1
|
Gustafsson T, Ulfhake B. Aging Skeletal Muscles: What Are the Mechanisms of Age-Related Loss of Strength and Muscle Mass, and Can We Impede Its Development and Progression? Int J Mol Sci 2024; 25:10932. [PMID: 39456714 PMCID: PMC11507513 DOI: 10.3390/ijms252010932] [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: 09/03/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
As we age, we lose muscle strength and power, a condition commonly referred to as sarcopenia (ICD-10-CM code (M62.84)). The prevalence of sarcopenia is about 5-10% of the elderly population, resulting in varying degrees of disability. In this review we emphasise that sarcopenia does not occur suddenly. It is an aging-induced deterioration that occurs over time and is only recognised as a disease when it manifests clinically in the 6th-7th decade of life. Evidence from animal studies, elite athletes and longitudinal population studies all confirms that the underlying process has been ongoing for decades once sarcopenia has manifested. We present hypotheses about the mechanism(s) underlying this process and their supporting evidence. We briefly review various proposals to impede sarcopenia, including cell therapy, reducing senescent cells and their secretome, utilising targets revealed by the skeletal muscle secretome, and muscle innervation. We conclude that although there are potential candidates and ongoing preclinical and clinical trials with drug treatments, the only evidence-based intervention today for humans is exercise. We present different exercise programmes and discuss to what extent the interindividual susceptibility to developing sarcopenia is due to our genetic predisposition or lifestyle factors.
Collapse
Affiliation(s)
| | - Brun Ulfhake
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden;
| |
Collapse
|
2
|
Sakai H, Uno H, Yamakawa H, Tanaka K, Ikedo A, Uezumi A, Ohkawa Y, Imai Y. The androgen receptor in mesenchymal progenitors regulates skeletal muscle mass via Igf1 expression in male mice. Proc Natl Acad Sci U S A 2024; 121:e2407768121. [PMID: 39292748 PMCID: PMC11441553 DOI: 10.1073/pnas.2407768121] [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: 04/18/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024] Open
Abstract
Androgens exert their effects primarily by binding to the androgen receptor (AR), a ligand-dependent nuclear receptor. While androgens have anabolic effects on skeletal muscle, previous studies reported that AR functions in myofibers to regulate skeletal muscle quality, rather than skeletal muscle mass. Therefore, the anabolic effects of androgens are exerted via nonmyofiber cells. In this context, the cellular and molecular mechanisms of AR in mesenchymal progenitors, which play a crucial role in maintaining skeletal muscle homeostasis, remain largely unknown. In this study, we demonstrated expression of AR in mesenchymal progenitors and found that targeted AR ablation in mesenchymal progenitors reduced limb muscle mass in mature adult, but not young or aged, male mice, although fatty infiltration of muscle was not affected. The absence of AR in mesenchymal progenitors led to remarkable perineal muscle hypotrophy, regardless of age, due to abnormal regulation of transcripts associated with cell death and extracellular matrix organization. Additionally, we revealed that AR in mesenchymal progenitors regulates the expression of insulin-like growth factor 1 (Igf1) and that IGF1 administration prevents perineal muscle atrophy in a paracrine manner. These findings indicate that the anabolic effects of androgens regulate skeletal muscle mass via, at least in part, AR signaling in mesenchymal progenitors.
Collapse
Affiliation(s)
- Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Hideaki Uno
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Harumi Yamakawa
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Aoi Ikedo
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| |
Collapse
|
3
|
Zhao C, Ikeya M. Novel insights from human induced pluripotent stem cells on origins and roles of fibro/adipogenic progenitors as heterotopic ossification precursors. Front Cell Dev Biol 2024; 12:1457344. [PMID: 39286484 PMCID: PMC11402712 DOI: 10.3389/fcell.2024.1457344] [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: 06/30/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Fibro/adipogenic progenitors (FAPs) that reside in muscle tissue are crucial for muscular homeostasis and regeneration as they secrete signaling molecules and components of the extracellular matrix. During injury or disease, FAPs differentiate into different cell types and significantly modulate muscular function. Recent advances in lineage tracing and single-cell transcriptomics have proven that FAPs are heterogeneous both in resting and post-injury or disease states. Their heterogeneity may be owing to the varied tissue microenvironments and their diverse developmental origins. Therefore, understanding FAPs' developmental origins can help predict their characteristics and behaviors under different conditions. FAPs are thought to be the major cell populations in the muscle connective tissue (MCT). During embryogenesis, the MCT directs muscular development throughout the body and serves as a prepattern for muscular morphogenesis. The developmental origins of FAPs as stromal cells in the MCT were studied previously. In adult tissues, FAPs are important precursors for heterotopic ossification, especially in the context of the rare genetic disorder fibrodysplasia ossificans progressiva. A new developmental origin for FAPs have been suggested that differs from conventional developmental perspectives. In this review, we summarize the developmental origins and functions of FAPs as stromal cells of the MCT and present novel insights obtained by using patient-derived induced pluripotent stem cells and mouse models of heterotopic ossification. This review broadens the current understanding of FAPs and suggests potential avenues for further investigation.
Collapse
Affiliation(s)
- Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Redden JT, Deng J, Cohen DJ, Schwartz Z, McClure MJ. Muscle Fibrosis, NF-κB, and TGF-β Are Differentially Altered in Two Models of Paralysis (Botox Versus Neurectomy). Adv Wound Care (New Rochelle) 2024. [PMID: 38877804 DOI: 10.1089/wound.2024.0045] [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: 06/16/2024] Open
Abstract
Objective: Volumetric muscle loss results in intramuscular axotomy, denervating muscle distal to the injury and leading to paralysis, denervation, and loss of muscle function. Once the nerve is damaged, paralyzed skeletal muscle will atrophy and accumulate noncontractile connective tissue. The objective of this study was to determine differences in connective tissue, atrophy, and inflammatory signaling between two paralysis models, botulinum toxin (Botox), which blocks acetylcholine transmission while keeping nerves intact, and neurectomy, which eliminates all nerve-to-muscle signaling. Approach: Twenty male Sprague Dawley rats were randomized and received a sciatic-femoral neurectomy (SFN), Botox-induced muscle paralysis of the proximal femur muscles, quadriceps femoris, hamstrings, and calf muscles (BTX), or sham. Muscle force was measured 52 days postsurgery, and samples were collected for histology, protein, and mRNA assays. Results: SFN and BTX decreased twitch and tetanic force, decreased fiber size by twofold, and increased myogenic expression compared with controls. SFN increased the levels of all major extracellular matrix proteins correlating with fibrosis [e.g., laminin, fibronectin, and collagen type(s) I, III, VI]. SFN also increased profibrotic and proinflammatory mRNA compared with BTX and controls. Innovation: SFN and BTX were similar in gross morphology and functional deficiencies. However, SFN exhibited a higher amount of fibrosis in histological sections and immunoblotting. The present study shows evidence that nerve signaling changes NF-κB and TGF-β signaling, warranting future studies to determine the mechanisms involved. Conclusion: These data indicate that nerve signaling may influence fibrogenesis following denervation, but the mechanisms involved may differ as a function of the method of paralysis.
Collapse
Affiliation(s)
- James T Redden
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jingyao Deng
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - David J Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Orthopedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
5
|
Cai CW, Grey JA, Hubmacher D, Han WM. Biomaterial-Based Regenerative Strategies for Volumetric Muscle Loss: Challenges and Solutions. Adv Wound Care (New Rochelle) 2024. [PMID: 38775429 DOI: 10.1089/wound.2024.0079] [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: 07/11/2024] Open
Abstract
Significance: Volumetric muscle loss (VML) is caused by the loss of significant amounts of skeletal muscle tissue. VML cannot be repaired by intrinsic regenerative processes, resulting in permanent loss of muscle function and disability. Current rehabilitative-focused treatment strategies lack efficacy and do not restore muscle function, indicating the need for the development of effective regenerative strategies. Recent Advances: Recent developments implicate biomaterial-based approaches for promoting muscle repair and functional restoration post-VML. Specifically, bioscaffolds transplanted in the injury site have been utilized to mimic endogenous cues of the ablated tissue to promote myogenic pathways, increase neo-myofiber synthesis, and ultimately restore contractile function to the injured unit. Critical Issues: Despite the development and preclinical testing of various biomaterial-based regenerative strategies, effective therapies for patients are not available. The unique challenges posed for biomaterial-based treatments of VML injuries, including its scalability and clinical applicability beyond small-animal models, impede progress. Furthermore, production of tissue-engineered constructs is technically demanding, with reproducibility issues at scale and complexities in achieving vascularization and innervation of large constructs. Future Directions: Biomaterial-based regenerative strategies designed to comprehensively address the pathophysiology of VML are needed. Considerations for clinical translation, including scalability and regulatory compliance, should also be considered when developing such strategies. In addition, an integrated approach that combines regenerative and rehabilitative strategies is essential for ensuring functional improvement.
Collapse
Affiliation(s)
- Charlene W Cai
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Biology, The College of New Jersey, Ewing, New Jersey, USA
| | - Josh A Grey
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dirk Hubmacher
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Woojin M Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
6
|
Loomis T, Kulkarni VA, Villalba M, Davids JR, Leach JK, Smith LR. Muscle satellite cells and fibro-adipogenic progenitors from muscle contractures of children with cerebral palsy have impaired regenerative capacity. Dev Med Child Neurol 2024. [PMID: 38937924 DOI: 10.1111/dmcn.16006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
AIM To evaluate the mechanosensitivity of muscle satellite cells (MuSCs) and fibro-adipogenic progenitors (FAPs) in cerebral palsy (CP) and the efficacy of the drug verteporfin in restoring cells' regenerative capacity. METHOD Muscle biopsies were collected from six children with CP and six typically developing children. MuSCs and FAPs were isolated and plated on collagen-coated polyacrylamide gels at stiffnesses of 0.2 kPa, 8 kPa, and 25 kPa. Cells were treated with verteporfin to block mechanosensing or with dimethyl sulfoxide as a negative control. MuSC differentiation and FAP activation into myofibroblasts were measured using immunofluorescence staining. RESULTS Surprisingly, MuSC differentiation was not affected by stiffness; however, stiff substrates resulted in large myonuclear clustering. Across all stiffnesses, MuSCs from children with CP had less differentiation than those of their typically developing counterparts. FAP activation into myofibroblasts was significantly higher in children with CP than their typically developing peers, but was not affected by stiffness. Verteporfin did not affect differentiation or activation in either cell population, but slightly decreased myonuclear clustering on stiff substrates. INTERPRETATION Cells from children with CP were less regenerative and more fibrotic compared to those of their typically developing counterparts, with MuSCs being sensitive to increases in stiffness. Therefore, the mechanosensitivity of MuSCs and FAPs may represent a new target to improve differentiation and activation in CP muscle.
