomic and phosphoproteomic studies have identified novel regulators of exercise adaptations, while single-cell/nucleus and spatial sequencing technologies promise to evolve our understanding of cellular specialization and communication in and around skeletal muscle cells. This narrative review highlights (a) the historical foundations of exercise omics in skeletal muscle, (b) current research at 3 layers of the omics cascade (DNA, RNA, and protein), and (c) applications of single-cell omics and spatial sequencing technologies to study skeletal muscle adaptation to exercise. Further elaboration of muscle's global molecular footprint using multi-omics methods will help researchers and practitioners develop more effective and targeted approaches to improve skeletal muscle health as well as athletic performance.
Collapse
Affiliation(s)
- Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR 72701, USA.
| | - James R Bagley
- Muscle Physiology Laboratory, Department of Kinesiology, College of Health and Social Sciences, San Francisco State University, San Francisco, CA 94132, USA.
| |
Collapse
|
8
|
Owen AM, Gonzalez-Velez S, Keeble AR, Thomas NT, Fry CS. Fork in the road: therapeutic and pathological actions for fibro-adipogenic progenitors following musculoskeletal injury. J Physiol 2025. [PMID: 39930980 DOI: 10.1113/jp286816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/20/2025] [Indexed: 02/19/2025] Open
Abstract
Musculoskeletal injuries are a substantial source of global disability through weakness and loss of function, which can be attributable, in part, to deficits in skeletal muscle quality. Poor muscle quality, resulting from fibrotic pathology or fatty infiltration, strongly predicts lower rates of patient recovery following injury and higher rates of re-injury. The cellular sources of fibrosis and fatty infiltration within skeletal muscle are mesenchymal fibro-adipogenic progenitors (FAPs), which are central effectors to support muscle homeostasis, regeneration and growth. However, following acute or chronic musculoskeletal injury, FAPs can promote fibro/fatty pathology within muscle that is likely to limit recovery and repair. Given their indispensable role within skeletal muscle, FAPs have emerged as a compelling cellular target to promote tissue recovery following acute and chronic injury. This review provides insight into the aetiology of FAP activity following various musculoskeletal injuries, in addition to signalling components that effect FAP differentiation. Contrasting pathology with therapeutic potential, insight into disease- and injury-specific FAP activation further cements their role as crucial effectors to improve muscle function and enhance patient outcomes.
Collapse
Affiliation(s)
- Allison M Owen
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Sara Gonzalez-Velez
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Alexander R Keeble
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Chung JD, Porrello ER, Lynch GS. Muscle regeneration and muscle stem cells in metabolic disease. Free Radic Biol Med 2025; 227:52-63. [PMID: 39581389 DOI: 10.1016/j.freeradbiomed.2024.11.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
Skeletal muscle has a high regenerative capacity due to its resident adult muscle stem cells (MuSCs), which can repair damaged tissue by forming myofibres de novo. Stem cell dependent regeneration is critical for maintaining skeletal muscle health, and different conditions can draw heavily on MuSC support to preserve muscle function, including metabolic diseases such as diabetes. The global incidence and burden of diabetes is increasing, and skeletal muscle is critical for maintaining systemic metabolic homeostasis and improving outcomes for diabetic patients. Thus, poor muscle health in diabetes, termed diabetic myopathy, is an important complication that must be addressed. The health of MuSCs is also affected by diabetes, responsible for the poor muscle regenerative capacity and contributing to the functional decline in diabetic patients. Here, we review the impact of diabetes and metabolic disease on MuSCs and skeletal muscle, including potential mechanisms for impaired muscle regeneration and MuSC dysfunction, and how these deficits could be addressed.
Collapse
Affiliation(s)
- Jin D Chung
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, 3010, VIC, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, 3052, VIC, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, 3052, VIC, Australia
| | - Enzo R Porrello
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, 3010, VIC, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, 3052, VIC, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, 3052, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, 3010, VIC, Australia.
| |
Collapse
|
10
|
Michel JM, Godwin JS, Plotkin DL, McIntosh MC, Mattingly ML, Agostinelli PJ, Mueller BJ, Anglin DA, Kontos NJ, Berry AC, Vega MM, Pipkin AA, Stock MS, Graham ZA, Baweja HS, Mobley CB, Bamman MM, Roberts MD. Effects of leg immobilization and recovery resistance training on skeletal muscle-molecular markers in previously resistance-trained versus untrained adults. J Appl Physiol (1985) 2025; 138:450-467. [PMID: 39819075 DOI: 10.1152/japplphysiol.00837.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
We sought to examine how resistance training (RT) status in young healthy individuals, either well resistance trained (T, n = 10) or untrained (UT, n = 11), affected molecular markers with leg immobilization followed by recovery RT. All participants underwent 2 wk of left leg immobilization via a locking leg brace. Afterward, all participants underwent 8 wk (3 days/wk) of knee extensor-focused progressive RT. Vastus lateralis (VL) ultrasound-derived thickness and muscle cross-sectional area were measured at baseline (PRE), immediately after disuse (MID), and after RT (POST) with VL muscle biopsies also being collected at these time points. Both groups presented lower ultrasound-derived VL size metrics at MID versus PRE (P ≤ 0.001), and values increased in both groups from MID to POST (P < 0.05); however, VL size increased from PRE to POST in UT only (P < 0.001). Mean and type II myofiber cross-sectional area values were greater at PRE and POST versus MID (P < 0.05), with T being greater than UT throughout (P ≤ 0.012). In both groups, satellite cell number was not affected by leg immobilization but increased in response to RT (P ≤ 0.014), with T being greater than UT throughout (P = 0.004). Total RNA (ribosome content) decreased (P = 0.010) from PRE to MID while total RNA and certain endoplasmic reticulum stress proteins increased from MID to POST regardless of training status. Immobilization-induced muscle atrophy and recovery RT hypertrophy outcomes are similar between UT and T participants, and the lack of molecular signature differences between groups supports these findings. However, results are limited to younger adults undergoing noncomplicated disuse.NEW & NOTEWORTHY Formerly trained and untrained individuals demonstrate similar atrophic responses to disuse while untrained individuals exhibited a greater hypertrophic response to subsequent resistance training. The molecular responses accompanying these changes were largely similar between groups and included increases in satellite cell content with resistance training and increases in ribosome biogenesis, which was largely driven by the formerly trained group.
Collapse
Affiliation(s)
- J Max Michel
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Joshua S Godwin
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Daniel L Plotkin
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Mason C McIntosh
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | | | | | - Breanna J Mueller
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Derick A Anglin
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Nicholas J Kontos
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Alexander C Berry
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Marina Meyer Vega
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Autumn A Pipkin
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Matt S Stock
- School of Kinesiology and Rehabilitation Sciences, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, Florida, United States
| | - Zachary A Graham
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| | - Harsimran S Baweja
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - C Brooks Mobley
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Marcas M Bamman
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
- Edward Via College of Osteopathic Medicine, Auburn, Alabama, United States
| |
Collapse
|
11
|
Sun J, Jia Y, Chen S, Bian Y, Liang H, Du X. Adipose mesenchymal stem cell-derived extracellular vesicles alleviate renal fibrosis by reducing epithelial-mesenchymal transition via the FOXS1/Wnt/β-catenin signaling pathway. Int Immunopharmacol 2025; 146:113880. [PMID: 39709913 DOI: 10.1016/j.intimp.2024.113880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/07/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Adipose mesenchymal stem cells (ADSCs) exert beneficial effects on kidney disease through a paracrine mechanism. However, the specific molecular mechanisms by which ADSCs treat renal fibrosis are not yet fully understood. Therefore, it is crucial to clarify the therapeutic effects of ADSC-derived extracellular vesicles (ADSC-EVs) on the progression of renal fibrosis and their underlying mechanisms. METHODS We investigated the therapeutic effects of ADSC-EVs on renal fibrosis both in vivo and in vitro. Key genes and signaling pathways were identified with RNA sequencing analysis of HK-2 cells. The role and underlying mechanism of the FOXS1/Wnt/β-catenin pathway in mediating antifibrotic effects were also verified. RESULTS In vivo, We found that ADSC-EV treatment significantly improves renal fibrosis in unilateral ureteral obstruction (UUO)-induced renal fibrosis mice models. And in vitro, our data suggested that ADSC-EVs can reduce epithelial-mesenchymal transition (EMT) to inhibit fibrosis in transforming growth factor-β1 (TGF-β1)-treated HK-2 cells. The RNA sequencing results showed that FOXS1 was the primary gene involved in ADSC-EV treatment of renal fibrosis. RT-qPCR suggested that ADSC-EV treatment reversed elevated FOXS1 level both in TGF-β1-treated HK-2 cells and UUO-induced renal fibrosis mice models. Moreover, Western blot analysis confirmed that ADSC-EVs alleviate renal fibrosis and EMT by inhibiting the expression of FOXS1 in HK-2 cells treated with TGF-β1. Furthermore, overexpression of FOXS1 in HK-2 cells could promote the occurrence of fibrosis and knockdown of FOXS1 could reduce the occurrence of fibrosis. Finally, ADSC-EVs may exert these effects via FOXS1-mediated activation of the Wnt/β-catenin pathway. CONCLUSION Taken together, we have confirmed that ADSC-EVs alleviate renal fibrosis by reducing EMT via the FOXS1/Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanyuan Jia
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Sha Chen
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haihai Liang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Xuanyi Du
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Tsuchiya Y, Yeung CYC, Svensson RB, Kjaer M. Effect of human myoblasts on tenogenic progression in 2D and 3D culture models. J Anat 2025. [PMID: 39854094 DOI: 10.1111/joa.14224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/04/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Tendon injuries and disorders associated with mechanical tendon overuse are common musculoskeletal problems. Even though tendons play a central role in human movement, the intrinsic healing process of tendon is very slow. So far, it is known that tendon cell activity is supported by several interstitial cells within the tendon. However, the interplay between the tendon and the adjacent muscle for tendon regeneration and development processes has not been fully investigated. Here, we tested whether factors released from muscle derived myogenic cells (myoblasts) enhance tenogenic progressions of human tendon derived cells (tendon fibroblasts) using two-dimensional (2D) culture model and a three-dimensional (3D)-engineered tendon construct culture model, which mimics tendon regeneration and development. The conditioned media from myoblasts and unconditioned media as control were applied to tendon fibroblasts. In 2D, immunofluorescence analysis revealed increased collagen type I expressing area and increased migration potential when conditioned media from myoblasts were applied. In the 3D-engineered human tendon construct model, wet weight, diameter, and cross-sectional area of the tendon constructs were increased in response to the application of conditioned media from myoblasts, whereas the collagen density was lower and mechanical function was reduced both at the functional level (maximum stiffness) and the material level (maximum stress and modulus). These results indicate that myoblast-derived factors extend collagen expressing area and enhance migration of tendon fibroblasts, while factors involved in the robustness of extra-cellular matrix deposition of tissue-engineered tendon constructs are lacking. Our findings suggest that adjacent muscle affects the signaling interplay in tendons.