Collapse
Affiliation(s)
- Taryn Loomis
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, CA, USA
| | - Vedant A Kulkarni
- Department of Orthopaedic Surgery, Shriners Children's Northern California, Sacramento, CA, USA
| | - Marie Villalba
- Department of Orthopaedic Surgery, Shriners Children's Northern California, Sacramento, CA, USA
| | - Jon R Davids
- Department of Orthopaedic Surgery, Shriners Children's Northern California, Sacramento, CA, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - Lucas R Smith
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, CA, USA
- Department of Physical Medicine and Rehabilitation, UC Davis Health, Sacramento, CA, USA
| |
Collapse
|
7
|
Xu HR, Le VV, Oprescu SN, Kuang S. Muscle stem cells as immunomodulator during regeneration. Curr Top Dev Biol 2024; 158:221-238. [PMID: 38670707 DOI: 10.1016/bs.ctdb.2024.01.010] [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 skeletal muscle is well known for its remarkable ability to regenerate after injuries. The regeneration is a complex and dynamic process that involves muscle stem cells (also called muscle satellite cells, MuSCs), fibro-adipogenic progenitors (FAPs), immune cells, and other muscle-resident cell populations. The MuSCs are the myogenic cell populaiton that contribute nuclei directly to the regenerated myofibers, while the other cell types collaboratively establish a microenvironment that facilitates myogenesis of MuSCs. The myogenic process includes activation, proliferation and differentiationof MuSCs, and subsequent fusion their descendent mononuclear myocytes into multinuclear myotubes. While the contributions of FAPs and immune cells to this microenvironment have been well studied, the influence of MuSCs on other cell types remains poorly understood. This review explores recent evidence supporting the potential role of MuSCs as immunomodulators during muscle regeneration, either through cytokine production or ligand-receptor interactions.
Collapse
Affiliation(s)
- H Rex Xu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Victor V Le
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States; Purdue University Institute for Cancer Research, West Lafayette, IN, United States.
| |
Collapse
|
8
|
Jiang H, Liu B, Lin J, Xue T, Han Y, Lu C, Zhou S, Gu Y, Xu F, Shen Y, Xu L, Sun H. MuSCs and IPCs: roles in skeletal muscle homeostasis, aging and injury. Cell Mol Life Sci 2024; 81:67. [PMID: 38289345 PMCID: PMC10828015 DOI: 10.1007/s00018-023-05096-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Skeletal muscle is a highly specialized tissue composed of myofibres that performs crucial functions in movement and metabolism. In response to external stimuli and injuries, a range of stem/progenitor cells, with muscle stem cells or satellite cells (MuSCs) being the predominant cell type, are rapidly activated to repair and regenerate skeletal muscle within weeks. Under normal conditions, MuSCs remain in a quiescent state, but become proliferative and differentiate into new myofibres in response to injury. In addition to MuSCs, some interstitial progenitor cells (IPCs) such as fibro-adipogenic progenitors (FAPs), pericytes, interstitial stem cells expressing PW1 and negative for Pax7 (PICs), muscle side population cells (SPCs), CD133-positive cells and Twist2-positive cells have been identified as playing direct or indirect roles in regenerating muscle tissue. Here, we highlight the heterogeneity, molecular markers, and functional properties of these interstitial progenitor cells, and explore the role of muscle stem/progenitor cells in skeletal muscle homeostasis, aging, and muscle-related diseases. This review provides critical insights for future stem cell therapies aimed at treating muscle-related diseases.
Collapse
Affiliation(s)
- Haiyan Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tong Xue
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Yimin Han
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Lingchi Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Demarbaix T, Daele UV, Meirte J, Anthonissen M, Maertens K, Moortgat P. Possible benefits of food supplementation or diet in scar management: A scoping review. Scars Burn Heal 2024; 10:20595131241282105. [PMID: 39280762 PMCID: PMC11402062 DOI: 10.1177/20595131241282105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Aim The evidence regarding a potential role of food supplementation as an adjunct therapy in scar aftercare is limited. In this scoping review we aim to provide an overview of the possible beneficial role of supplementations in aftercare settings. Method After formulating the research question and accompanying key words, a comprehensive search for relevant publications was performed using PubMed and Web of Science. Two authors independently identified and checked each study against the inclusion criteria. All data was collected and summarized for further discussion. Results After screening, 11 studies were included in the qualitative synthesis. Four studies including human subjects showed a promising connection between scar improvement and supplementation of vitamin D, omega-3 fatty-acids or a Solanaceae-free diet and lower omega-6 fatty-acid intake. Most of the studies were performed on in-vitro models. Preliminary evidence confirmed the beneficial role of vitamin D. Curcumin- and quercetin-supplementation were linked to decreased fibroblast proliferation. Vitamin C enhanced collagen production in healthy as well as keloidal dermal fibroblasts. Chitin stimulated cell-proliferation in human fibroblasts and keratinocytes. Conclusion The findings suggest early potential benefits of additional food supplementation in scar management for scars but provide no clear evidence. To establish guidelines or gather more evidence on food supplementation, studies involving human subjects (in vivo) are essential. The intricacies associated with nutritional studies in vivo present multifaceted challenges. It should be emphasized that substantial additional evidence is required before aspects such as timing and dosage of supplementation could be addressed for clinical application. Lay Summary Aim: This scoping review looks at whether taking food supplements might help with scar care alongside standard scar management following burn injury. Little information is thought to be available on this subject. An up-to-date review of the literature was undertaken to assimilate the body of evidence and determine if a consensus could be drawn.Method: A specific research question was designed and search conducted in scientific databases like PubMed and Web of Science. Two of our team members carefully selected and reviewed each study to determine which studies met the inclusion or exclusion criteria. All studies that met the inclusion criteria were then reviewed and the information collated to enable conclusions to be drawn.Results: Eleven studies met the inclusion criteria and were used to formulate the conclusions drawn. Four studies showed that taking vitamin D, omega-3 fatty acids, a diet without certain vegetables (Solanaceae), and eating less omega-6 fatty acids might help improve scars. It is important to note that most studies (seven out of 11) were carried out in a laboratory and not with real people. These lab studies showed that vitamin D might be helpful. Supplements like curcumin and quercetin seemed to slow down the growth of skin cells like fibroblasts and keratinocytes. Vitamin C aided collagen synthesis, which is important for healthy skin, in both normal and keloid scar cells. Another substance, chitin, was also found to help skin cells and keratinocytes grow better.Conclusion: Our findings point to some early possible benefits of taking extra nutrient supplements for managing scars but do not provide clear evidence. More research is required to enable the development of supplement recommendation and guidelines to be produced. Future research should focus on human trials but do keep in mind that carrying out supplement studies with people is more complicated. The evidence provided by this scoping review is insufficient to recommend the intake of any supplements or the imposition of dietary restrictions for the purpose of managing scars.
Collapse
Affiliation(s)
- Thibau Demarbaix
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
| | - Ulrike Van Daele
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
| | - Jill Meirte
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
| | - Mieke Anthonissen
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Koen Maertens
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Vrije Universiteit Brussel, Clinical and Lifespan Psychology, Brussels, Belgium
| | - Peter Moortgat
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
| |
Collapse
|
10
|
Brightwell CR, Latham CM, Keeble AR, Thomas NT, Owen AM, Reeves KA, Long DE, Patrick M, Gonzalez-Velez S, Abed V, Annamalai RT, Jacobs C, Conley CE, Hawk GS, Stone AV, Fry JL, Thompson KL, Johnson DL, Noehren B, Fry CS. GDF8 inhibition enhances musculoskeletal recovery and mitigates posttraumatic osteoarthritis following joint injury. SCIENCE ADVANCES 2023; 9:eadi9134. [PMID: 38019905 PMCID: PMC10686569 DOI: 10.1126/sciadv.adi9134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Musculoskeletal disorders contribute substantially to worldwide disability. Anterior cruciate ligament (ACL) tears result in unresolved muscle weakness and posttraumatic osteoarthritis (PTOA). Growth differentiation factor 8 (GDF8) has been implicated in the pathogenesis of musculoskeletal degeneration following ACL injury. We investigated GDF8 levels in ACL-injured human skeletal muscle and serum and tested a humanized monoclonal GDF8 antibody against a placebo in a mouse model of PTOA (surgically induced ACL tear). In patients, muscle GDF8 was predictive of atrophy, weakness, and periarticular bone loss 6 months following surgical ACL reconstruction. In mice, GDF8 antibody administration substantially mitigated muscle atrophy, weakness, and fibrosis. GDF8 antibody treatment rescued the skeletal muscle and articular cartilage transcriptomic response to ACL injury and attenuated PTOA severity and deficits in periarticular bone microarchitecture. Furthermore, GDF8 genetic deletion neutralized musculoskeletal deficits in response to ACL injury. Our findings support an opportunity for rapid targeting of GDF8 to enhance functional musculoskeletal recovery and mitigate the severity of PTOA after injury.
Collapse
Affiliation(s)
- Camille R. Brightwell
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Christine M. Latham
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Alexander R. Keeble
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Nicholas T. Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Allison M. Owen
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Kelsey A. Reeves
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Douglas E. Long
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Matthew Patrick
- Department of Biomedical Engineering, College of Engineering, University of Kentucky, Lexington, KY, USA
| | | | - Varag Abed
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Ramkumar T. Annamalai
- Department of Biomedical Engineering, College of Engineering, University of Kentucky, Lexington, KY, USA
| | - Cale Jacobs
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Caitlin E. Conley
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Gregory S. Hawk
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Austin V. Stone
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jean L. Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Katherine L. Thompson
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Darren L. Johnson
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Brian Noehren
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Christopher S. Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
11
|
Ma R, Feng L, Wu P, Liu Y, Ren HM, Li SW, Tang L, Zhong CB, Han D, Zhang WB, Tang JY, Zhou XQ, Jiang WD. A new insight on copper: Promotion of collagen synthesis and myofiber growth and development in juvenile grass carp ( Ctenopharyngodon idella). ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:22-33. [PMID: 37771856 PMCID: PMC10522946 DOI: 10.1016/j.aninu.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 09/30/2023]
Abstract
Copper (Cu) is a trace element, essential for fish growth. In the current study, in addition to growth performance, we first explored the effects of Cu on collagen synthesis and myofiber growth and development in juvenile grass carp (Ctenopharyngodon idella). A total of 1080 fish (11.16 ± 0.01 g) were randomly divided into 6 treatments (3 replicates per treatment) to receive five doses of organic Cu, which were Cu citrate (CuCit) at 0.99 (basal diet), 2.19, 4.06, 6.15, and 8.07 mg/kg, and one dose of inorganic Cu (CuSO4·5H2O at 3.15 mg/kg), for 9 weeks. The results showed appropriate Cu level (4.06 mg/kg) enhanced growth performance, improved nutritional Cu status, and downregulated Cu-transporting ATPase 1 mRNA levels in the hepatopancreas, intestine, and muscle of juvenile grass carp. Meanwhile, collagen content in fish muscle was increased after Cu intake, which was probably due to the following pathways: (1) activating CTGF/TGF-β1/Smads signaling pathway to regulate collagen transcription; (2) upregulating of La ribonucleoprotein domain family 6 (LARP6) mRNA levels to regulate translation initiation; (3) increasing proline hydroxylase, lysine hydroxylase, and lysine oxidase activities to regulate posttranslational modifications. In addition, optimal Cu group increased myofiber diameters and the frequency of myofibers with diameter >50 μm, which might be associated with upregulation of cyclin B, cyclin D, cyclin E, proliferating cell nuclear antigen, myogenic determining factor (MyoD), myogenic factor 5, myogenin (MyoG), myogenic regulatory factor 4 and myosin heavy chain (MyHC) and downregulation of myostatin mRNA levels, increasing protein levels of MyoD, MyoG and MyHC in fish muscle. Finally, based on percentage weight gain (PWG), serum ceruloplasmin (Cp) activity and collagen content in fish muscle, Cu requirements were determined as 4.74, 4.37 and 4.62 mg/kg diet (CuCit as Cu source) of juvenile grass carp, respectively. Based on PWG and Cp activity, compared to CuSO4·5H2O, the efficacy of CuCit were 131.80% and 115.38%, respectively. Our findings provide new insights into Cu supplementation to promote muscle growth in fish, and help improve the overall productivity of aquaculture.
Collapse
Affiliation(s)
- Rui Ma
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Cheng-Bo Zhong
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Dong Han
- State Key Laboratory of Fresh Water Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Wen-Bing Zhang
- The Key Laboratory of Mariculture, Ministry of Education, The Key Laboratory of Aquaculture Nutrition and Feeds, Ministry of Agriculture, Ocean University of China, Qingdao, 266003, China
| | - Jia-Yong Tang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| |
Collapse
|
12
|
Mathieu M, Girousse A, Sengenès C. [What if the origin of FAPs was contributing to their heterogeneity in muscle?]. Med Sci (Paris) 2023; 39 Hors série n° 1:15-21. [PMID: 37975765 DOI: 10.1051/medsci/2023129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Fibro-adipogenic progenitors (FAPs) are resident mesenchymal stromal cells (MSCs) of skeletal muscle. They play a crucial role in muscle homeostasis and regeneration through their paracrine activity. Recent technological advances in single-cell RNA sequencing have allowed the characterization of the heterogeneity within this cell population. In this article, we will present the different subpopulations of FAPs under basal, injury, or degenerative conditions, as well as their associated functions in mice and humans. We will then discuss the potential extramuscular origin of a post-injury FAP population. Indeed, our recent work demonstrates that MSCs from adipose tissue, infiltrating the muscle, could contribute to FAP heterogeneity.
Collapse
Affiliation(s)
- Maxime Mathieu
- Institut RESTORE, UMR Inserm 1301 / CNRS 5070, Toulouse, France
| | | | | |
Collapse
|
13
|
Loomis T, Smith LR. Thrown for a loop: fibro-adipogenic progenitors in skeletal muscle fibrosis. Am J Physiol Cell Physiol 2023; 325:C895-C906. [PMID: 37602412 DOI: 10.1152/ajpcell.00245.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
Fibro-adipogenic progenitors (FAPs) are key regulators of skeletal muscle regeneration and homeostasis. However, dysregulation of these cells leads to fibro-fatty infiltration across various muscle diseases. FAPs are the key source of extracellular matrix (ECM) deposition in muscle, and disruption to this process leads to a pathological accumulation of ECM, known as fibrosis. The replacement of contractile tissue with fibrotic ECM functionally impairs the muscle and increases muscle stiffness. FAPs and fibrotic muscle form a progressively degenerative feedback loop where, as a muscle becomes fibrotic, it induces a fibrotic FAP phenotype leading to further development of fibrosis. In this review, we summarize FAPs' role in fibrosis in terms of their activation, heterogeneity, contributions to fibrotic degeneration, and role across musculoskeletal diseases. We also discuss current research on potential therapeutic avenues to attenuate fibrosis by targeting FAPs.
Collapse
Affiliation(s)
- Taryn Loomis
- Biomedical Engineering Graduate Group, University of California, Davis, California, United States
| | - Lucas R Smith
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California, United States
- Department of Physical Medicine and Rehabilitation, University of California, Davis, California, United States
| |
Collapse
|
14
|
Zhao P, Liu X, Feng L, Jiang WD, Wu P, Liu Y, Ren HM, Jin XW, Yang J, Zhou XQ. New perspective on mechanism in muscle toxicity of ochratoxin A: Model of juvenile grass carp (Ctenopharyngodon idella). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 263:106701. [PMID: 37776711 DOI: 10.1016/j.aquatox.2023.106701] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Ochratoxin A (OTA) is a common fungal toxin that pollutes raw materials of aquatic feeds (such as corn, soybean meal, and wheat). This study explored the effects of OTA through diet on muscle toxicity in juvenile grass carp (Ctenopharyngodon idella). The following results were obtained for the muscle. (1) With an increase in dietary OTA, the residue of OTA in muscle increased, muscle fiber diameter and density decreased, and even muscle fiber breakage. (2) OTA caused oxidative stress by downregulating GPx1 (a, b) and Trx via inhibited the PGC1-α/Nrf2 signaling pathway. (3) OTA exacerbated endoplasmic reticulum stress in the muscle by causing endoplasmic reticulum expansion (results of transmission electron microscopy) and upregulating the expression of GRP78, eIF2α, ATF6, PERK, and CHOP. (4) OTA reduced muscle fiber diameter by inhibiting protein synthesis (AKT, TOR, and S6K1) and promoting the mRNA expression of protein degradation-related genes (MURF1, MAFBX, and FoxO3a), as well as by reducing AKT and promoting the immunofluorescence expression of FoxO3. (5) OTA inhibits collagen deposition by downregulating TGF-β1, TGF-βR1, Smad2, Smad3, Smad4, CTGF, TIMP, PHD, and LOX mRNA expressions as well as the CTGF immunofluorescence expression. Moreover, based on the GSH and collagen content contents, the upper safe dose for OTA-induced toxicity was 963.6 and 1129.6 μg/kg diet, respectively. Using the example of OTA, our research has provided new insights that raise concerns about the quality of aquatic products by exploring muscle toxicity caused by mycotoxins.
Collapse
Affiliation(s)
- Piao Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xin Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China
| | - Juan Yang
- Tongwei Co., Ltd., Chengdu, China, Healthy Aquaculture Key Laboratory of Sichuan Province, Sichuan 610041, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Key Laboratory of Sichuan Province, Ministry of Education, Ministry of Agriculture and Rural Affairs, Sichuan 611130, China.
| |
Collapse
|
15
|
Pratt J, Whitton L, Ryan A, Juliusdottir T, Dolan J, Conroy J, Narici M, De Vito G, Boreham C. Genes encoding agrin (AGRN) and neurotrypsin (PRSS12) are associated with muscle mass, strength and plasma C-terminal agrin fragment concentration. GeroScience 2023:10.1007/s11357-022-00721-1. [PMID: 36609795 PMCID: PMC10400504 DOI: 10.1007/s11357-022-00721-1] [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: 10/10/2022] [Accepted: 12/26/2022] [Indexed: 01/09/2023] Open
Abstract
Although physiological data suggest that neuromuscular junction (NMJ) dysfunction is a principal mechanism underpinning sarcopenia, genetic studies have implicated few genes involved in NMJ function. Accordingly, we explored whether genes encoding agrin (AGRN) and neurotrypsin (PRSS12) were associated with sarcopenia phenotypes: muscle mass, strength and plasma C-terminal agrin fragment (CAF). PhenoScanner was used to determine if AGRN and/or PRSS12 variants had previously been implicated with sarcopenia phenotypes. For replication, we combined genotype from whole genome sequencing with phenotypic data from 6715 GenoFit participants aged 18-83 years. Dual energy X-ray absorptiometry assessed whole body lean mass (WBLM) and appendicular lean mass (ALM), hand dynamometry determined grip strength and ELISA measured plasma CAF in a subgroup (n = 260). Follow-up analyses included eQTL analyses, carrier analyses, single-variant and gene-burden tests. rs2710873 (AGRN) and rs71608359 (PRSS12) associate with muscle mass and strength phenotypes, respectively, in the UKBB (p = 8.9 × 10-6 and p = 8.4 × 10-6) and GenoFit cohort (p = 0.019 and p = 0.014). rs2710873 and rs71608359 are eQTLs for AGRN and PRSS12, respectively, in ≥ three tissues. Compared to non-carriers, carriers of rs2710873 had 4.0% higher WBLM and ALM (both p < 0.001), and 9.5% lower CAF concentrations (p < 0.001), while carriers of rs71608359 had 2.3% lower grip strength (p = 0.034). AGRN and PRSS12 are associated with muscle strength and mass in single-variant analyses, while PRSS12 has further associations with muscle strength in gene-burden tests. Our findings provide novel evidence of the relevance of AGRN and PRSS12 to sarcopenia phenotypes and support existing physiological data illustrating the importance of the NMJ in maintaining muscle health during ageing.