Collapse
Affiliation(s)
- Yoshifumi Tsuchiya
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Sports Science, Doshisha University, Kyoto, Japan
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Japan
| | - Ching-Yan Chloé Yeung
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg-Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Millozzi F, Milán-Rois P, Sett A, Delli Carpini G, De Bardi M, Gisbert-Garzarán M, Sandonà M, Rodríguez-Díaz C, Martínez-Mingo M, Pardo I, Esposito F, Viscomi MT, Bouché M, Parolini O, Saccone V, Toulmé JJ, Somoza Á, Palacios D. Aptamer-conjugated gold nanoparticles enable oligonucleotide delivery into muscle stem cells to promote regeneration of dystrophic muscles. Nat Commun 2025; 16:577. [PMID: 39794309 PMCID: PMC11724063 DOI: 10.1038/s41467-024-55223-9] [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: 02/01/2024] [Accepted: 12/03/2024] [Indexed: 01/13/2025] Open
Abstract
Inefficient targeting of muscle stem cells (MuSCs), also called satellite cells, represents a major bottleneck of current therapeutic strategies for muscular dystrophies, as it precludes the possibility of promoting compensatory regeneration. Here we describe a muscle-targeting delivery platform, based on gold nanoparticles, that enables the release of therapeutic oligonucleotides into MuSCs. We demonstrate that AuNPs conjugation to an aptamer against α7/β1 integrin dimers directs either local or systemic delivery of microRNA-206 to MuSCs, thereby promoting muscle regeneration and improving muscle functionality, in a mouse model of Duchenne Muscular Dystrophy. We show here that this platform is biocompatible, non-toxic, and non-immunogenic, and it can be easily adapted for the release of a wide range of therapeutic oligonucleotides into diseased muscles.
Collapse
Affiliation(s)
- Francesco Millozzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
| | | | - Arghya Sett
- Bordeaux University, Inserm U1212, CNRS UMR5320, Bordeaux, France
- ERIN Department, Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Giovanni Delli Carpini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | - Martina Sandonà
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Santa Lucia IRCCS, Rome, Italy
| | | | | | | | - Federica Esposito
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
- Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico, Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marina Bouché
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, Rome, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico, Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valentina Saccone
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico, Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Jean-Jacques Toulmé
- Bordeaux University, Inserm U1212, CNRS UMR5320, Bordeaux, France.
- Novaptech, Gradignan, France.
| | - Álvaro Somoza
- IMDEA Nanociencia, Madrid, Spain.
- Unidad Asociada de Nanobiomedicina, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| | - Daniela Palacios
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.
- Institute for Systems Analysis and Computer Science "Antonio Ruberti" (IASI), National Research Council (CNR), Rome, Italy.
| |
Collapse
|
14
|
Jia J, Wang L, Zhou Y, Zhang P, Chen X. Muscle-derived extracellular vesicles mediate crosstalk between skeletal muscle and other organs. Front Physiol 2025; 15:1501957. [PMID: 39844898 PMCID: PMC11750798 DOI: 10.3389/fphys.2024.1501957] [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: 09/26/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Skeletal muscle (SKM) has crucial roles in locomotor activity and posture within the body and also functions have been recognized as an actively secretory organ. Numerous bioactive molecules are secreted by SKM and transported by extracellular vesicles (EVs), a novel class of mediators of communication between cells and organs that contain various types of cargo molecules including lipids, proteins and nucleic acids. SKM-derived EVs (SKM-EVs) are intercellular communicators with significant roles in the crosstalk between SKM and other organs. In this review, we briefly describe the biological characteristics, composition, and uptake mechanisms of EVs, particularly exosomes, comprehensively summarize the regulatory effects of SKM-EVs on the function of, which include myogenesis, muscle repair and regeneration, as well as metabolic regulation. Furthermore, we explore the impact of SKM- EVs on various organs including bone, the cardiovascular system, adipose tissue, and nervous system. As emerging evidence suggests that SKM-EVs are involved in the development and regulation of type 2 diabetes (T2D), systemic inflammation, and other chronic diseases, we also highlight the potential of SKM-EVs as therapeutic targets and diagnostic biomarkers, emphasizing the need for further research to elucidate the complex mechanisms underlying intercellular communication in physiological and pathological contexts.
Collapse
Affiliation(s)
- Jiajie Jia
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lu Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Peng Zhang
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
15
|
Fei S, Rule BD, Godwin JS, Mobley CB, Roberts MD, von Walden F, Vechetti IJ. miRNA-1 regulation is necessary for mechanical overload-induced muscle hypertrophy in male mice. Physiol Rep 2025; 13:e70166. [PMID: 39761956 PMCID: PMC11705529 DOI: 10.14814/phy2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that play a critical role in regulating gene expression post-transcriptionally. They are involved in various developmental and physiological processes, and their dysregulation is linked to various diseases. Skeletal muscle-specific miRNAs, including miR-1, play a crucial role in the development and maintenance of skeletal muscle. It has been demonstrated that the expression of miR-1 decreases by approximately 50% in response to hypertrophic stimuli, suggesting its potential involvement in muscle hypertrophy. In our study, we hypothesize that reduction of miR-1 levels is necessary for skeletal muscle growth due to its interaction to essential pro-growth genes. Promoting a smaller reduction of miR-1 levels, we observed a blunted hypertrophic response in mice undergoing a murine model of muscle hypertrophy. In addition, our results suggest that miR-1 inhibits the expression of Itm2a, a membrane-related protein, as potential miR-1-related candidate for skeletal muscle hypertrophy. While the exact mechanism in muscle hypertrophy has not been identified, our results suggest that miR-1-regulated membrane proteins are important for skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- Shengyi Fei
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| | - Blake D. Rule
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| | | | | | | | | | - Ivan J. Vechetti
- Department of Nutrition and Health SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| |
Collapse
|
16
|
Wu L, Zhuang Z, Jia W, Li Y, Lu Y, Xu M, Bai H, Wang Z, Chang G, Jiang Y. Exploring the molecular basis of efficient feed utilization in low residual feed intake slow-growing ducks based on breast muscle transcriptome. Poult Sci 2025; 104:104613. [PMID: 39631277 PMCID: PMC11652873 DOI: 10.1016/j.psj.2024.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Residual feed intake (RFI) has recently gained attention as a key indicator of feed efficiency in poultry. In this study, 800 slow-growing ducks with similar initial body weights were reared in an experimental facility until they were culled at 42 d of age. Thirty high RFI (HRFI) and 30 low RFI (LRFI) birds were selected to evaluate their growth performance, carcass characteristics, and muscle development. Transcriptome and weighted gene co-expression correlation network analyses of pectoral muscles were conducted on six LRFI and six HRFI ducks. The results revealed that selecting for LRFI significantly reduced feed consumption (P < 0.05) and improved feed efficiency without affecting the growth performance, slaughter rate, or meat quality of ducks (P > 0.05). Moreover, compared with HRFI ducks, LRFI ducks had a lower pectoral muscle fat content (P < 0.05), larger muscle fiber diameter and area (P < 0.05), and lower muscle fiber density (P < 0.05). There were significant differences in gene expression between LRFI and HRFI ducks, with 102 upregulated and 258 downregulated genes, which were enriched in the PPAR signaling pathway, adipocytokine signaling pathway, actin cytoskeleton regulation, ECM-receptor interaction, and focal adhesion. The expression of genes associated with fat and energy metabolism, including ACSL6, PCK1, APOC3, HMGCS2, PRKAG3, and G6PC1, was downregulated in LRFI ducks, and weighted gene co-expression correlation network analysis identified PRKAG3 as a hub gene. Our findings indicate that reduced mitochondrial energy metabolism may contribute to the RFI of slow-growing ducks, with PRKAG3 playing a pivotal role in this biological process. These findings provide novel insights into the molecular changes underlying RFI variation in slow-growing ducks.
Collapse
Affiliation(s)
- Lei Wu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Wenqian Jia
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Yongpeng Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Yijia Lu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Minghong Xu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, PR China
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
17
|
Aussieker T, Fuchs CJ, Zorenc AH, Verdijk LB, van Loon LJC, Snijders T. Daily blood flow restriction does not affect muscle fiber capillarization and satellite cell content during 2 wk of bed rest in healthy young men. J Appl Physiol (1985) 2025; 138:89-98. [PMID: 39625459 DOI: 10.1152/japplphysiol.00461.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025] Open
Abstract
The present study assessed whether single-leg daily blood flow restriction (BFR) treatment attenuates the decline in muscle fiber size, capillarization, and satellite cell (SC) content during 2 wk of bed rest in healthy, young men. Twelve healthy, young men (age: 24 ± 3 yr; BMI: 23.7 ± 3.1 kg/m2) were subjected to 2 wk of bed rest, during which one leg was exposed to three times daily 5 min of BFR, whereas the contralateral leg received sham treatment [control (CON)]. Muscle biopsies were obtained from the m. vastus lateralis from both the BFR and CON legs before and immediately after 2 wk of bed rest. Types I and II muscle fiber size, myonuclear content, capillarization, and SC content were assessed by immunohistochemistry. No significant decline in either type I or type II muscle fiber size was observed following bed rest, with no differences between the CON and BFR legs (P > 0.05). Type I muscle fiber capillary density increased in response to bed rest in both legs (P < 0.05), whereas other muscle fiber capillarization measures remained unaltered. SC content decreased in both type I (from 7.4 ± 3.2 to 5.9 ± 2.7 per 100 fibers) and type II (from 7.2 ± 3.4 to 6.5 ± 3.2 per 100 fibers) muscle fibers (main effect of time P = 0.018), with no significant differences between the BFR and CON legs (P > 0.05). In conclusion, 2 wk of bed rest has no effect on muscle capillarization and decreases the SC content, and daily BFR treatment does not affect skeletal muscle fiber size and SC content in healthy, young men.NEW & NOTEWORTHY We recently reported that the application of daily blood flow restriction (BFR) treatment does not preserve muscle mass or strength and does not modulate daily muscle protein synthesis rates during 2 wk of bed rest. Here, we show that 2 wk of bed rest resulted in a decrease in satellite cell (SC) content. In addition, the BFR treatment did not affect muscle fiber size, capillarization, and SC content during 2 wk of bed rest.
Collapse
Affiliation(s)
- Thorben Aussieker
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Cas J Fuchs
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Antoine H Zorenc
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Lex B Verdijk
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Tim Snijders
- Department of Human Biology, NUTRIM, Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
18
|
Borowik AK, Murach KA, Miller BF. The expanding roles of myonuclei in adult skeletal muscle health and function. Biochem Soc Trans 2024; 52:1-14. [PMID: 39700019 DOI: 10.1042/bst20241637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Skeletal muscle cells (myofibers) require multiple nuclei to support a cytoplasmic volume that is larger than other mononuclear cell types. It is dogmatic that mammalian resident myonuclei rely on stem cells (specifically satellite cells) for adding new DNA to muscle fibers to facilitate cytoplasmic expansion that occurs during muscle growth. In this review, we discuss the relationship between cell size and supporting genetic material. We present evidence that myonuclei may undergo DNA synthesis as a strategy to increase genetic material in myofibers independent from satellite cells. We then describe the details of our experiments that demonstrated that mammalian myonuclei can replicate DNA in vivo. Finally, we present our findings in the context of expanding knowledge about myonuclear heterogeneity, myonuclear mobility and shape. We also address why myonuclear replication is potentially important and provide future directions for remaining unknowns. Myonuclear DNA replication, coupled with new discoveries about myonuclear transcription, morphology, and behavior in response to stress, may provide opportunities to leverage previously unappreciated skeletal muscle biological processes for therapeutic targets that support muscle mass, function, and plasticity.
Collapse
Affiliation(s)
- Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, U.S.A
| | - Kevin A Murach
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, U.S.A
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, U.S.A
- Oklahoma City VA Medical Center, Oklahoma City, OK, U.S.A
| |
Collapse
|
19
|
Taye N, Rodriguez L, Iatridis JC, Han WM, Hubmacher D. Myoblast-derived ADAMTS-like 2 promotes skeletal muscle regeneration after injury. NPJ Regen Med 2024; 9:39. [PMID: 39702607 DOI: 10.1038/s41536-024-00383-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Skeletal muscle regeneration and functional recovery after minor injuries requires the activation of muscle-resident myogenic muscle stem cells (i.e. satellite cells) and their subsequent differentiation into myoblasts, myocytes, and ultimately myofibers. We recently identified secreted ADAMTS-like 2 (ADAMTSL2) as a pro-myogenic regulator of muscle development, where it promoted myoblast differentiation. Since myoblast differentiation is a key process in skeletal muscle regeneration, we here examined the role of ADAMTSL2 during muscle regeneration after BaCl2 injury. Specifically, we found that muscle regeneration was delayed after ablation of ADAMTSL2 in myogenic precursor cells and accelerated following injection of pro-myogenic ADAMTSL2 protein domains. Mechanistically, ADAMTSL2 regulated the number of committed myoblasts, which are the precursors for myocytes and regenerating myofibers. Collectively, our data support a role for myoblast-derived ADAMTSL2 as a positive regulator of muscle regeneration and provide a proof-of-concept for potential therapeutic applications.
Collapse
Affiliation(s)
- Nandaraj Taye
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Levon Rodriguez
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - James C Iatridis
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Woojin M Han
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dirk Hubmacher
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine, New York, NY, 10029, USA.
| |
Collapse
|
20
|
Alakhdar AA, Sivakumar S, Kopchak RM, Hunter AN, Ambrosio F, Washburn NR. Age-Related ECM Stiffness Mediates TRAIL Activation in Muscle Stem Cell Differentiation. Adv Biol (Weinh) 2024; 8:e2400334. [PMID: 39601528 PMCID: PMC11889993 DOI: 10.1002/adbi.202400334] [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: 06/13/2024] [Revised: 10/01/2024] [Indexed: 11/29/2024]
Abstract
The stiffening of the extracellular matrix (ECM) with age hinders muscle regeneration by causing intrinsic muscle stem cell (MuSC) dysfunction through a poorly understood mechanism. Here, the study aims to study those age-related molecular changes in the differentiation of MuSCs due to age and/or stiffness. Hence, young and aged MuSCs are seeded onto substrates engineered to mimic a soft and stiff ECM microenvironment to study those molecular changes using single-cell RNA sequencing (scRNA). The trajectory of scRNA data of the MuSCs under four different conditions undergoing differentiation is analyzed as well as the active molecular pathways and transcription factors driving those differentiation fates. Data revealed the presence of a branching point within the trajectory leading to the emergence of an age-related fibroblastic population characterized by activation of the TNF-related apoptosis-inducing ligand (TRAIL) pathway, which is significantly activated in aged cells cultured on stiff substrates. Next, using the collagen cross-linking inhibitor β-aminopropionitrile (BAPN) in vivo, the study elucidates stiffness changes on TRAIL downstream apoptotic targets (caspase 8 and caspase 3) using immunostaining. TRAIL activity is significantly inhibited by BAPN in aged animals, indicating a complex mechanism of age-related declines in muscle function through inflammatory and apoptotic mediators.
Collapse
Affiliation(s)
- Amira A. Alakhdar
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | | | - Rylee M. Kopchak
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
| | - Allison N. Hunter
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Newell R. Washburn
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| |
Collapse
|
21
|
Bourgeois BL, Gallegos EM, Levitt DE, Bergeaux PJ, Molina PE, Simon L. Extracellular vesicle miR-206 improves chronic binge alcohol-mediated decreased myoblast differentiation in SIV-infected female macaques. Am J Physiol Cell Physiol 2024; 327:C1626-C1637. [PMID: 39099419 PMCID: PMC11684875 DOI: 10.1152/ajpcell.00290.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/03/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
Alcohol misuse in people with human immunodeficiency virus (HIV) (PWH) and chronic binge alcohol (CBA) administration in simian immunodeficiency virus (SIV)-infected macaques are associated with increased physical frailty and impaired functional skeletal muscle mass, respectively. Previous studies by our group demonstrate that muscle-enriched microRNAs (myomiRs) are differentially expressed in skeletal muscle (SKM) from CBA-administered SIV-infected male macaques and their altered expression contributes to impaired differentiation of SKM stem cells or myoblasts. MicroRNAs can be transported in extracellular vesicles (EVs) to mediate numerous cellular responses through intercellular communication. The present study tested the hypothesis that EV-mediated delivery of miR-206 can ameliorate CBA-mediated decreases in myoblast differentiation. Myoblasts were isolated from SKM of female SIV-infected, antiretroviral therapy-treated macaques that received either CBA (2.5 g/kg/day, CBA/SIV) or water (VEH/SIV) for 14.5 mo. Myotube and myotube-derived EV myomiR expression, including miR-206, was lower in the CBA/SIV group. Overexpression of miR-206 decreased histone deacetylase 4 (HDAC4) and paired box 7 (PAX7) expression in myotubes and increased fusion index, a differentiation index, in CBA/SIV-derived myotubes. Similarly, EV-mediated delivery of miR-206 increased both fusion index and myotube density of CBA/SIV-derived myoblasts. These results support the potential therapeutic utility of EVs in delivering myomiRs to improve SKM stem cell differentiation.NEW & NOTEWORTHY Alcohol decreases skeletal muscle myoblast differentiation into myotubes, which is associated with decreased expression of microRNA-206. We show that delivering exogenous miR-206 in plasma-derived extracellular vesicles (EVs) to myoblasts derived from alcohol-administered animals increases myotube differentiation. These results support the potential therapeutic utility of EVs in delivering muscle-enriched microRNAs to improve skeletal muscle stem cell differentiation.
Collapse
Affiliation(s)
- Brianna L Bourgeois
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| | - Eden M Gallegos
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| | - Danielle E Levitt
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| | - Peter J Bergeaux
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| | - Patricia E Molina
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| | - Liz Simon
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research CenterLouisiana State University Health Sciences Center, New Orleans, Louisiana, United States
| |
Collapse
|
22
|
Keane AJ, Sanz-Nogués C, Jayasooriya D, Creane M, Chen X, Lyons CJ, Sikri I, Goljanek-Whysall K, O'Brien T. miR-1, miR-133a, miR-29b and skeletal muscle fibrosis in chronic limb-threatening ischaemia. Sci Rep 2024; 14:29393. [PMID: 39592654 PMCID: PMC11599917 DOI: 10.1038/s41598-024-76415-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024] Open
Abstract
Chronic limb-threatening ischaemia (CLTI), the most severe manifestation of peripheral arterial disease (PAD), is associated with a poor prognosis and high amputation rates. Despite novel therapeutic approaches being investigated, no significant clinical benefits have been observed yet. Understanding the molecular pathways of skeletal muscle dysfunction in CLTI is crucial for designing successful treatments. This study aimed to identify miRNAs dysregulated in muscle biopsies from PAD cohorts. Using MIcroRNA ENrichment TURned NETwork (MIENTURNET) on a publicly accessible RNA-sequencing dataset of PAD cohorts, we identified a list of miRNAs that were over-represented among the upregulated differentially expressed genes (DEGs) in CLTI. Next, we validated the altered expression of these miRNAs and their targets in mice with hindlimb ischaemia (HLI). Our results showed a significant downregulation of miR-1, miR-133a, and miR-29b levels in the ischaemic limbs versus the contralateral non-ischaemic limb. A miRNA target protein-protein interaction network identified extracellular matrix components, including collagen-1a1, -3a1, and -4a1, fibronectin-1, fibrin-1, matrix metalloproteinase-2 and -14, and Sparc, which were upregulated in the ischaemic muscle of mice. This is the first study to identify miR-1, miR-133a, and miR-29b as potential contributors to fibrosis and vascular pathology in CLTI muscle, which supports their potential as novel therapeutic agents for this condition.
Collapse
Affiliation(s)
- Alan J Keane
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Clara Sanz-Nogués
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland.
- CÚRAM SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| | - Dulan Jayasooriya
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Michael Creane
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Xizhe Chen
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Caomhán J Lyons
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Isha Sikri
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
| | - Katarzyna Goljanek-Whysall
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), University of Galway, Biomedical Sciences 1st Floor South, Corrib Village, Dangan, Galway, Ireland
- CÚRAM SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
23
|
Luo J, Pu Q, Wu X. Recent Advances of Exosomes Derived from Skeletal Muscle and Crosstalk with Other Tissues. Int J Mol Sci 2024; 25:10877. [PMID: 39456658 PMCID: PMC11507631 DOI: 10.3390/ijms252010877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Skeletal muscle plays a crucial role in movement, metabolism, and energy homeostasis. As the most metabolically active endocrine organ in the body, it has recently attracted widespread attention. Skeletal muscle possesses the ability to release adipocytokines, bioactive peptides, small molecular metabolites, nucleotides, and other myogenic cell factors; some of which have been shown to be encapsulated within small vesicles, particularly exosomes. These skeletal muscle exosomes (SKM-Exos) are released into the bloodstream and subsequently interact with receptor cell membranes to modulate the physiological and pathological characteristics of various tissues. Therefore, SKM-Exos may facilitate diverse interactions between skeletal muscle and other tissues while also serving as biomarkers that reflect the physiological and pathological states of muscle function. This review delves into the pivotal role and intricate molecular mechanisms of SKM-Exos and its derived miRNAs in the maturation and rejuvenation of skeletal muscle, along with their intercellular signaling dynamics and physiological significance in interfacing with other tissues.