Collapse
Affiliation(s)
- Jedd Pratt
- Institute for Sport and Health, University College Dublin, Dublin, Ireland. .,Genuity Science, Dublin, Ireland. .,Department of Biomedical Sciences, CIR-Myo Myology Centre, Neuromuscular Physiology Laboratory, University of Padova, Padua, Italy.
| | | | | | | | | | | | - Marco Narici
- Department of Biomedical Sciences, CIR-Myo Myology Centre, Neuromuscular Physiology Laboratory, University of Padova, Padua, Italy
| | - Giuseppe De Vito
- Department of Biomedical Sciences, CIR-Myo Myology Centre, Neuromuscular Physiology Laboratory, University of Padova, Padua, Italy
| | - Colin Boreham
- Institute for Sport and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Cai L, Shi L, Peng Z, Sun Y, Chen J. Ageing of skeletal muscle extracellular matrix and mitochondria: finding a potential link. Ann Med 2023; 55:2240707. [PMID: 37643318 PMCID: PMC10732198 DOI: 10.1080/07853890.2023.2240707] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/13/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Aim: To discuss the progress of extracellular matrix (ECM) characteristics, mitochondrial homeostasis, and their potential crosstalk in the pathogenesis of sarcopenia, a geriatric syndrome characterized by a generalized and progressive reduction in muscle mass, strength, and physical performance.Methods: This review focuses on the anatomy and physiology of skeletal muscle, alterations of ECM and mitochondria during ageing, and the role of the interplay between ECM and mitochondria in the pathogenesis of sarcopenia.Results: Emerging evidence points to a clear interplay between mitochondria and ECM in various tissues and organs. Under the ageing process, the ECM undergoes changes in composition and physical properties that may mediate mitochondrial changes via the systematic metabolism, ROS, SPARC pathway, and AMPK/PGC-1α signalling, which in turn exacerbate muscle degeneration. However, the precise effects of such crosstalk on the pathobiology of ageing, particularly in skeletal muscle, have not yet been fully understood.Conclusion: The changes in skeletal muscle ECM and mitochondria are partially responsible for the worsened muscle function during the ageing process. A deeper understanding of their alterations and interactions in sarcopenic patients can help prevent sarcopenia and improve its prognoses.
Collapse
Affiliation(s)
- Lubing Cai
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luze Shi
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Yang J, Vamvini M, Nigro P, Ho LL, Galani K, Alvarez M, Tanigawa Y, Renfro A, Carbone NP, Laakso M, Agudelo LZ, Pajukanta P, Hirshman MF, Middelbeek RJW, Grove K, Goodyear LJ, Kellis M. Single-cell dissection of the obesity-exercise axis in adipose-muscle tissues implies a critical role for mesenchymal stem cells. Cell Metab 2022; 34:1578-1593.e6. [PMID: 36198295 PMCID: PMC9558082 DOI: 10.1016/j.cmet.2022.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/29/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022]
Abstract
Exercise training is critical for the prevention and treatment of obesity, but its underlying mechanisms remain incompletely understood given the challenge of profiling heterogeneous effects across multiple tissues and cell types. Here, we address this challenge and opposing effects of exercise and high-fat diet (HFD)-induced obesity at single-cell resolution in subcutaneous and visceral white adipose tissue and skeletal muscle in mice with diet and exercise training interventions. We identify a prominent role of mesenchymal stem cells (MSCs) in obesity and exercise-induced tissue adaptation. Among the pathways regulated by exercise and HFD in MSCs across the three tissues, extracellular matrix remodeling and circadian rhythm are the most prominent. Inferred cell-cell interactions implicate within- and multi-tissue crosstalk centered around MSCs. Overall, our work reveals the intricacies and diversity of multi-tissue molecular responses to exercise and obesity and uncovers a previously underappreciated role of MSCs in tissue-specific and multi-tissue beneficial effects of exercise.
Collapse
Affiliation(s)
- Jiekun Yang
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Li-Lun Ho
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kyriakitsa Galani
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yosuke Tanigawa
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Renfro
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nicholas P Carbone
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Leandro Z Agudelo
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Grove
- Novo Nordisk Research Center, Seattle, WA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
18
|
Scripture-Adams DD, Chesmore KN, Barthélémy F, Wang RT, Nieves-Rodriguez S, Wang DW, Mokhonova EI, Douine ED, Wan J, Little I, Rabichow LN, Nelson SF, Miceli MC. Single nuclei transcriptomics of muscle reveals intra-muscular cell dynamics linked to dystrophin loss and rescue. Commun Biol 2022; 5:989. [PMID: 36123393 PMCID: PMC9485160 DOI: 10.1038/s42003-022-03938-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/01/2022] [Indexed: 02/05/2023] Open
Abstract
In Duchenne muscular dystrophy, dystrophin loss leads to chronic muscle damage, dysregulation of repair, fibro-fatty replacement, and weakness. We develop methodology to efficiently isolate individual nuclei from minute quantities of frozen skeletal muscle, allowing single nuclei sequencing of irreplaceable archival samples and from very small samples. We apply this method to identify cell and gene expression dynamics within human DMD and mdx mouse muscle, characterizing effects of dystrophin rescue by exon skipping therapy at single nuclei resolution. DMD exon 23 skipping events are directly observed and increased in myonuclei from treated mice. We describe partial rescue of type IIa and IIx myofibers, expansion of an MDSC-like myeloid population, recovery of repair/remodeling M2-macrophage, and repression of inflammatory POSTN1 + fibroblasts in response to exon skipping and partial dystrophin restoration. Use of this method enables exploration of cellular and transcriptomic mechanisms of dystrophin loss and repair within an intact muscle environment. Our initial findings will scaffold our future work to more directly examine muscular dystrophies and putative recovery pathways.
Collapse
Affiliation(s)
- Deirdre D Scripture-Adams
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kevin N Chesmore
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Florian Barthélémy
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard T Wang
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shirley Nieves-Rodriguez
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Derek W Wang
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Amgen, Thousand Oaks, CA, USA
| | - Ekaterina I Mokhonova
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emilie D Douine
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jijun Wan
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Isaiah Little
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laura N Rabichow
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - M Carrie Miceli
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Giha HA, Sater MS, Alamin OAO. Diabetes mellitus tendino-myopathy: epidemiology, clinical features, diagnosis and management of an overlooked diabetic complication. Acta Diabetol 2022; 59:871-883. [PMID: 35291027 DOI: 10.1007/s00592-022-01860-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/22/2022] [Indexed: 12/13/2022]
Abstract
Tendino-myopathy, an unexplored niche, is a non-vascular unstated T2DM complication, which is largely disregarded in clinical practice, thus, we aim to explore it in this review. Literature search using published data from different online resources. Epidemiologically, reported prevalence varies around 10-90%, which is marked variable and unreliable. Clinically, diabetic tendino-myopathy is typified by restriction of movement, pain/tenderness, cramps and decreased functions. Moreover, myopathy is characterized by muscle atrophy, weakness and ischemia, and tendinopathy by deformities and reduced functions/precision. In tendonapthy, the three most affected regions are: the hand (cheiroarthropathy, Dupuytren's contracture, flexor tenosynovitis and carpel tunnel syndrome), shoulder (adhesive capsulitis, rotator cuff tendinopathy and tenosynovitis) and foot (Achilles tendinopathy with the risk of tear/rupture), in addition to diffuse idiopathic skeletal hyperostosis. Pathologically, it is characterized by decreased muscle fiber mass and increased fibrosis, with marked extracellular matrix remodeling and deposition of collagens. The tendon changes include decreased collagen fibril diameter, changed morphology, increased packing and disorganization, with overall thickening, and calcification. Diagnosis is basically clinical and radiological, while diagnostic biomarkers are awaited. Management is done by diabetes control, special nutrition and physiotherapy, while analgesics, steroids and surgery are used in tendinopathy. Several antisarcopenic drugs are in the pipeline. This review aims to bridge clinical practice with research and update routine diabetic checkup by inclusion of tendino-myopathies in the list with an emphasis on management.
Collapse
Affiliation(s)
- Hayder A Giha
- Medical Biochemistry and Molecular Biology, Khartoum, Sudan.
| | - Mai S Sater
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Kingdom of Bahrain
| | - Osman A O Alamin
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Alneelain University, Khartoum, Sudan
- Interventional Cardiology, Ahmad Gasim Cardiac Centre, Ahmad Gasim Hospital, Khartoum North, Sudan
| |
Collapse
|
20
|
Identification of distinct non-myogenic skeletal-muscle-resident mesenchymal cell populations. Cell Rep 2022; 39:110785. [PMID: 35545045 PMCID: PMC9535675 DOI: 10.1016/j.celrep.2022.110785] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/23/2022] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal progenitors of the lateral plate mesoderm give rise to various cell fates within limbs, including a heterogeneous group of muscle-resident mesenchymal cells. Often described as fibro-adipogenic progenitors, these cells are key players in muscle development, disease, and regeneration. To further define this cell population(s), we perform lineage/reporter analysis, flow cytometry, single-cell RNA sequencing, immunofluorescent staining, and differentiation assays on normal and injured murine muscles. Here we identify six distinct Pdgfra+ non-myogenic muscle-resident mesenchymal cell populations that fit within a bipartite differentiation trajectory from a common progenitor. One branch of the trajectory gives rise to two populations of immune-responsive mesenchymal cells with strong adipogenic potential and the capability to respond to acute and chronic muscle injury, whereas the alternative branch contains two cell populations with limited adipogenic capacity and inherent mineralizing capabilities; one of the populations displays a unique neuromuscular junction association and an ability to respond to nerve injury. Leinroth et al. explore the heterogeneity of Pdgfra+ muscle-resident mesenchymal cells, demonstrating that Pdgfra+ subpopulations have unique gene expression profiles, exhibit two distinct cell trajectories from a common progenitor, differ in their abilities to respond to muscle injuries, and show variable adipogenic and mineralizing capacities.
Collapse
|
21
|
Zhang X, Zhang C, Yang C, Kuang L, Zheng J, Tang L, Lei M, Li C, Ren Y, Guo Z, Ji Y, Deng X, Huang D, Wang G, Xie X. Circular RNA, microRNA and Protein Profiles of the Longissimus Dorsi of Germany ZIKA and Sichuan White Rabbits. Front Genet 2022; 12:777232. [PMID: 35003217 PMCID: PMC8740122 DOI: 10.3389/fgene.2021.777232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Due to the dietetic properties and remarkable nutritive value of rabbit meat, its industry is increasing rapidly. However, the association between circular RNAs, microRNAs, and proteins and muscle fiber type, and meat quality of rabbit is still unknown. Here, using deep sequencing and iTRAQ proteomics technologies we first identified 3159 circRNAs, 356 miRNAs, and 755 proteins in the longissimus dorsi tissues from Sichuan white (SCWrabs) and Germany great line ZIKA rabbits (ZIKArabs). Next, we identified 267 circRNAs, 3 miRNAs, and 29 proteins differentially expressed in the muscle tissues of SCWrabs and ZIKArabs. Interaction network analysis revealed some key regulation relationships between noncoding RNAs and proteins that might be associated with the muscle fiber type and meat quality of rabbit. Further, miRNA isoforms and gene variants identified in SCWrabs and ZIKArabs revealed some pathways and biological processes related to the muscle development. This is the first study of noncoding RNA and protein profiles for the two rabbit breeds. It provides a valuable resource for future studies in rabbits and will improve our understanding of the molecular regulation mechanisms in the muscle development of livestock. More importantly, the output of our study will benefit the researchers and producers in the rabbit breeding program.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Cuixia Zhang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Chao Yang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Liangde Kuang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Jie Zheng
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Li Tang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Min Lei
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Congyan Li
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Yongjun Ren
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhiqiang Guo
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Yang Ji
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | | | - Dengping Huang
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Gaofu Wang
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Xiaohong Xie
- Sichuan Animal Sciences Academy, Chengdu, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
22
|
Molina T, Fabre P, Dumont NA. Fibro-adipogenic progenitors in skeletal muscle homeostasis, regeneration and diseases. Open Biol 2021; 11:210110. [PMID: 34875199 PMCID: PMC8651418 DOI: 10.1098/rsob.210110] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle possesses a remarkable regenerative capacity that relies on the activity of muscle stem cells, also known as satellite cells. The presence of non-myogenic cells also plays a key role in the coordination of skeletal muscle regeneration. Particularly, fibro-adipogenic progenitors (FAPs) emerged as master regulators of muscle stem cell function and skeletal muscle regeneration. This population of muscle resident mesenchymal stromal cells has been initially characterized based on its bi-potent ability to differentiate into fibroblasts or adipocytes. New technologies such as single-cell RNAseq revealed the cellular heterogeneity of FAPs and their complex regulatory network during muscle regeneration. In acute injury, FAPs rapidly enter the cell cycle and secrete trophic factors that support the myogenic activity of muscle stem cells. Conversely, deregulation of FAP cell activity is associated with the accumulation of fibrofatty tissue in pathological conditions such as muscular dystrophies and ageing. Considering their central role in skeletal muscle pathophysiology, the regulatory mechanisms of FAPs and their cellular and molecular crosstalk with muscle stem cells are highly investigated in the field. In this review, we summarize the current knowledge on FAP cell characteristics, heterogeneity and the cellular crosstalk during skeletal muscle homeostasis and regeneration. We further describe their role in muscular disorders, as well as different therapeutic strategies targeting these cells to restore muscle regeneration.
Collapse
Affiliation(s)
- Thomas Molina
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Paul Fabre
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas A. Dumont
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Wei X, Nicoletti C, Puri PL. Fibro-Adipogenic Progenitors: Versatile keepers of skeletal muscle homeostasis, beyond the response to myotrauma. Semin Cell Dev Biol 2021; 119:23-31. [PMID: 34332886 PMCID: PMC8552908 DOI: 10.1016/j.semcdb.2021.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
While Fibro-Adipogenic Progenitors (FAPs) have been originally identified as muscle-interstitial mesenchymal cells activated in response to muscle injury and endowed with inducible fibrogenic and adipogenic potential, subsequent studies have expanded their phenotypic and functional repertoire and revealed their contribution to skeletal muscle response to a vast range of perturbations. Here we review the emerging contribution of FAPs to skeletal muscle responses to motor neuron injuries and to systemic physiological (e.g., exercise) or pathological metabolic (e.g., diabetes) perturbations. We also provide an initial blueprint of discrete sub-clusters of FAPs that are activated by specific perturbations and discuss their role in muscle adaptation to these conditions.
Collapse
Affiliation(s)
- X Wei
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - C Nicoletti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - P L Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
24
|
Lieber RL, Binder-Markey B. Biochemical and structural basis of the passive mechanical properties of whole skeletal muscle. J Physiol 2021; 599:3809-3823. [PMID: 34101193 PMCID: PMC8364503 DOI: 10.1113/jp280867] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/06/2021] [Indexed: 01/18/2023] Open
Abstract
Passive mechanical properties of whole skeletal muscle are not as well understood as active mechanical properties. Both the structural basis for passive mechanical properties and the properties themselves are challenging to determine because it is not clear which structures within skeletal muscle actually bear passive loads and there are not established standards by which to make mechanical measurements. Evidence suggests that titin bears the majority of the passive load within the single muscle cell. However, at larger scales, such as fascicles and muscles, there is emerging evidence that the extracellular matrix bears the major part of the load. Complicating the ability to quantify and compare across size scales, muscles and species, definitions of muscle passive properties such as stress, strain, modulus and stiffness can be made relative to many reference parameters. These uncertainties make a full understanding of whole muscle passive mechanical properties and modelling these properties very difficult. Future studies defining the specific load bearing structures and their composition and organization are required to fully understand passive mechanics of the whole muscle and develop therapies to treat disorders in which passive muscle properties are altered such as muscular dystrophy, traumatic laceration, and contracture due to upper motor neuron lesion as seen in spinal cord injury, stroke and cerebral palsy.
Collapse
Affiliation(s)
- Richard L. Lieber
- Shirley Ryan AbilityLab
- Departments of Physical Medicine and Rehabilitation and
Biomedical Engineering, Northwestern University, Chicago, IL, USA
- Edward Hines V.A. Medical Center, Hines, IL USA
| | - Ben Binder-Markey
- Department of Physical Therapy and Rehabilitation Sciences
and School of Biomedical Engineering, Sciences and Health Systems, Drexel
University, Philadelphia, PA USA
| |
Collapse
|
25
|
Contreras O, Rossi FMV, Theret M. Origins, potency, and heterogeneity of skeletal muscle fibro-adipogenic progenitors-time for new definitions. Skelet Muscle 2021; 11:16. [PMID: 34210364 PMCID: PMC8247239 DOI: 10.1186/s13395-021-00265-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Striated muscle is a highly plastic and regenerative organ that regulates body movement, temperature, and metabolism-all the functions needed for an individual's health and well-being. The muscle connective tissue's main components are the extracellular matrix and its resident stromal cells, which continuously reshape it in embryonic development, homeostasis, and regeneration. Fibro-adipogenic progenitors are enigmatic and transformative muscle-resident interstitial cells with mesenchymal stem/stromal cell properties. They act as cellular sentinels and physiological hubs for adult muscle homeostasis and regeneration by shaping the microenvironment by secreting a complex cocktail of extracellular matrix components, diffusible cytokines, ligands, and immune-modulatory factors. Fibro-adipogenic progenitors are the lineage precursors of specialized cells, including activated fibroblasts, adipocytes, and osteogenic cells after injury. Here, we discuss current research gaps, potential druggable developments, and outstanding questions about fibro-adipogenic progenitor origins, potency, and heterogeneity. Finally, we took advantage of recent advances in single-cell technologies combined with lineage tracing to unify the diversity of stromal fibro-adipogenic progenitors. Thus, this compelling review provides new cellular and molecular insights in comprehending the origins, definitions, markers, fate, and plasticity of murine and human fibro-adipogenic progenitors in muscle development, homeostasis, regeneration, and repair.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, 2052, Australia.
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fabio M V Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
26
|
Leiva-Cepas F, Benito-Ysamat A, Jimena I, Jimenez-Diaz F, Gil-Belmonte MJ, Ruz-Caracuel I, Villalba R, Peña-Amaro J. Ultrasonographic and Histological Correlation after Experimental Reconstruction of a Volumetric Muscle Loss Injury with Adipose Tissue. Int J Mol Sci 2021; 22:ijms22136689. [PMID: 34206557 PMCID: PMC8268690 DOI: 10.3390/ijms22136689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 02/05/2023] Open
Abstract
Different types of scaffolds are used to reconstruct muscle volume loss injuries. In this experimental study, we correlated ultrasound observations with histological findings in a muscle volume loss injury reconstructed with autologous adipose tissue. The outcome is compared with decellularized and porous matrix implants. Autologous adipose tissue, decellularized matrix, and a porous collagen matrix were implanted in volumetric muscle loss (VML) injuries generated on the anterior tibial muscles of Wistar rats. Sixty days after implantation, ultrasound findings were compared with histological and histomorphometric analysis. The muscles with an autologous adipose tissue implant exhibited an ultrasound pattern that was quite similar to that of the regenerative control muscles. From a histological point of view, the defects had been occupied by newly formed muscle tissue with certain structural abnormalities that would explain the differences between the ultrasound patterns of the normal control muscles and the regenerated ones. While the decellularized muscle matrix implant resulted in fibrosis and an inflammatory response, the porous collagen matrix implant was replaced by regenerative muscle fibers with neurogenic atrophy and fibrosis. In both cases, the ultrasound images reflected echogenic, echotextural, and vascular changes compatible with the histological findings of failed muscle regeneration. The ultrasound analysis confirmed the histological findings observed in the VML injuries reconstructed by autologous adipose tissue implantation. Ultrasound can be a useful tool for evaluating the structure of muscles reconstructed through tissue engineering.
Collapse
Affiliation(s)
- Fernando Leiva-Cepas
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Department of Pathology, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Alberto Benito-Ysamat
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
- Radiology Department, Musculoskeletal Section, Reina Sofia University Hospital, 14004 Cordoba, Spain
| | - Ignacio Jimena
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Fernando Jimenez-Diaz
- Sport Sciences Faculty, Castilla La Mancha University, 45071 Toledo, Spain;
- Department of Health Sciences, Faculty of Medicine, Campus de los Jerónimos, San Antonio Catholic University (UCAM), 30107 Murcia, Spain
| | - Maria Jesus Gil-Belmonte
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
| | - Ignacio Ruz-Caracuel
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Department of Pathology, Ramon y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Rafael Villalba
- Tissue of Establishment of the Center for Transfusion, Tissues and Cells, 14004 Cordoba, Spain;
| | - Jose Peña-Amaro
- Research Group in Muscle Regeneration, Department of Morphological Sciences, Faculty of Medicine and Nursing, University of Cordoba, 14004 Cordoba, Spain; (F.L.-C.); (A.B.-Y.); (I.J.); (M.J.G.-B.); (I.R.-C.)