Collapse
Affiliation(s)
- Jia Luo
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Qiang Pu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xiaoqian Wu
- College of Food Science, Southwest University, Chongqing 400715, China
| |
Collapse
|
24
|
Yu N, Chang X, Hu J, Li J, Ma J, Huang L. mRNA expression profiles in muscle-derived extracellular vesicles of Large White and wild boar piglets reveal their potential roles in immunity and muscle phenotype. Front Vet Sci 2024; 11:1452704. [PMID: 39421829 PMCID: PMC11484452 DOI: 10.3389/fvets.2024.1452704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Extracellular vesicles (EVs) known for their pivotal role in intercellular communication through RNA delivery, hold paramount implications for understanding muscle phenotypic variations in diverse pig breeds. Methods In this study, we compared the mRNA expression profiles of longissimus dorsi muscles and muscle-derived extracellular vesicles (M-EVs), and also examined the diversity of enriched genes in M-EVs between weaned wild boars and commercial Large White pigs with respect to their numbers and biological functions. Results The results of the study showed that the variation in the expression profiles of mRNAs between muscles and M-EVs was much greater than the variability between the respective breeds. Meanwhile, the enrichment trend of low-expressed genes (ranked <1,000) was significantly (p-value ≤ 0.05) powerful in M-EVs compared to highly expressed genes in muscles. In addition, M-EVs carried a smaller proportion of coding sequences and a larger proportion of untranslated region sequences compared to muscles. There were 2,110 genes enriched in M-EVs (MEGs) in Large White pigs and 2,322 MEGs in wild boars, with 1,490 MEGs shared interbreeds including cyclin D2 (CCND2), which inhibits myogenic differentiation. Of the 89 KEGG pathways that were significantly enriched (p-value ≤ 0.05) for these MEGs, 13 unique to Large White pigs were mainly related to immunity, 27 unique to wild boars were functionally diverse but included cell fate regulation such as the Notch signaling pathway and the TGF-beta signaling pathway, and 49 were common to both breeds were also functionally complex but partially related to innate immunity, such as the Complement and coagulation cascades and the Fc gamma R-mediated phagocytosis. Discussion These findings suggest that mRNAs in M-EVs have the potential to serve as indicators of muscle phenotype differences between the two pig breeds, highlighting the need for further exploration into the role of EV-RNAs in pig phenotype formation.
Collapse
Affiliation(s)
| | | | | | | | - Junwu Ma
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Lusheng Huang
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| |
Collapse
|
25
|
Villa C, Secchi V, Macchi M, Tripodi L, Trombetta E, Zambroni D, Padelli F, Mauri M, Molinaro M, Oddone R, Farini A, De Palma A, Varela Pinzon L, Santarelli F, Simonutti R, Mauri P, Porretti L, Campione M, Aquino D, Monguzzi A, Torrente Y. Magnetic-field-driven targeting of exosomes modulates immune and metabolic changes in dystrophic muscle. NATURE NANOTECHNOLOGY 2024; 19:1532-1543. [PMID: 39039121 PMCID: PMC11486659 DOI: 10.1038/s41565-024-01725-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 06/18/2024] [Indexed: 07/24/2024]
Abstract
Exosomes are promising therapeutics for tissue repair and regeneration to induce and guide appropriate immune responses in dystrophic pathologies. However, manipulating exosomes to control their biodistribution and targeting them in vivo to achieve adequate therapeutic benefits still poses a major challenge. Here we overcome this limitation by developing an externally controlled delivery system for primed annexin A1 myo-exosomes (Exomyo). Effective nanocarriers are realized by immobilizing the Exomyo onto ferromagnetic nanotubes to achieve controlled delivery and localization of Exomyo to skeletal muscles by systemic injection using an external magnetic field. Quantitative muscle-level analyses revealed that macrophages dominate the uptake of Exomyo from these ferromagnetic nanotubes in vivo to synergistically promote beneficial muscle responses in a murine animal model of Duchenne muscular dystrophy. Our findings provide insights into the development of exosome-based therapies for muscle diseases and, in general, highlight the formulation of effective functional nanocarriers aimed at optimizing exosome biodistribution.
Collapse
Grants
- Regione Lombardia (Region of Lombardy)
- Fondazione Telethon (Telethon Foundation)
- RF-2016-02362263 "Multimodal nanotracking for exosome-based therapy in DMD" (theory enhancing) “At the origin of congenital muscular dystrophy: shedding light on the Tdark proteins DPM2 and DPM3”, Bando “Cariplo Telethon Alliance GJC2021” 2022
- Multiomics pRofiling of patient spEcific Models to predict druggable targets in severe neuromuscular rare diseases (REMODEL)”, Unmet Medical Needs, Fondazione Regionale per la Ricerca Biomedica (FRRB), 2022 Nanoparticles in Freidreich Ataxia” National Center for Gene Therapy and Drugs based on RNA Technology, Spoke #1: Genetic diseases, PNRR CN3 RNA, 2022
- “Isolamento di nanoparticelle naturali da utilizzare come agenti anti-infiammatori/anti-fibrotici”, 5X1000, Fondazione Patrimonio e dalla Direzione Scientifica Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico (2022)
- PNRR CN3 RNA, 2022, PNRR project ANTHEM: AdvaNced Technologies for Human-centrEd Medicine - PNC0000003 Spoke #2 – NextGenerationEU RF-2016-02362263 "Multimodal nanotracking for exosome-based therapy in DMD" (theory enhancing)
Collapse
Affiliation(s)
- Chiara Villa
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Valeria Secchi
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Mirco Macchi
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Luxembourg Centre for Systems Biomedicine, Department of Biomedical Data Science, Luxembourg City, Luxembourg
| | - Luana Tripodi
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Elena Trombetta
- Flow Cytometry Service, Clinical Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Desiree Zambroni
- Advanced Light and Electron Microscopy Bioimaging Center ALEMBIC, San Raffaele Scientific Institute - OSR, Milan, Italy
| | - Francesco Padelli
- Department of Neuroradiology, IRCCS Foundation Neurological Institute 'Carlo Besta', Milan, Italy
| | - Michele Mauri
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Monica Molinaro
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Rebecca Oddone
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonella De Palma
- National Research Council of Italy, Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
- Clinical Proteomics Laboratory, ITB-CNR, CNR.Biomics Infrastructure, Elixir, Milan, Italy
| | - Laura Varela Pinzon
- Veterinary Medicine, Department Clinical Sciences, Equine Sciences, Equine Musculoskeletal Biology. Utrecht University, Utrecht, Netherlands
| | - Federica Santarelli
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Roberto Simonutti
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - PierLuigi Mauri
- National Research Council of Italy, Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
- Clinical Proteomics Laboratory, ITB-CNR, CNR.Biomics Infrastructure, Elixir, Milan, Italy
| | - Laura Porretti
- Flow Cytometry Service, Clinical Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marcello Campione
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
- Department of Earth and Environmental Sciences, University of Milano Bicocca, Milano, Italy
| | - Domenico Aquino
- Department of Neuroradiology, IRCCS Foundation Neurological Institute 'Carlo Besta', Milan, Italy
| | - Angelo Monguzzi
- Department of Materials Science, University of Milano Bicocca, Milan, Italy
- NANOMIB, Nanomedicine Center, University of Milano Bicocca, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
26
|
Li F, Zhang F, Shi H, Xia H, Wei X, Liu S, Wu T, Li Y, Shu F, Chen M, Li J, Duan R. Aerobic exercise suppresses CCN2 secretion from senescent muscle stem cells and boosts muscle regeneration in aged mice. J Cachexia Sarcopenia Muscle 2024; 15:1733-1749. [PMID: 38925632 PMCID: PMC11446704 DOI: 10.1002/jcsm.13526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/18/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Aging negatively impacts tissue repair, particularly in skeletal muscle, where the regenerative capacity of muscle stem cells (MuSCs) diminishes with age. Although aerobic exercise is known to attenuate skeletal muscle atrophy, its specific impact on the regenerative and repair capacity of MuSCs remains unclear. METHODS Mice underwent moderate-intensity continuous training (MICT) from 9 months (aged + Ex-9M) or 20 months (aged + Ex-20M) to 25 months, with age-matched (aged) and adult controls. Histological examinations and MuSC transplantation assays assessed aerobic exercise effects on MuSC function and muscle regeneration. CCN2/connective tissue growth factor modulation (overexpression and knockdown) in MuSCs and AICAR supplementation effects were explored. RESULTS Aged mice displayed significantly reduced running duration (65.33 ± 4.32 vs. 161.9 ± 1.29 min, mean ± SD, P < 0.001) and distance (659.17 ± 103.64 vs. 3058.28 ± 46.26 m, P < 0.001) compared with adults. This reduction was accompanied by skeletal muscle weight loss and decreased myofiber cross-sectional area (CSA). However, MICT initiated at 9 or 20 months led to a marked increase in running duration (142.75 ± 3.14 and 133.86 ± 20.47 min, respectively, P < 0.001 compared with aged mice) and distance (2347.58 ± 145.11 and 2263 ± 643.87 m, respectively, P < 0.001). Additionally, MICT resulted in increased skeletal muscle weight and enhanced CSA. In a muscle injury model, aged mice exhibited fewer central nuclear fibres (CNFs; 266.35 ± 68.66/mm2), while adult, aged + Ex-9M and aged + Ex-20M groups showed significantly higher CNF counts (610.82 ± 46.76, 513.42 ± 47.19 and 548.29 ± 71.82/mm2, respectively; P < 0.001 compared with aged mice). MuSCs isolated from aged mice displayed increased CCN2 expression, which was effectively suppressed by MICT. Transplantation of MuSCs overexpressing CCN2 (Lenti-CCN2, Lenti-CON as control) into injured tibialis anterior muscle compromised regeneration capacity, resulting in significantly fewer CNFs in the Lenti-CCN2 group compared with Lenti-CON (488.07 ± 27.63 vs. 173.99 ± 14.28/mm2, P < 0.001) at 7 days post-injury (dpi). Conversely, knockdown of CCN2 (Lenti-CCN2shR, Lenti-NegsiR as control) in aged MuSCs improved regeneration capacity, significantly increasing the CNF count from 254.5 ± 26.36 to 560.39 ± 48.71/mm2. Lenti-CCN2 MuSCs also increased fibroblast proliferation and exacerbated skeletal muscle fibrosis, while knockdown of CCN2 in aged MuSCs mitigated this pattern. AICAR supplementation, mimicking exercise, replicated the beneficial effects of aerobic exercise by mitigating muscle weight decline, enhancing satellite cell activity and reducing fibrosis. CONCLUSIONS Aerobic exercise effectively reverses the decline in endurance capacity and mitigates muscle atrophy in aged mice. It inhibits CCN2 secretion from senescent MuSCs, thereby enhancing skeletal muscle regeneration and preventing fibrosis in aged mice. AICAR supplementation mimics the beneficial effects of aerobic exercise.
Collapse
Affiliation(s)
- Fan Li
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Fulong Zhang
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
- School of Physical EducationShanxi Datong UniversityDatongChina
| | - Haiwang Shi
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Honglin Xia
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Xiaobei Wei
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Siqi Liu
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Tao Wu
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Yuecheng Li
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Feng Shu
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Mengjie Chen
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Jie Li
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| | - Rui Duan
- School of Physical Education and Sports ScienceSouth China Normal UniversityGuangzhou510006China
| |
Collapse
|
27
|
Smirnova O, Efremov Y, Klyucherev T, Peshkova M, Senkovenko A, Svistunov A, Timashev P. Direct and cell-mediated EV-ECM interplay. Acta Biomater 2024; 186:63-84. [PMID: 39043290 DOI: 10.1016/j.actbio.2024.07.029] [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: 02/02/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Extracellular vesicles (EV) are a heterogeneous group of lipid particles excreted by cells. They play an important role in regeneration, development, inflammation, and cancer progression, together with the extracellular matrix (ECM), which they constantly interact with. In this review, we discuss direct and indirect interactions of EVs and the ECM and their impact on different physiological processes. The ECM affects the secretion of EVs, and the properties of the ECM and EVs modulate EVs' diffusion and adhesion. On the other hand, EVs can affect the ECM both directly through enzymes and indirectly through the modulation of the ECM synthesis and remodeling by cells. This review emphasizes recently discovered types of EVs bound to the ECM and isolated by enzymatic digestion, including matrix-bound nanovesicles (MBV) and tissue-derived EV (TiEV). In addition to the experimental studies, computer models of the EV-ECM-cell interactions, from all-atom models to quantitative pharmacology models aiming to improve our understanding of the interaction mechanisms, are also considered. STATEMENT OF SIGNIFICANCE: Application of extracellular vesicles in tissue engineering is an actively developing area. Vesicles not only affect cells themselves but also interact with the matrix and change it. The matrix also influences both cells and vesicles. In this review, different possible types of interactions between vesicles, matrix, and cells are discussed. Furthermore, the united EV-ECM system and its regulation through the cellular activity are presented.