- Maimonides Institute for Biomedical Research IMIBIC, Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
- Correspondence:
| |
Collapse
|
27
|
Sacher F, Feregrino C, Tschopp P, Ewald CY. Extracellular matrix gene expression signatures as cell type and cell state identifiers. Matrix Biol Plus 2021; 10:100069. [PMID: 34195598 PMCID: PMC8233473 DOI: 10.1016/j.mbplus.2021.100069] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Transcriptomic signatures based on cellular mRNA expression profiles can be used to categorize cell types and states. Yet whether different functional groups of genes perform better or worse in this process remains largely unexplored. Here we test the core matrisome - that is, all genes coding for structural proteins of the extracellular matrix - for its ability to delineate distinct cell types in embryonic single-cell RNA-sequencing (scRNA-seq) data. We show that even though expressed core matrisome genes correspond to less than 2% of an entire cellular transcriptome, their RNA expression levels suffice to recapitulate essential aspects of cell type-specific clustering. Notably, using scRNA-seq data from the embryonic limb, we demonstrate that core matrisome gene expression outperforms random gene subsets of similar sizes and can match and exceed the predictive power of transcription factors. While transcription factor signatures generally perform better in predicting cell types at early stages of chicken and mouse limb development, i.e., when cells are less differentiated, the information content of the core matrisome signature increases in more differentiated cells. Moreover, using cross-species analyses, we show that these cell type-specific signatures are evolutionarily conserved. Our findings suggest that each cell type produces its own unique extracellular matrix, or matreotype, which becomes progressively more refined and cell type-specific as embryonic tissues mature.
Collapse
Affiliation(s)
- Fabio Sacher
- Laboratory of Regulatory Evolution, DUW Zoology, University of Basel, Basel CH-4051, Switzerland
| | - Christian Feregrino
- Laboratory of Regulatory Evolution, DUW Zoology, University of Basel, Basel CH-4051, Switzerland
| | - Patrick Tschopp
- Laboratory of Regulatory Evolution, DUW Zoology, University of Basel, Basel CH-4051, Switzerland
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| |
Collapse
|
28
|
Murach KA, Peck BD, Policastro RA, Vechetti IJ, Van Pelt DW, Dungan CM, Denes LT, Fu X, Brightwell CR, Zentner GE, Dupont-Versteegden EE, Richards CI, Smith JJ, Fry CS, McCarthy JJ, Peterson CA. Early satellite cell communication creates a permissive environment for long-term muscle growth. iScience 2021; 24:102372. [PMID: 33948557 PMCID: PMC8080523 DOI: 10.1016/j.isci.2021.102372] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 12/22/2022] Open
Abstract
Using in vivo muscle stem cell (satellite cell)-specific extracellular vesicle (EV) tracking, satellite cell depletion, in vitro cell culture, and single-cell RNA sequencing, we show satellite cells communicate with other cells in skeletal muscle during mechanical overload. Early satellite cell EV communication primes the muscle milieu for proper long-term extracellular matrix (ECM) deposition and is sufficient to support sustained hypertrophy in adult mice, even in the absence of fusion to muscle fibers. Satellite cells modulate chemokine gene expression across cell types within the first few days of loading, and EV delivery of miR-206 to fibrogenic cells represses Wisp1 expression required for appropriate ECM remodeling. Late-stage communication from myogenic cells during loading is widespread but may be targeted toward endothelial cells. Satellite cells coordinate adaptation by influencing the phenotype of recipient cells, which extends our understanding of their role in muscle adaptation beyond regeneration and myonuclear donation.
Collapse
Affiliation(s)
- Kevin A. Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Bailey D. Peck
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Robert A. Policastro
- Department of Biology, College of Arts and Sciences, University of Indiana, Bloomington, IN 47405, USA
| | - Ivan J. Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska, Lincoln, NE 68588, USA
| | - Douglas W. Van Pelt
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Cory M. Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Lance T. Denes
- Department of Molecular Genetics and Microbiology, Center for Neurogenetics, University of Florida, Gainesville, FL 32611, USA
| | - Xu Fu
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Camille R. Brightwell
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Gabriel E. Zentner
- Department of Biology, College of Arts and Sciences, University of Indiana, Bloomington, IN 47405, USA
| | - Esther E. Dupont-Versteegden
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher I. Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jeramiah J. Smith
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506, USA
| | - Christopher S. Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J. McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Charlotte A. Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
29
|
Theret M, Rossi FMV, Contreras O. Evolving Roles of Muscle-Resident Fibro-Adipogenic Progenitors in Health, Regeneration, Neuromuscular Disorders, and Aging. Front Physiol 2021; 12:673404. [PMID: 33959042 PMCID: PMC8093402 DOI: 10.3389/fphys.2021.673404] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Normal skeletal muscle functions are affected following trauma, chronic diseases, inherited neuromuscular disorders, aging, and cachexia, hampering the daily activities and quality of life of the affected patients. The maladaptive accumulation of fibrous intramuscular connective tissue and fat are hallmarks of multiple pathologies where chronic damage and inflammation are not resolved, leading to progressive muscle replacement and tissue degeneration. Muscle-resident fibro-adipogenic progenitors are adaptable stromal cells with multilineage potential. They are required for muscle homeostasis, neuromuscular integrity, and tissue regeneration. Fibro-adipogenic progenitors actively regulate and shape the extracellular matrix and exert immunomodulatory functions via cross-talk with multiple other residents and non-resident muscle cells. Remarkably, cumulative evidence shows that a significant proportion of activated fibroblasts, adipocytes, and bone-cartilage cells, found after muscle trauma and disease, descend from these enigmatic interstitial progenitors. Despite the profound impact of muscle disease on human health, the fibrous, fatty, and ectopic bone tissues' origins are poorly understood. Here, we review the current knowledge of fibro-adipogenic progenitor function on muscle homeostatic integrity, regeneration, repair, and aging. We also discuss how scar-forming pathologies and disorders lead to dysregulations in their behavior and plasticity and how these stromal cells can control the onset and severity of muscle loss in disease. We finally explore the rationale of improving muscle regeneration by understanding and modulating fibro-adipogenic progenitors' fate and behavior.
Collapse
Affiliation(s)
- Marine Theret
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Osvaldo Contreras
- Departamento de Biología Celular y Molecular, Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, Australia
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| |
Collapse
|
30
|
Extracellular matrix: an important regulator of cell functions and skeletal muscle development. Cell Biosci 2021; 11:65. [PMID: 33789727 PMCID: PMC8011170 DOI: 10.1186/s13578-021-00579-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular matrix (ECM) is a kind of connective tissue in the cell microenvironment, which is of great significance to tissue development. ECM in muscle fiber niche consists of three layers: the epimysium, the perimysium, and the endomysium (basal lamina). These three layers of connective tissue structure can not only maintain the morphology of skeletal muscle, but also play an important role in the physiological functions of muscle cells, such as the transmission of mechanical force, the regeneration of muscle fiber, and the formation of neuromuscular junction. In this paper, detailed discussions are made for the structure and key components of ECM in skeletal muscle tissue, the role of ECM in skeletal muscle development, and the application of ECM in biomedical engineering. This review will provide the reader with a comprehensive overview of ECM, as well as a comprehensive understanding of the structure, physiological function, and application of ECM in skeletal muscle tissue.
Collapse
|
31
|
Carraro U, Yablonka-Reuveni Z. Translational research on Myology and Mobility Medicine: 2021 semi-virtual PDM3 from Thermae of Euganean Hills, May 26 - 29, 2021. Eur J Transl Myol 2021; 31:9743. [PMID: 33733717 PMCID: PMC8056169 DOI: 10.4081/ejtm.2021.9743] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
On 19-21 November 2020, the meeting of the 30 years of the Padova Muscle Days was virtually held while the SARS-CoV-2 epidemic was hitting the world after a seemingly quiet summer. During the 2020-2021 winter, the epidemic is still active, despite the start of vaccinations. The organizers hope to hold the 2021 Padua Days on Myology and Mobility Medicine in a semi-virtual form (2021 S-V PDM3) from May 26 to May 29 at the Thermae of Euganean Hills, Padova, Italy. Here the program and the Collection of Abstracts are presented. Despite numerous world problems, the number of submitted/selected presentations (lectures and oral presentations) has increased, prompting the organizers to extend the program to four dense days.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences of the University of Padova, Italy; CIR-Myo - Myology Centre, University of Padova, Italy; A-C Mioni-Carraro Foundation for Translational Myology, Padova.
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
32
|
Synergistic short-term and long-term effects of TGF-β1 and 3 on collagen production in differentiating myoblasts. Biochem Biophys Res Commun 2021; 547:176-182. [PMID: 33618224 DOI: 10.1016/j.bbrc.2021.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Skeletal muscle fibrosis and regeneration are modulated by transforming growth factor β (TGF-β) superfamily. Amongst them, TGF-β1 is a highly potent pro-fibrotic factor, while TGF-β3 has been implicated to reduce scar formation and collagen production in skin and vocal mucosa. However, little is known about the individual and combined short- and long-term effects of TGF-β1 and TGF-β3 on collagen expression in myoblasts and myotubes. Here we show that in C2C12 myoblasts TGF-β1 and/or TGF-β3 increased mRNA expression of Ctgf and Fgf-2 persistently after 3 h and of Col1A1 after 24 h, while TGF-β1+TGF-β3 mitigated these effects after 48 h incubation. Gene expression of Tgf-β1 was enhanced by TGF-β1 and/or TGF-β3 after 24 h and 48 h. However, Tgfbr1 mRNA expression was reduced at 48 h. After 48 h incubation with TGF-β1 and/or TGF-β3, Col3A1 and Col4A1 mRNA expression levels were decreased. Myoblasts produced collagen after three days incubation with TGF-β1 and/or TGF-β3 in a dose independent manner. Collagen deposition was doubled when myoblasts differentiated into myotubes and TGF-β1 and/or TGF-β3 did not stimulate collagen production any further. TGF-β type I receptor (TGFBR1) inhibitor, LY364947, suppressed TGF-βs-induced collagen production. Collagen I expression was higher in myotubes than in myoblasts. TGF-β1 and/or TGF-β3 inhibited myotube differentiation which was antagonized by LY364947. These results indicate that both C2C12 myoblasts and myotubes produce collagen. Whereas TGF-β1 and TGF-β3 individually and simultaneously stimulate collagen production in C2C12 differentiating myoblasts, in myotubes these effects are less prominent. In muscle cells, TGF-β3 is ineffective to antagonize TGF-β1-induced collagen production.
Collapse
|
33
|
Mukund K, Subramaniam S. Skeletal muscle: A review of molecular structure and function, in health and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1462. [PMID: 31407867 PMCID: PMC6916202 DOI: 10.1002/wsbm.1462] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
Decades of research in skeletal muscle physiology have provided multiscale insights into the structural and functional complexity of this important anatomical tissue, designed to accomplish the task of generating contraction, force and movement. Skeletal muscle can be viewed as a biomechanical device with various interacting components including the autonomic nerves for impulse transmission, vasculature for efficient oxygenation, and embedded regulatory and metabolic machinery for maintaining cellular homeostasis. The "omics" revolution has propelled a new era in muscle research, allowing us to discern minute details of molecular cross-talk required for effective coordination between the myriad interacting components for efficient muscle function. The objective of this review is to provide a systems-level, comprehensive mapping the molecular mechanisms underlying skeletal muscle structure and function, in health and disease. We begin this review with a focus on molecular mechanisms underlying muscle tissue development (myogenesis), with an emphasis on satellite cells and muscle regeneration. We next review the molecular structure and mechanisms underlying the many structural components of the muscle: neuromuscular junction, sarcomere, cytoskeleton, extracellular matrix, and vasculature surrounding muscle. We highlight aberrant molecular mechanisms and their possible clinical or pathophysiological relevance. We particularly emphasize the impact of environmental stressors (inflammation and oxidative stress) in contributing to muscle pathophysiology including atrophy, hypertrophy, and fibrosis. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of BioengineeringUniversity of CaliforniaSan DiegoCalifornia
| | - Shankar Subramaniam
- Department of Bioengineering, Bioinformatics & Systems BiologyUniversity of CaliforniaSan DiegoCalifornia
- Department of Computer Science and EngineeringUniversity of CaliforniaSan DiegoCalifornia
- Department of Cellular and Molecular Medicine and NanoengineeringUniversity of CaliforniaSan DiegoCalifornia
| |
Collapse
|
34
|
Role of hypoxia in skeletal muscle fibrosis: Synergism between hypoxia and TGF-β signaling upregulates CCN2/CTGF expression specifically in muscle fibers. Matrix Biol 2019; 87:48-65. [PMID: 31669521 DOI: 10.1016/j.matbio.2019.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023]
Abstract
Several skeletal muscle diseases are characterized by fibrosis, the excessive accumulation of extracellular matrix. Transforming growth factor-β (TGF-β) and connective tissue growth factor (CCN2/CTGF) are two profibrotic factors augmented in fibrotic skeletal muscle, together with signs of reduced vasculature that implies a decrease in oxygen supply. We observed that fibrotic muscles are characterized by the presence of positive nuclei for hypoxia-inducible factor-1α (HIF-1α), a key mediator of the hypoxia response. However, it is not clear how a hypoxic environment could contribute to the fibrotic phenotype in skeletal muscle. We evaluated the role of hypoxia and TGF-β on CCN2 expression in vitro. Fibroblasts, myoblasts and differentiated myotubes were incubated with TGF-β1 under hypoxic conditions. Hypoxia and TGF-β1 induced CCN2 expression synergistically in myotubes but not in fibroblasts or undifferentiated muscle progenitors. This induction requires HIF-1α and the Smad-independent TGF-β signaling pathway. We performed in vivo experiments using pharmacological stabilization of HIF-1α or hypoxia-induced via hindlimb ischemia together with intramuscular injections of TGF-β1, and we found increased CCN2 expression. These observations suggest that hypoxic signaling together with TGF-β signaling, which are both characteristics of a fibrotic skeletal muscle environment, induce the expression of CCN2 in skeletal muscle fibers and myotubes.
Collapse
|
35
|
Lieber RL, Fridén J. Muscle contracture and passive mechanics in cerebral palsy. J Appl Physiol (1985) 2019; 126:1492-1501. [PMID: 30571285 PMCID: PMC6589815 DOI: 10.1152/japplphysiol.00278.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 11/06/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle contractures represent the permanent shortening of a muscle-tendon unit, resulting in loss of elasticity and, in extreme cases, joint deformation. They may result from cerebral palsy, spinal cord injury, stroke, muscular dystrophy, and other neuromuscular disorders. Contractures are the prototypic and most severe clinical presentation of increased passive mechanical muscle force in humans, often requiring surgical correction. Intraoperative experiments demonstrate that high muscle passive force is associated with sarcomeres that are abnormally stretched, although otherwise normal, with fewer sarcomeres in series. Furthermore, changes in the amount and arrangement of collagen in the extracellular matrix also increase muscle stiffness. Structural light and electron microscopy studies demonstrate that large bundles of collagen, referred to as perimysial cables, may be responsible for this increased stiffness and are regulated by interaction of a number of cell types within the extracellular matrix. Loss of muscle satellite cells may be related to changes in both sarcomeres and extracellular matrix. Future studies are required to determine the underlying mechanism for changes in muscle satellite cells and their relationship (if any) to contracture. A more complete understanding of this mechanism may lead to effective nonsurgical treatments to relieve and even prevent muscle contractures.
Collapse
Affiliation(s)
- Richard L Lieber
- Shirley Ryan AbilityLab, Chicago, Illinois
- Departments of Physical Medicine and Rehabilitation and Biomedical Engineering, Northwestern University , Chicago, Illinois
| | - Jan Fridén
- Swiss Paraplegic Center, Nottwil, Switzerland
| |
Collapse
|
36
|
Bechshøft CJL, Jensen SM, Schjerling P, Andersen JL, Svensson RB, Eriksen CS, Mkumbuzi NS, Kjaer M, Mackey AL. Age and prior exercise in vivo determine the subsequent in vitro molecular profile of myoblasts and nonmyogenic cells derived from human skeletal muscle. Am J Physiol Cell Physiol 2019; 316:C898-C912. [PMID: 30917034 DOI: 10.1152/ajpcell.00049.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The decline in skeletal muscle regenerative capacity with age is partly attributed to muscle stem cell (satellite cell) dysfunction. Recent evidence has pointed to a strong interaction between myoblasts and fibroblasts, but the influence of age on this interaction is unknown. Additionally, while the native tissue environment is known to determine the properties of myogenic cells in vitro, how the aging process alters this cell memory has not been established at the molecular level. We recruited 12 young and 12 elderly women, who performed a single bout of heavy resistance exercise with the knee extensor muscles of one leg. Five days later, muscle biopsies were collected from both legs, and myogenic cells and nonmyogenic cells were isolated for in vitro experiments with mixed or separated cells and analyzed by immunostaining and RT-PCR. A lower myogenic fusion index was detected in the cells from the old versus young women, in association with differences in gene expression levels of key myogenic regulatory factors and senescence, which were further altered by performing exercise before tissue sampling. Coculture with nonmyogenic cells from the elderly led to a higher myogenic differentiation index compared with nonmyogenic cells from the young. These findings show that the in vitro phenotype and molecular profile of human skeletal muscle myoblasts and fibroblasts is determined by the age and exercise state of the original in vivo environment and help explain how exercise can enhance muscle stem cell function in old age.
Collapse
Affiliation(s)
- Cecilie J L Bechshøft
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Simon M Jensen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jesper L Andersen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Christian S Eriksen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Nonhlanhla S Mkumbuzi
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Newlands, South Africa
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
37
|
Verma M, Asakura Y, Murakonda BSR, Pengo T, Latroche C, Chazaud B, McLoon LK, Asakura A. Muscle Satellite Cell Cross-Talk with a Vascular Niche Maintains Quiescence via VEGF and Notch Signaling. Cell Stem Cell 2018; 23:530-543.e9. [PMID: 30290177 PMCID: PMC6178221 DOI: 10.1016/j.stem.2018.09.007] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/19/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022]
Abstract
Skeletal muscle is a complex tissue containing tissue resident muscle stem cells (satellite cells) (MuSCs) important for postnatal muscle growth and regeneration. Quantitative analysis of the biological function of MuSCs and the molecular pathways responsible for a potential juxtavascular niche for MuSCs is currently lacking. We utilized fluorescent reporter mice and muscle tissue clearing to investigate the proximity of MuSCs to capillaries in 3 dimensions. We show that MuSCs express abundant VEGFA, which recruits endothelial cells (ECs) in vitro, whereas blocking VEGFA using both a vascular endothelial growth factor (VEGF) inhibitor and MuSC-specific VEGFA gene deletion reduces the proximity of MuSCs to capillaries. Importantly, this proximity to the blood vessels was associated with MuSC self-renewal in which the EC-derived Notch ligand Dll4 induces quiescence in MuSCs. We hypothesize that MuSCs recruit capillary ECs via VEGFA, and in return, ECs maintain MuSC quiescence though Dll4.
Collapse
Affiliation(s)
- Mayank Verma
- Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Bhavani Sai Rohit Murakonda
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Claire Latroche
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | | | - Linda K McLoon
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Ophthalmology and Visual Neurosciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
38
|
Visualization of specific collagen-producing cells by Col1-GFP transgenic mice revealed novel type I collagen-producing cells other than fibroblasts in systemic organs/tissues. Biochem Biophys Res Commun 2018; 505:267-273. [PMID: 30245134 DOI: 10.1016/j.bbrc.2018.09.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 01/18/2023]
Abstract
Type I collagen is one of the most abundant proteins in mammals and plays important roles in maintaining the integrity of many tissues. Although fibroblasts are the main source of type I collagen, other cells also produce it; however, these cells are not well-defined owing to the lack of a specific marker. A transgenic (Tg) mouse line has been generated in which type I collagen-producing cells are labeled with enhanced green fluorescent protein (EGFP), which enables the monitoring of these cells without requiring an additional cell marker. This Tg mouse line has since been widely used to study type I collagen-producing cells and fibrosis; one study revealed that podocytes, which were not previously considered to produce type I collagen, expressed EGFP. This raises a question regarding the specificity of EGFP expression in this Tg mouse line. To exclude the possibility of non-specific EGFP expression in the existing Tg mouse line and specifically monitor type I collagen-producing cells, we generated a new Tg mouse line and histologically confirmed the specificity of EGFP expression throughout the body. Moreover, we explored type I collagen-producing cells other than fibroblasts and revealed for the first time that Leydig cells have the ability to produce type I collagen. Because of its highly specific and physiologically accurate expression, our new Tg mouse line will help to accurately elucidate not only type I collagen-producing cells in normal tissues but also the potential cells in fibrotic tissues, providing new insights into the pathology of fibrosis.