Collapse
Affiliation(s)
- Olga Smirnova
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Yuri Efremov
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Timofey Klyucherev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 119991 Moscow, Russia
| | - Alexey Senkovenko
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | | | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, 119991 Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia.
| |
Collapse
|
28
|
Lin KH, Hibbert JE, Flynn CG, Lemens JL, Torbey MM, Steinert ND, Flejsierowicz PM, Melka KM, Lindley GT, Lares M, Setaluri V, Wagers AJ, Hornberger TA. Satellite cell-derived TRIM28 is pivotal for mechanical load- and injury-induced myogenesis. EMBO Rep 2024; 25:3812-3841. [PMID: 39143258 PMCID: PMC11387408 DOI: 10.1038/s44319-024-00227-1] [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: 01/04/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/16/2024] Open
Abstract
Satellite cells are skeletal muscle stem cells that contribute to postnatal muscle growth, and they endow skeletal muscle with the ability to regenerate after a severe injury. Here we discover that this myogenic potential of satellite cells requires a protein called tripartite motif-containing 28 (TRIM28). Interestingly, different from the role reported in a previous study based on C2C12 myoblasts, multiple lines of both in vitro and in vivo evidence reveal that the myogenic function of TRIM28 is not dependent on changes in the phosphorylation of its serine 473 residue. Moreover, the functions of TRIM28 are not mediated through the regulation of satellite cell proliferation or differentiation. Instead, our findings indicate that TRIM28 regulates the ability of satellite cells to progress through the process of fusion. Specifically, we discover that TRIM28 controls the expression of a fusogenic protein called myomixer and concomitant fusion pore formation. Collectively, the outcomes of this study expose the framework of a novel regulatory pathway that is essential for myogenesis.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jamie E Hibbert
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Corey Gk Flynn
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Jake L Lemens
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Melissa M Torbey
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Nathaniel D Steinert
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Philip M Flejsierowicz
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Kiley M Melka
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Garrison T Lindley
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Marcos Lares
- Department of Dermatology, University of Wisconsin - Madison, Madison, WI, USA
| | | | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Joslin Diabetes Center, Boston, MA, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA.
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
29
|
Ismaeel A, Peck BD, Montgomery MM, Burke BI, Goh J, Kang G, Franco AB, Xia Q, Goljanek-Whysall K, McDonagh B, McLendon JM, Koopmans PJ, Jacko D, Schaaf K, Bloch W, Gehlert S, Wen Y, Murach KA, Peterson CA, Boudreau RL, Fisher-Wellman KH, McCarthy JJ. microRNA-1 Regulates Metabolic Flexibility in Skeletal Muscle via Pyruvate Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607377. [PMID: 39149347 PMCID: PMC11326265 DOI: 10.1101/2024.08.09.607377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
MicroRNA-1 (miR-1) is the most abundant miRNA in adult skeletal muscle. To determine the function of miR-1 in adult skeletal muscle, we generated an inducible, skeletal muscle-specific miR-1 knockout (KO) mouse. Integration of RNA-sequencing (RNA-seq) data from miR-1 KO muscle with Argonaute 2 enhanced crosslinking and immunoprecipitation sequencing (AGO2 eCLIP-seq) from human skeletal muscle identified miR-1 target genes involved with glycolysis and pyruvate metabolism. The loss of miR-1 in skeletal muscle induced cancer-like metabolic reprogramming, as shown by higher pyruvate kinase muscle isozyme M2 (PKM2) protein levels, which promoted glycolysis. Comprehensive bioenergetic and metabolic phenotyping combined with skeletal muscle proteomics and metabolomics further demonstrated that miR-1 KO induced metabolic inflexibility as a result of pyruvate oxidation resistance. While the genetic loss of miR-1 reduced endurance exercise performance in mice and in C. elegans, the physiological down-regulation of miR-1 expression in response to a hypertrophic stimulus in both humans and mice causes a similar metabolic reprogramming that supports muscle cell growth. Taken together, these data identify a novel post-translational mechanism of adult skeletal muscle metabolism regulation mediated by miR-1.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Bailey D Peck
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - McLane M Montgomery
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - Benjamin I Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Jensen Goh
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Gyumin Kang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Abigail B Franco
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Mass Spectrometry and Proteomics Core, University of Kentucky, Lexington, KY, USA
| | - Qin Xia
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Katarzyna Goljanek-Whysall
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Jared M McLendon
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Pieter J Koopmans
- Department Health, Human Performance, & Recreation, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA
| | - Daniel Jacko
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Olympic Base Center, North Rhine-Westphalia/Rhineland, Cologne, Germany
| | - Kirill Schaaf
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Olympic Base Center, North Rhine-Westphalia/Rhineland, Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports Medicine, German Sport University, Cologne, Germany
- Department for the Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department Health, Human Performance, & Recreation, University of Arkansas, Fayetteville, AR, USA
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, AR, USA
| | - Charlotte A Peterson
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Ryan L Boudreau
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kelsey H Fisher-Wellman
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
30
|
Fernández‐Rhodes M, Buchan E, Gagnon SD, Qian J, Gethings L, Lees R, Peacock B, Capel AJ, Martin NRW, Oppenheimer PG, Lewis MP, Davies OG. Extracellular vesicles may provide an alternative detoxification pathway during skeletal muscle myoblast ageing. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e171. [PMID: 39169919 PMCID: PMC11336379 DOI: 10.1002/jex2.171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/20/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024]
Abstract
Skeletal muscle (SM) acts as a secretory organ, capable of releasing myokines and extracellular vesicles (SM-EVs) that impact myogenesis and homeostasis. While age-related changes have been previously reported in murine SM-EVs, no study has comprehensively profiled SM-EV in human models. To this end, we provide the first comprehensive comparison of SM-EVs from young and old human primary skeletal muscle cells (HPMCs) to map changes associated with SM ageing. HPMCs, isolated from young (24 ± 1.7 years old) and older (69 ± 2.6 years old) participants, were immunomagnetically sorted based on the presence of the myogenic marker CD56 (N-CAM) and cultured as pure (100% CD56+) or mixed populations (MP: 90% CD56+). SM-EVs were isolated using an optimised protocol combining ultrafiltration and size exclusion chromatography (UF + SEC) and their biological content was extensively characterised using Raman spectroscopy (RS) and liquid chromatography mass spectrometry (LC-MS). Minimal variations in basic EV parameters (particle number, size, protein markers) were observed between young and old populations. However, biochemical fingerprinting by RS highlighted increased protein (amide I), lipid (phospholipids and phosphatidylcholine) and hypoxanthine signatures for older SM-EVs. Through LC-MS, we identified 84 shared proteins with functions principally related to cell homeostasis, muscle maintenance and transcriptional regulation. Significantly, SM-EVs from older participants were comparatively enriched in proteins involved in oxidative stress and DNA/RNA mutagenesis, such as E3 ubiquitin-protein ligase TTC3 (TTC3), little elongation complex subunit 1 (ICE1) and Acetyl-CoA carboxylase 1 (ACACA). These data suggest SM-EVs could provide an alternative pathway for homeostasis and detoxification during SM ageing.
Collapse
Affiliation(s)
| | - Emma Buchan
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamBirminghamUK
| | - Stephanie D. Gagnon
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| | - Jiani Qian
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| | - Lee Gethings
- Waters CorporationWilmslowUK
- School of Biological SciencesUniversity of ManchesterManchesterUK
- Medical SchoolUniversity of SurreySurreyUK
| | | | - Ben Peacock
- School of Biological SciencesUniversity of ManchesterManchesterUK
| | - Andrew J. Capel
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| | - Neil R. W. Martin
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamBirminghamUK
| | - Mark P. Lewis
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| | - Owen G. Davies
- School of SportExercise and Health Sciences, Loughborough UniversityLoughboroughUK
| |
Collapse
|
31
|
Hour TC, Lan Nhi NT, Lai IJ, Chuu CP, Lin PC, Chang HW, Su YF, Chen CH, Chen YK. Kaempferol-Enhanced Migration and Differentiation of C2C12 Myoblasts via ITG1B/FAK/Paxillin and IGF1R/AKT/mTOR Signaling Pathways. Mol Nutr Food Res 2024; 68:e2300685. [PMID: 38860356 DOI: 10.1002/mnfr.202300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/30/2024] [Indexed: 06/12/2024]
Abstract
SCOPE Kaempferol (KMP), a bioactive flavonoid compound found in fruits and vegetables, contributes to human health in many ways but little is known about its relationship with muscle mass. The effect of KMP on C2C12 myoblast differentiation and the mechanisms that might underlie that effect are studied. METHODS AND RESULTS This study finds that KMP (1, 10 µM) increases the migration and differentiation of C2C12 myoblasts in vitro. Studying the possible mechanism underlying its effect on migration, the study finds that KMP activates Integrin Subunit Beta 1 (ITGB1) in C2C12 myoblasts, increasing p-FAK (Tyr398) and its downstream cell division cycle 42 (CDC42), a protein previously associated with cell migration. Regarding differentiation, KMP upregulates the expression of myosin heavy chain (MHC) and activates IGF1/AKT/mTOR/P70S6K. Interestingly, pretreatment with an AKT inhibitor (LY294002) and siRNA knockdown of IGF1R leads to a decrease in cell differentiation, suggesting that IGF1/AKT activation is required for KMP to induce C2C12 myoblast differentiation. CONCLUSION Together, the findings suggest that KMP enhances the migration and differentiation of C2C12 myoblasts through the ITG1B/FAK/paxillin and IGF1R/AKT/mTOR pathways. Thus, KMP supplementation might potentially be used to prevent or delay age-related loss of muscle mass and help maintain muscle health.
Collapse
Affiliation(s)
- Tzyh-Chyuan Hour
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807378, Taiwan
| | - Nguyen Thai Lan Nhi
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - I-Ju Lai
- Department of Nutrition, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 350401, Taiwan
| | - Pei-Chen Lin
- Department of Oral Hygiene, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Hsi-Wen Chang
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Ying-Fang Su
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center and Department of Orthopedics, Kaohsiung Medical University Hospital and Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Yu-Kuei Chen
- Department of Nutrition, I-Shou University, Kaohsiung, 82445, Taiwan
| |
Collapse
|
32
|
Cappelli E, Ravera S, Bertola N, Grilli F, Squillario M, Regis S, Degan P. Advanced Analysis and Validation of a microRNA Signature for Fanconi Anemia. Genes (Basel) 2024; 15:820. [PMID: 39062599 PMCID: PMC11276059 DOI: 10.3390/genes15070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Some years ago, we reported the generation of a Fanconi anemia (FA) microRNA signature. This study aims to develop an analytical strategy to select a smaller and more reliable set of molecules that could be tested for potential benefits for the FA phenotype, elucidate its biochemical and molecular mechanisms, address experimental activity, and evaluate its possible impact on FA therapy. In silico analyses of the data obtained in the original study were thoroughly processed and anenrichment analysis was employed to identify the classes of genes that are over-represented in the FA-miRNA population under study. Primary bone marrow mononuclear cells (MNCs) from sixFA patients and sixhealthy donors as control samples were employed in the study. RNAs containing the small RNA fractions were reverse-transcribed and real-time PCR was performed in triplicate using the specific primers. Experiments were performed in triplicate.The in-silico analysis reported six miRNAs as likely contributors to the complex pathological spectrum of FA. Among these, three miRNAs were validated by real-time PCR. Primary bone marrow mononuclear cells (MNCs) reported a significant reduction in the expression level of miRNA-1246 and miRNA-206 in the FA samples in comparison to controls.This study highlights several biochemical pathways as culprits in the phenotypic manifestations and the pathophysiological mechanisms acting in FA. A relatively low number of miRNAs appear involved in all these different phenotypes, demonstrating the extreme plasticity of the gene expression modulation. This study further highlights miR-206 as a pivotal player in regulatory functions and signaling in the bone marrow mesenchymal stem cell (BMSC) process in FA. Due to this evidence, the activity of miR-206 in FA deserves specific experimental scrutiny. The results, here presented, might be relevant in the management of FA.
Collapse
Affiliation(s)
- Enrico Cappelli
- Haematology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via De Toni 14, 16132 Genova, Italy;
| | - Nadia Bertola
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, L. go R. Benzi 10, 16132 Genoa, Italy;
| | - Federica Grilli
- Haematology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | | | - Stefano Regis
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | - Paolo Degan
- Mutagenesi e Prevenzione Oncologica Unit—IRCCS Ospedale Policlinico San Martino, L. go R. Benzi 10, 16132 Genoa, Italy;
| |
Collapse
|
33
|
Carata E, Muci M, Di Giulio S, Di Giulio T, Mariano S, Panzarini E. The Neuromuscular Disorder Mediated by Extracellular Vesicles in Amyotrophic Lateral Sclerosis. Curr Issues Mol Biol 2024; 46:5999-6017. [PMID: 38921029 PMCID: PMC11202069 DOI: 10.3390/cimb46060358] [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: 03/28/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) represents a neurodegenerative disorder characterized by the progressive loss of both upper and lower motor neurons, resulting in muscular atrophy and eventual paralysis. While much research has concentrated on investigating the impact of major mutations associated with ALS on motor neurons and central nervous system (CNS) cells, recent studies have unveiled that ALS pathogenesis extends beyond CNS imbalances, encompassing dysregulation in other tissues such as skeletal muscle. Evidence from animal models and patients supports this broader perspective. Skeletal muscle, once considered solely as an effector organ, is now recognized as possessing significant secretory activity capable of influencing motor neuron survival. However, the precise cellular and molecular mechanisms underlying the detrimental effects observed in muscle and its associated structures in ALS remain poorly understood. Additionally, emerging data suggest that extracellular vesicles (EVs) may play a role in the establishment and function of the neuromuscular junction (NMJ) under both physiological and pathological conditions and in wasting and regeneration of skeletal muscles, particularly in neurodegenerative diseases like ALS. This review aims to explore the key findings about skeletal muscle involvement in ALS, shedding light on the potential underlying mechanisms and contributions of EVs and their possible application for the design of biosensors.
Collapse
Affiliation(s)
- Elisabetta Carata
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (E.C.); (M.M.); (T.D.G.); (S.M.)
| | - Marco Muci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (E.C.); (M.M.); (T.D.G.); (S.M.)
| | - Simona Di Giulio
- Department of Mathematics and Physics, University of Salento, 73100 Lecce, Italy;
| | - Tiziano Di Giulio
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (E.C.); (M.M.); (T.D.G.); (S.M.)
| | - Stefania Mariano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (E.C.); (M.M.); (T.D.G.); (S.M.)
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (E.C.); (M.M.); (T.D.G.); (S.M.)
| |
Collapse
|
34
|
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
|
35
|
Fennel ZJ, Bourrant P, Kurian AS, Petrocelli JJ, de Hart NMMP, Yee EM, Boudina S, Keirstead HS, Nistor G, Greilach SA, Berchtold NC, Lane TE, Drummond MJ. Stem cell secretome treatment improves whole-body metabolism, reduces adiposity, and promotes skeletal muscle function in aged mice. Aging Cell 2024; 23:e14144. [PMID: 38500398 PMCID: PMC11296109 DOI: 10.1111/acel.14144] [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: 09/12/2023] [Revised: 02/09/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024] Open
Abstract
Aging coincides with the progressive loss of muscle mass and strength, increased adiposity, and diminished physical function. Accordingly, interventions aimed at improving muscle, metabolic, and/or physical health are of interest to mitigate the adverse effects of aging. In this study, we tested a stem cell secretome product, which contains extracellular vesicles and growth, cytoskeletal remodeling, and immunomodulatory factors. We examined the effects of 4 weeks of 2×/week unilateral intramuscular secretome injections (quadriceps) in ambulatory aged male C57BL/6 mice (22-24 months) compared to saline-injected aged-matched controls. Secretome delivery substantially increased whole-body lean mass and decreased fat mass, corresponding to higher myofiber cross-sectional area and smaller adipocyte size, respectively. Secretome-treated mice also had greater whole-body physical function (grip strength and rotarod performance) and had higher energy expenditure and physical activity levels compared to control mice. Furthermore, secretome-treated mice had greater skeletal muscle Pax7+ cell abundance, capillary density, collagen IV turnover, reduced intramuscular lipids, and greater Akt and hormone sensitive lipase phosphorylation in adipose tissue. Finally, secretome treatment in vitro directly enhanced muscle cell growth and IL-6 production, and in adipocytes, it reduced lipid content and improved insulin sensitivity. Moreover, indirect treatment with secretome-treated myotube culture media also enhanced muscle cell growth and adipocyte size reduction. Together, these data suggest that intramuscular treatment with a stem cell secretome improves whole-body metabolism, physical function, and remodels skeletal muscle and adipose tissue in aged mice.
Collapse
Affiliation(s)
- Zachary J. Fennel
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | - Paul‐Emile Bourrant
- Division of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Anu Susan Kurian
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | - Jonathan J. Petrocelli
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | | | - Elena M. Yee
- Division of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Sihem Boudina
- Division of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | | | | | | | | | - Thomas E. Lane
- Immunis, Inc.IrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Micah J. Drummond
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
- Division of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
36
|
Duran P, Yang BA, Plaster E, Eiken M, Loebel C, Aguilar CA. Tracking of Nascent Matrix Deposition during Muscle Stem Cell Activation across Lifespan Using Engineered Hydrogels. Adv Biol (Weinh) 2024; 8:e2400091. [PMID: 38616175 DOI: 10.1002/adbi.202400091] [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: 02/18/2024] [Revised: 03/22/2024] [Indexed: 04/16/2024]
Abstract
Adult stem cells occupy a niche that contributes to their function, but how stem cells rebuild their microenvironment after injury remains an open-ended question. Herein, biomaterial-based systems and metabolic labeling are utilized to evaluate how skeletal muscle stem cells deposit extracellular matrix. Muscle stem cells and committed myoblasts are observed to generate less nascent matrix than muscle resident fibro-adipogenic progenitors. When cultured on substrates that matched the stiffness of physiological uninjured and injured muscles, muscle stem cells increased nascent matrix deposition with activation kinetics. Reducing the ability to deposit nascent matrix by an inhibitor of vesicle trafficking (Exo-1) attenuated muscle stem cell function and mimicked impairments observed from muscle stem cells isolated from old muscles. Old muscle stem cells are observed to deposit less nascent matrix than young muscle stem cells, which is rescued with therapeutic supplementation of insulin-like growth factors. These results highlight the role of nascent matrix production with muscle stem cell activation.
Collapse
Affiliation(s)
- Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin A Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleanor Plaster
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madeline Eiken
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
37
|
Guvatova ZG, Kobelyatskaya AA, Kudasheva ER, Pudova EA, Bulavkina EV, Churov AV, Tkacheva ON, Moskalev AA. Matrisome Transcriptome Dynamics during Tissue Aging. Life (Basel) 2024; 14:593. [PMID: 38792614 PMCID: PMC11121957 DOI: 10.3390/life14050593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is a complex three-dimensional network of macromolecules that provides structural support for the cells and plays a significant role in tissue homeostasis and repair. Growing evidence indicates that dysregulation of ECM remodeling contributes to various pathological conditions in the body, including age-associated diseases. In this work, gene expression data of normal human tissues obtained from the Genotype-Tissue Expression project, as well as data from MatrisomeDB 2.0, the ECM-protein knowledge database, are used to estimate the age-dependent matrisome transcriptome dynamics in the blood, heart, brain, liver, kidneys, lungs, and muscle. Differential gene expression (DE) analysis revealed dozens of matrisome genes encoding both structural elements of the ECM and ECM-associated proteins, which had a tissue-specific expression profile with age. Among common DE genes that changed expression with age in at least three tissues, COL18A1, MFAP1, IGFBP7, AEBP1, LTBP2, LTBP4, LG14, EFEMP1, PRELP, BGN, FAM20B, CTSC, CTSS, and CLEC2B were observed. The findings of the study also reveal that there are sex-specific alterations during aging in the matrisome gene expression. Taken together, the results obtained in this work may help in understanding the role of the ECM in tissue aging and might prove valuable for the future development of the field of ECM research in general.