Collapse
|
39
|
Xu P, Werner JU, Milerski S, Hamp CM, Kuzenko T, Jähnert M, Gottmann P, de Roy L, Warnecke D, Abaei A, Palmer A, Huber-Lang M, Dürselen L, Rasche V, Schürmann A, Wabitsch M, Knippschild U. Diet-Induced Obesity Affects Muscle Regeneration After Murine Blunt Muscle Trauma-A Broad Spectrum Analysis. Front Physiol 2018; 9:674. [PMID: 29922174 PMCID: PMC5996306 DOI: 10.3389/fphys.2018.00674] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Injury to skeletal muscle affects millions of people worldwide. The underlying regenerative process however, is a very complex mechanism, time-wise highly coordinated, and subdivided in an initial inflammatory, a regenerative and a remodeling phase. Muscle regeneration can be impaired by several factors, among them diet-induced obesity (DIO). In order to evaluate if obesity negatively affects healing processes after trauma, we utilized a blunt injury approach to damage the extensor iliotibialis anticus muscle on the left hind limb of obese and normal weight C57BL/6J without showing any significant differences in force input between normal weight and obese mice. Magnetic resonance imaging (MRI) of the injury and regeneration process revealed edema formation and hemorrhage exudate in muscle tissue of normal weight and obese mice. In addition, morphological analysis of physiological changes revealed tissue necrosis, immune cell infiltration, extracellular matrix (ECM) remodeling, and fibrosis formation in the damaged muscle tissue. Regeneration was delayed in muscles of obese mice, with a higher incidence of fibrosis formation due to hampered expression levels of genes involved in ECM organization. Furthermore, a detailed molecular fingerprint in different stages of muscle regeneration underlined a delay or even lack of a regenerative response to injury in obese mice. A time-lapse heatmap determined 81 differentially expressed genes (DEG) with at least three hits in our model at all-time points, suggesting key candidates with a high impact on muscle regeneration. Pathway analysis of the DEG revealed five pathways with a high confidence level: myeloid leukocyte migration, regulation of tumor necrosis factor production, CD4-positive, alpha-beta T cell differentiation, ECM organization, and toll-like receptor (TLR) signaling. Moreover, changes in complement-, Wnt-, and satellite cell-related genes were found to be impaired in obese animals after trauma. Furthermore, histological satellite cell evaluation showed lower satellite cell numbers in the obese model upon injury. Ankrd1, C3ar1, Ccl8, Mpeg1, and Myog expression levels were also verified by qPCR. In summary, increased fibrosis formation, the reduction of Pax7+ satellite cells as well as specific changes in gene expression and signaling pathways could explain the delay of tissue regeneration in obese mice post trauma.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Jens-Uwe Werner
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Sebastian Milerski
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Carmen M Hamp
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Tatjana Kuzenko
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Potsdam, Germany
| | - Pascal Gottmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Potsdam, Germany
| | - Luisa de Roy
- Institute of Orthopaedic Research and Biomechanics, Center for Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Daniela Warnecke
- Institute of Orthopaedic Research and Biomechanics, Center for Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Alireza Abaei
- Core facility "Small Animal Imaging", Ulm University, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Hospital, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Hospital, Ulm, Germany
| | - Lutz Dürselen
- Institute of Orthopaedic Research and Biomechanics, Center for Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Volker Rasche
- Core facility "Small Animal Imaging", Ulm University, Ulm, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Potsdam, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Ulm University Hospital for Pediatrics and Adolescent Medicine, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
40
|
Smith LR, Barton ER. Regulation of fibrosis in muscular dystrophy. Matrix Biol 2018; 68-69:602-615. [PMID: 29408413 DOI: 10.1016/j.matbio.2018.01.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 02/08/2023]
Abstract
The production of force and power are inherent properties of skeletal muscle, and regulated by contractile proteins within muscle fibers. However, skeletal muscle integrity and function also require strong connections between muscle fibers and their extracellular matrix (ECM). A well-organized and pliant ECM is integral to muscle function and the ability for many different cell populations to efficiently migrate through ECM is critical during growth and regeneration. For many neuromuscular diseases, genetic mutations cause disruption of these cytoskeletal-ECM connections, resulting in muscle fragility and chronic injury. Ultimately, these changes shift the balance from myogenic pathways toward fibrogenic pathways, culminating in the loss of muscle fibers and their replacement with fatty-fibrotic matrix. Hence a common pathological hallmark of muscular dystrophy is prominent fibrosis. This review will cover the salient features of muscular dystrophy pathogenesis, highlight the signals and cells that are important for myogenic and fibrogenic actions, and discuss how fibrosis alters the ECM of skeletal muscle, and the consequences of fibrosis in developing therapies.
Collapse
Affiliation(s)
- Lucas R Smith
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
41
|
α-smooth muscle actin is not a marker of fibrogenic cell activity in skeletal muscle fibrosis. PLoS One 2018; 13:e0191031. [PMID: 29320561 PMCID: PMC5761950 DOI: 10.1371/journal.pone.0191031] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 12/27/2017] [Indexed: 01/18/2023] Open
Abstract
α-Smooth muscle actin (α-SMA) is used as a marker for a subset of activated fibrogenic cells, myofibroblasts, which are regarded as important effector cells of tissue fibrogenesis. We address whether α-SMA-expressing myofibroblasts are detectable in fibrotic muscles of mdx5cv mice, a mouse model for Duchenne muscular dystrophy (DMD), and whether the α-SMA expression correlates with the fibrogenic function of intramuscular fibrogenic cells. α-SMA immunostaining signal was not detected in collagen I (GFP)-expressing cells in fibrotic muscles of ColI-GFP/mdx5cv mice, but it was readily detected in smooth muscle cells lining intramuscular blood vessel walls. α-SMA expression was detected by quantitative RT-PCR and Western blot in fibrogenic cells sorted from diaphragm and quadriceps muscles of the ColI-GFP/mdx5cv mice. Consistent with the more severe fibrosis in the ColI-GFP/mdx5cv diaphragm, the fibrogenic cells in the diaphragm exerted a stronger fibrogenic function than the fibrogenic cells in the quadriceps as gauged by their extracellular matrix gene expression. However, both gene and protein expression of α-SMA was lower in the diaphragm fibrogenic cells than in the quadriceps fibrogenic cells in the ColI-GFP/mdx5cv mice. We conclude that myofibroblasts are present in fibrotic skeletal muscles, but their expression of α-SMA is not detectable by immunostaining. The level of α-SMA expression by intramuscular fibrogenic cells does not correlate positively with the level of collagen gene expression or the severity of skeletal muscle fibrosis in the mdx5cv mice. α-SMA is not a functional marker of fibrogenic cells in skeletal muscle fibrosis associated with muscular dystrophy.
Collapse
|
42
|
Endothelial fibrosis induced by suppressed STAT3 expression mediated by signaling involving the TGF-β1/ALK5/Smad pathway. J Transl Med 2017; 97:1033-1046. [PMID: 28737766 DOI: 10.1038/labinvest.2017.61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 12/22/2022] Open
Abstract
During systemic inflammatory pathologies, mediators of inflammation circulate in the bloodstream and interact with endothelial cells (ECs), resulting in endothelial dysfunction that maintains and enhances the pathological condition. Inflammatory mediators change the protein expression profile of ECs, which become activated fibroblasts via endothelial-to-mesenchymal transition. This process is characterized by downregulated endothelial proteins and strongly upregulated fibrotic-specific genes and extracellular matrix-forming proteins. The main inductor of endothelial fibrosis is transforming growth factor-β1 (TGF-β1), which acts through the TGF-β1/activin receptor-like kinase 5 (ALK5)/Smads intracellular signaling pathway. The signal transducer and activator of transcription 3 (STAT3) is also involved in fibrosis in several tissues (e.g. heart and vascular system), where STAT3 signaling decreases TGF-β1-induced responses by directly interacting with Smad proteins, suggesting that decreased STAT3 could induce TGF-β1-mediated fibrosis. However, it is unknown if suppressed STAT3 expression induces EC fibrosis through a mechanism involving the TGF-β signaling pathway. The present study evaluated the fibrotic actions of STAT3 suppression in ECs and investigated TGF-β1/ALK5/Smad4 signaling pathway participation. Suppressed STAT3 expression stimulated fibrotic conversion in ECs, as mediated by protein expression reprograming that decreased endothelial marker expression and increased fibrotic and extracellular matrix protein levels. The potential mechanism underlying these changes was dependent on TGF-β1 secretion, the ALK5 activation pathway, and Smad4 translocation into the nucleus. We conclude that suppressed STAT3 expression converts ECs into activated fibroblasts via TGF-β1/ALK5/Smad4 signaling pathway involvement.
Collapse
|
43
|
Gillies AR, Chapman MA, Bushong EA, Deerinck TJ, Ellisman MH, Lieber RL. High resolution three-dimensional reconstruction of fibrotic skeletal muscle extracellular matrix. J Physiol 2016; 595:1159-1171. [PMID: 27859324 DOI: 10.1113/jp273376] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/24/2016] [Indexed: 01/18/2023] Open
Abstract
KEY POINTS Fibrosis occurs secondary to many skeletal muscle diseases and injuries, and can alter muscle function. It is unknown how collagen, the most abundant extracellular structural protein, alters its organization during fibrosis. Quantitative and qualitative high-magnification electron microscopy shows that collagen is organized into perimysial cables which increase in number in a model of fibrosis, and cables have unique interactions with collagen-producing cells. Fibrotic muscles are stiffer and have a higher concentration of collagen-producing cells. These results improve our understanding of the organization of fibrotic skeletal muscle extracellular matrix and identify novel structures that might be targeted by antifibrotic therapy. ABSTRACT Skeletal muscle extracellular matrix (ECM) structure and organization are not well understood, yet the ECM plays an important role in normal tissue homeostasis and disease processes. Fibrosis is common to many muscle diseases and is typically quantified based on an increase in ECM collagen. Through the use of multiple imaging modalities and quantitative stereology, we describe the structure and composition of wild-type and fibrotic ECM, we show that collagen in the ECM is organized into large bundles of fibrils, or collagen cables, and the number of these cables (but not their size) increases in desmin knockout muscle (a fibrosis model). The increase in cable number is accompanied by increased muscle stiffness and an increase in the number of collagen producing cells. Unique interactions between ECM cells and collagen cables were also observed and reconstructed by serial block face scanning electron microscopy. These results demonstrate that the muscle ECM is more highly organized than previously reported. Therapeutic strategies for skeletal muscle fibrosis should consider the organization of the ECM to target the structures and cells contributing to fibrotic muscle function.
Collapse
Affiliation(s)
- Allison R Gillies
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mark A Chapman
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.,Karolinska Institute, Stockholm, Sweden
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Thomas J Deerinck
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093, USA.,Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Richard L Lieber
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.,Department of Orthopaedic Surgery, University of California San Diego, La Jolla, CA, 92093, USA.,Rehabilitation Institute of Chicago, Chicago, IL, 60611, USA
| |
Collapse
|