Collapse
Affiliation(s)
- Zulfiya G. Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia
| | | | - Eveline R. Kudasheva
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia
| | - Elena A. Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elizaveta V. Bulavkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey V. Churov
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia
| | - Olga N. Tkacheva
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia
| | - Alexey A. Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia
| |
Collapse
|
38
|
Ruple BA, Mattingly ML, Godwin JS, McIntosh MC, Kontos NJ, Agyin-Birikorang A, Michel JM, Plotkin DL, Chen SY, Ziegenfuss TN, Fruge AD, Gladden LB, Robinson AT, Mobley CB, Mackey AL, Roberts MD. The effects of resistance training on denervated myofibers, senescent cells, and associated protein markers in middle-aged adults. FASEB J 2024; 38:e23621. [PMID: 38651653 PMCID: PMC11047210 DOI: 10.1096/fj.202302103rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Denervated myofibers and senescent cells are hallmarks of skeletal muscle aging. However, sparse research has examined how resistance training affects these outcomes. We investigated the effects of unilateral leg extensor resistance training (2 days/week for 8 weeks) on denervated myofibers, senescent cells, and associated protein markers in apparently healthy middle-aged participants (MA, 55 ± 8 years old, 17 females, 9 males). We obtained dual-leg vastus lateralis (VL) muscle cross-sectional area (mCSA), VL biopsies, and strength assessments before and after training. Fiber cross-sectional area (fCSA), satellite cells (Pax7+), denervated myofibers (NCAM+), senescent cells (p16+ or p21+), proteins associated with denervation and senescence, and senescence-associated secretory phenotype (SASP) proteins were analyzed from biopsy specimens. Leg extensor peak torque increased after training (p < .001), while VL mCSA trended upward (interaction p = .082). No significant changes were observed for Type I/II fCSAs, NCAM+ myofibers, or senescent (p16+ or p21+) cells, albeit satellite cells increased after training (p = .037). While >90% satellite cells were not p16+ or p21+, most p16+ and p21+ cells were Pax7+ (>90% on average). Training altered 13 out of 46 proteins related to muscle-nerve communication (all upregulated, p < .05) and 10 out of 19 proteins related to cellular senescence (9 upregulated, p < .05). Only 1 out of 17 SASP protein increased with training (IGFBP-3, p = .031). In conclusion, resistance training upregulates proteins associated with muscle-nerve communication in MA participants but does not alter NCAM+ myofibers. Moreover, while training increased senescence-related proteins, this coincided with an increase in satellite cells but not alterations in senescent cell content or SASP proteins. These latter findings suggest shorter term resistance training is an unlikely inducer of cellular senescence in apparently healthy middle-aged participants. However, similar study designs are needed in older and diseased populations before definitive conclusions can be drawn.
Collapse
Affiliation(s)
| | | | | | | | | | | | - J. Max Michel
- School of Kinesiology, Auburn University, Auburn, AL, USA
| | | | | | | | | | | | | | | | - Abigail L. Mackey
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | |
Collapse
|
39
|
Kim TK, Gil HY. Effects of Paraspinal Intramuscular Injection of Atelocollagen in Patients with Chronic Low Back Pain: A Retrospective Observational Study. J Clin Med 2024; 13:2607. [PMID: 38731135 PMCID: PMC11084233 DOI: 10.3390/jcm13092607] [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: 03/12/2024] [Revised: 04/21/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Background/Objectives: Atelocollagen is used for soft tissue repair and reconstruction by replacing defective or damaged muscles, membranes, ligaments, and tendons. This study aimed to evaluate the clinical efficacy and safety of additional paraspinal intramuscular injection of atelocollagen on lumbar epidural steroid injection for reducing pain and improving functional capacity of patients with chronic low back pain (CLBP). Methods: We retrospectively enrolled 608 consecutive patients with CLBP who received lumbar epidural steroid injection with or without additional paraspinal intramuscular injection of atelocollagen. The Numerical Rating Scale and the Oswestry Disability Index were used to assess pain and functional capacity, respectively, before the procedure, and three months after the injection. Also, we analyzed the relationship between the additional paraspinal intramuscular injection of atelocollagen and the success rate. Results: Both Numerical Rating Scale and the Oswestry Disability Index scores were significantly reduced in both groups at three months after injection. However, there was a significant difference between the two groups. Furthermore, the success rate was significantly higher in the additional paraspinal intramuscular injection of atelocollagen group. Conclusions: This study's results showed that additional paraspinal intramuscular injection of atelocollagen on lumbar epidural steroid injection reduced pain and improved functional capacity for patients with CLBP. Therefore, the paraspinal intramuscular injection of atelocollagen may be a promising option for the treatment of patients with CLBP.
Collapse
Affiliation(s)
- Tae Kwang Kim
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Ho Young Gil
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Gumi Hospital, Soonchunhyang University College of Medicine, Gumi 39371, Republic of Korea
| |
Collapse
|
40
|
Fard D, Barbiera A, Dobrowolny G, Tamagnone L, Scicchitano BM. Semaphorins: Missing Signals in Age-dependent Alteration of Neuromuscular Junctions and Skeletal Muscle Regeneration. Aging Dis 2024; 15:517-534. [PMID: 37728580 PMCID: PMC10917540 DOI: 10.14336/ad.2023.0801] [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: 05/03/2023] [Accepted: 08/01/2023] [Indexed: 09/21/2023] Open
Abstract
Skeletal muscle is characterized by a remarkable capacity to rearrange after physiological changes and efficiently regenerate. However, during aging, extensive injury, or pathological conditions, the complete regenerative program is severely affected, with a progressive loss of muscle mass and function, a condition known as sarcopenia. The compromised tissue repair program is attributable to the gradual depletion of stem cells and to altered regulatory signals. Defective muscle regeneration can severely affect re-innervation by motor axons, and neuromuscular junctions (NMJs) development, ultimately leading to skeletal muscle atrophy. Defects in NMJ formation and maintenance occur physiologically during aging and are responsible for the pathogenesis of several neuromuscular disorders. However, it is still largely unknown how neuromuscular connections are restored on regenerating fibers. It has been suggested that attractive and repelling signals used for axon guidance could be implicated in this process; in particular, guidance molecules called semaphorins play a key role. Semaphorins are a wide family of extracellular regulatory signals with a multifaceted role in cell-cell communication. Originally discovered as axon guidance factors, they have been implicated in cancer progression, embryonal organogenesis, skeletal muscle innervation, and other physiological and developmental functions in different tissues. In particular, in skeletal muscle, specific semaphorin molecules are involved in the restoration and remodeling of the nerve-muscle connections, thus emphasizing their plausible role to ensure the success of muscle regeneration. This review article aims to discuss the impact of aging on skeletal muscle regeneration and NMJs remodeling and will highlight the most recent insights about the role of semaphorins in this context.
Collapse
Affiliation(s)
- Damon Fard
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Alessandra Barbiera
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Gabriella Dobrowolny
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy.
| | - Luca Tamagnone
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| | - Bianca Maria Scicchitano
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| |
Collapse
|
41
|
Yin Y, He GJ, Hu S, Tse EHY, Cheung TH. Muscle stem cell niche dynamics during muscle homeostasis and regeneration. Curr Top Dev Biol 2024; 158:151-177. [PMID: 38670704 DOI: 10.1016/bs.ctdb.2024.02.008] [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] [Indexed: 04/28/2024]
Abstract
The process of skeletal muscle regeneration involves a coordinated interplay of specific cellular and molecular interactions within the injury site. This review provides an overview of the cellular and molecular components in regenerating skeletal muscle, focusing on how these cells or molecules in the niche regulate muscle stem cell functions. Dysfunctions of muscle stem cell-to-niche cell communications during aging and disease will also be discussed. A better understanding of how niche cells coordinate with muscle stem cells for muscle repair will greatly aid the development of therapeutic strategies for treating muscle-related disorders.
Collapse
Affiliation(s)
- Yishu Yin
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China
| | - Gary J He
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China
| | - Shenyuan Hu
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China
| | - Erin H Y Tse
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, P.R. China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, P.R. China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, P.R. China.
| |
Collapse
|
42
|
Kwon Y. YAP/TAZ as Molecular Targets in Skeletal Muscle Atrophy and Osteoporosis. Aging Dis 2024; 16:AD.2024.0306. [PMID: 38502585 PMCID: PMC11745433 DOI: 10.14336/ad.2024.0306] [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: 01/09/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
Skeletal muscles and bones are closely connected anatomically and functionally. Age-related degeneration in these tissues is associated with physical disability in the elderly and significantly impacts their quality of life. Understanding the mechanisms of age-related musculoskeletal tissue degeneration is crucial for identifying molecular targets for therapeutic interventions for skeletal muscle atrophy and osteoporosis. The Hippo pathway is a recently identified signaling pathway that plays critical roles in development, tissue homeostasis, and regeneration. The Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the mammalian Hippo signaling pathway. This review highlights the fundamental roles of YAP and TAZ in the homeostatic maintenance and regeneration of skeletal muscles and bones. YAP/TAZ play a significant role in stem cell function by relaying various environmental signals to stem cells. Skeletal muscle atrophy and osteoporosis are related to stem cell dysfunction or senescence triggered by YAP/TAZ dysregulation resulting from reduced mechanosensing and mitochondrial function in stem cells. In contrast, the maintenance of YAP/TAZ activation can suppress stem cell senescence and tissue dysfunction and may be used as a basis for the development of potential therapeutic strategies. Thus, targeting YAP/TAZ holds significant therapeutic potential for alleviating age-related muscle and bone dysfunction and improving the quality of life in the elderly.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Yu Y, Su Y, Wang G, Lan M, Liu J, Garcia Martin R, Brandao BB, Lino M, Li L, Liu C, Kahn CR, Meng Q. Reciprocal communication between FAPs and muscle cells via distinct extracellular vesicle miRNAs in muscle regeneration. Proc Natl Acad Sci U S A 2024; 121:e2316544121. [PMID: 38442155 PMCID: PMC10945765 DOI: 10.1073/pnas.2316544121] [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: 09/27/2023] [Accepted: 02/06/2024] [Indexed: 03/07/2024] Open
Abstract
Muscle regeneration is a complex process relying on precise teamwork between multiple cell types, including muscle stem cells (MuSCs) and fibroadipogenic progenitors (FAPs). FAPs are also the main source of intramuscular adipose tissue (IMAT). Muscles without FAPs exhibit decreased IMAT infiltration but also deficient muscle regeneration, indicating the importance of FAPs in the repair process. Here, we demonstrate the presence of bidirectional crosstalk between FAPs and MuSCs via their secretion of extracellular vesicles (EVs) containing distinct clusters of miRNAs that is crucial for normal muscle regeneration. Thus, after acute muscle injury, there is activation of FAPs leading to a transient rise in IMAT. These FAPs also release EVs enriched with a selected group of miRNAs, a number of which come from an imprinted region on chromosome 12. The most abundant of these is miR-127-3p, which targets the sphingosine-1-phosphate receptor S1pr3 and activates myogenesis. Indeed, intramuscular injection of EVs from immortalized FAPs speeds regeneration of injured muscle. In late stages of muscle repair, in a feedback loop, MuSCs and their derived myoblasts/myotubes secrete EVs enriched in miR-206-3p and miR-27a/b-3p. The miRNAs repress FAP adipogenesis, allowing full muscle regeneration. Together, the reciprocal communication between FAPs and muscle cells via miRNAs in their secreted EVs plays a critical role in limiting IMAT infiltration while stimulating muscle regeneration, hence providing an important mechanism for skeletal muscle repair and homeostasis.
Collapse
Affiliation(s)
- Yingying Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Yang Su
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
- Department of Cell Biology, Third Military Medical University, Chongqing400038, China
| | - Guoxiao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Miaomiao Lan
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
| | - Jin Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
| | - Ruben Garcia Martin
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Bruna Brasil Brandao
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Marsel Lino
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Lei Li
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
| | - Chang Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA02215
| | - Qingyong Meng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, Department of Genetics and Molecular biology, China Agricultural University, Beijing100193, China
| |
Collapse
|
44
|
Li Z, Hou D, Tang Z, Xiong L, Yan Y. The potential role of stem cells-derived extracellular vesicles in the treatment of musculoskeletal system diseases. Cell Biol Int 2024; 48:237-252. [PMID: 38100269 DOI: 10.1002/cbin.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/29/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
The therapeutic potential of stem cells-derived extracellular vesicles (EVs) has shown a great progress in the regenerative medicine. EVs are rich in a variety of bioactive substances, which are important carriers of signal transmission and interactions between cells, and they play an important role in the processes of tissue repair and regeneration. Several studies have shown that stem cells-derived EVs regulate immunity, promote cell proliferation and differentiation, enhance bone and vascular regeneration, and play an increasingly important role in musculoskeletal system. This review aimed to describe the biological characteristics of stem cells-derived EVs and discuss their potential role in the therapy of musculoskeletal system diseases.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Demiao Hou
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Zijin Tang
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Lishun Xiong
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yiguo Yan
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
45
|
Cao X, Xue L, Yu X, Yan Y, Lu J, Luo X, Wang H, Wang J. Myogenic exosome miR-140-5p modulates skeletal muscle regeneration and injury repair by regulating muscle satellite cells. Aging (Albany NY) 2024; 16:4609-4630. [PMID: 38428405 PMCID: PMC10968704 DOI: 10.18632/aging.205617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Muscle satellite cells (SCs) play a crucial role in the regeneration and repair of skeletal muscle injuries. Previous studies have shown that myogenic exosomes can enhance satellite cell proliferation, while the expression of miR-140-5p is significantly reduced during the repair process of mouse skeletal muscle injuries induced by BaCl2. This study aims to investigate the potential of myogenic exosomes carrying miR-140-5p inhibitors to activate SCs and influence the regeneration of injured muscles. Myogenic progenitor cell exosomes (MPC-Exo) and contained miR-140-5p mimics/inhibitors myogenic exosomes (MPC-Exo140+ and MPC-Exo140-) were employed to treat SCs and use the model. The results demonstrate that miR-140-5p regulates SC proliferation by targeting Pax7. Upon the addition of MPC-Exo and MPC-Exo140-, Pax7 expression in SCs significantly increased, leading to the transition of the cell cycle from G1 to S phase and an enhancement in cell proliferation. Furthermore, the therapeutic effect of MPC-Exo140- was validated in animal model, where the expression of muscle growth-related genes substantially increased in the gastrocnemius muscle. Our research demonstrates that MPC-Exo140- can effectively activate dormant muscle satellite cells, initiating their proliferation and differentiation processes, ultimately leading to the formation of new skeletal muscle cells and promoting skeletal muscle repair and remodeling.
Collapse
Affiliation(s)
- Xiaorui Cao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Linli Xue
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiuju Yu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Juan Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
46
|
Li DCW, Rudloff S, Langer HT, Norman K, Herpich C. Age-Associated Differences in Recovery from Exercise-Induced Muscle Damage. Cells 2024; 13:255. [PMID: 38334647 PMCID: PMC10854791 DOI: 10.3390/cells13030255] [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: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Understanding the intricate mechanisms governing the cellular response to resistance exercise is paramount for promoting healthy aging. This narrative review explored the age-related alterations in recovery from resistance exercise, focusing on the nuanced aspects of exercise-induced muscle damage in older adults. Due to the limited number of studies in older adults that attempt to delineate age differences in muscle discovery, we delve into the multifaceted cellular influences of chronic low-grade inflammation, modifications in the extracellular matrix, and the role of lipid mediators in shaping the recovery landscape in aging skeletal muscle. From our literature search, it is evident that aged muscle displays delayed, prolonged, and inefficient recovery. These changes can be attributed to anabolic resistance, the stiffening of the extracellular matrix, mitochondrial dysfunction, and unresolved inflammation as well as alterations in satellite cell function. Collectively, these age-related impairments may impact subsequent adaptations to resistance exercise. Insights gleaned from this exploration may inform targeted interventions aimed at enhancing the efficacy of resistance training programs tailored to the specific needs of older adults, ultimately fostering healthy aging and preserving functional independence.
Collapse
Affiliation(s)
- Donna Ching Wah Li
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Stefan Rudloff
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| | | | - Kristina Norman
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Catrin Herpich
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| |
Collapse
|
47
|
Zhang J, Gao Y, Yan J. Roles of Myokines and Muscle-Derived Extracellular Vesicles in Musculoskeletal Deterioration under Disuse Conditions. Metabolites 2024; 14:88. [PMID: 38392980 PMCID: PMC10891558 DOI: 10.3390/metabo14020088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
Prolonged inactivity and disuse conditions, such as those experienced during spaceflight and prolonged bedrest, are frequently accompanied by detrimental effects on the motor system, including skeletal muscle atrophy and bone loss, which greatly increase the risk of osteoporosis and fractures. Moreover, the decrease in glucose and lipid utilization in skeletal muscles, a consequence of muscle atrophy, also contributes to the development of metabolic syndrome. Clarifying the mechanisms involved in disuse-induced musculoskeletal deterioration is important, providing therapeutic targets and a scientific foundation for the treatment of musculoskeletal disorders under disuse conditions. Skeletal muscle, as a powerful endocrine organ, participates in the regulation of physiological and biochemical functions of local or distal tissues and organs, including itself, in endocrine, autocrine, or paracrine manners. As a motor organ adjacent to muscle, bone tissue exhibits a relative lag in degenerative changes compared to skeletal muscle under disuse conditions. Based on this phenomenon, roles and mechanisms involved in the communication between skeletal muscle and bone, especially from muscle to bone, under disuse conditions have attracted widespread attention. In this review, we summarize the roles and regulatory mechanisms of muscle-derived myokines and extracellular vesicles (EVs) in the occurrence of muscle atrophy and bone loss under disuse conditions, as well as discuss future perspectives based on existing research.
Collapse
Affiliation(s)
- Jie Zhang
- Institute of Special Medicine, Shanxi Medical University, Jinzhong 030619, China;
| | - Yunfang Gao
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Jiangwei Yan
- Institute of Special Medicine, Shanxi Medical University, Jinzhong 030619, China;
| |
Collapse
|
48
|
Hidalgo-Alvarez V, Madl CM. Leveraging Biomaterial Platforms to Study Aging-Related Neural and Muscular Degeneration. Biomolecules 2024; 14:69. [PMID: 38254669 PMCID: PMC10813704 DOI: 10.3390/biom14010069] [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: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Aging is a complex multifactorial process that results in tissue function impairment across the whole organism. One of the common consequences of this process is the loss of muscle mass and the associated decline in muscle function, known as sarcopenia. Aging also presents with an increased risk of developing other pathological conditions such as neurodegeneration. Muscular and neuronal degeneration cause mobility issues and cognitive impairment, hence having a major impact on the quality of life of the older population. The development of novel therapies that can ameliorate the effects of aging is currently hindered by our limited knowledge of the underlying mechanisms and the use of models that fail to recapitulate the structure and composition of the cell microenvironment. The emergence of bioengineering techniques based on the use of biomimetic materials and biofabrication methods has opened the possibility of generating 3D models of muscular and nervous tissues that better mimic the native extracellular matrix. These platforms are particularly advantageous for drug testing and mechanistic studies. In this review, we discuss the developments made in the creation of 3D models of aging-related neuronal and muscular degeneration and we provide a perspective on the future directions for the field.
Collapse
Affiliation(s)
| | - Christopher M. Madl
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
49
|
Miyauchi Y, Kiyama M, Soga S, Nanri H, Ogiwara T, Yonamine S, Kon R, Ikarashi N, Chiba Y, Hosoe T, Sakai H. Downregulation of Genes for Skeletal Muscle Extracellular Matrix Components by Cisplatin. Biol Pharm Bull 2024; 47:1846-1850. [PMID: 39522978 DOI: 10.1248/bpb.b24-00428] [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] [Indexed: 11/16/2024]
Abstract
The extracellular matrix (ECM) in skeletal muscle is involved in a variety of physiological functions beyond the mechanical support of muscle tissue, nerves, and blood vessels; however, the role of the ECM in skeletal muscle remains unclear. There is little information regarding changes in the expression of factors comprising the ECM during cisplatin-induced muscle atrophy. In the present study, we examined the changes in gene expressions for skeletal muscle extracellular matrix components in skeletal muscle during cisplatin-induced muscle atrophy. Intraperitoneal administration of cisplatin caused muscle atrophy in mice and during this cisplatin-induced muscle atrophy, the expression of many procollagen genes (Col1a1, Col1a2, Col3a1, Col4a1, Col5a1, and Col5a2), elastin (Eln), fibronectin (Fn1), Laminin (Lama1, Lama2, and Lamb1) decorin (Dcn), heparan sulphate proteoglycans (Hspg2) and integrin (Itgb1) constituting the ECM was suppressed. Additional studies are needed to elucidate the pathological significance and mechanisms of reduced gene expression of ECM components associated with cisplatin-induced muscle atrophy.
Collapse
Affiliation(s)
- Yu Miyauchi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Miho Kiyama
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Shinki Soga
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Hayato Nanri
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Takayuki Ogiwara
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Shiori Yonamine
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Risako Kon
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Nobutomo Ikarashi
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, School of Pharmacy, Hoshi University
| | - Tomoo Hosoe
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
- Department of Bioregulatory Science, School of Pharmacy, Hoshi University
| | - Hiroyasu Sakai
- Department of Biomolecular Pharmacology, School of Pharmacy, Hoshi University
| |
Collapse
|
50
|
Lin KH, Hibbert JE, Lemens JL, Torbey MM, Steinert ND, Flejsierowicz PM, Melka KM, Lares M, Setaluri V, Hornberger TA. The role of satellite cell-derived TRIM28 in mechanical load- and injury-induced myogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572566. [PMID: 38187693 PMCID: PMC10769277 DOI: 10.1101/2023.12.20.572566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Satellite cells are skeletal muscle stem cells that contribute to postnatal muscle growth, and they endow skeletal muscle with the ability to regenerate after a severe injury. Here we discovered that this myogenic potential of satellite cells requires a protein called tripartite motif-containing 28 (TRIM28). Unexpectedly, multiple lines of both in vitro and in vivo evidence revealed that the myogenic function of TRIM28 is not dependent on changes in the phosphorylation of its serine 473 residue. Moreover, the functions of TRIM28 were not mediated through the regulation of satellite cell proliferation or differentiation. Instead, our findings indicate that TRIM28 regulates the ability of satellite cells to progress through the process of fusion. Specifically, we discovered that TRIM28 controls the expression of a fusogenic protein called myomixer and concomitant fusion pore formation. Collectively, the outcomes of this study expose the framework of a novel regulatory pathway that is essential for myogenesis.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jamie E. Hibbert
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Jake L. Lemens
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Melissa M. Torbey
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Nathaniel D. Steinert
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Philip M. Flejsierowicz
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Kiley M. Melka
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Marcos Lares
- Department of Dermatology, University of Wisconsin - Madison, WI, USA
| | | | - Troy A. Hornberger
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| |
Collapse
|