1
|
Characterisation of Progressive Skeletal Muscle Fibrosis in the Mdx Mouse Model of Duchenne Muscular Dystrophy: An In Vivo and In Vitro Study. Int J Mol Sci 2022; 23:ijms23158735. [PMID: 35955872 PMCID: PMC9369129 DOI: 10.3390/ijms23158735] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disease leading to progressive muscle wasting, respiratory failure, and cardiomyopathy. Although muscle fibrosis represents a DMD hallmark, the organisation of the extracellular matrix and the molecular changes in its turnover are still not fully understood. To define the architectural changes over time in muscle fibrosis, we used an mdx mouse model of DMD and analysed collagen and glycosaminoglycans/proteoglycans content in skeletal muscle sections at different time points during disease progression and in comparison with age-matched controls. Collagen significantly increased particularly in the diaphragm, quadriceps, and gastrocnemius in adult mdx, with fibrosis significantly correlating with muscle degeneration. We also analysed collagen turnover pathways underlying fibrosis development in cultured primary quadriceps-derived fibroblasts. Collagen secretion and matrix metalloproteinases (MMPs) remained unaffected in both young and adult mdx compared to wt fibroblasts, whereas collagen cross-linking and tissue inhibitors of MMP (TIMP) expression significantly increased. We conclude that, in the DMD model we used, fibrosis mostly affects diaphragm and quadriceps with a higher collagen cross-linking and inhibition of MMPs that contribute differently to progressive collagen accumulation during fibrotic remodelling. This study offers a comprehensive histological and molecular characterisation of DMD-associated muscle fibrosis; it may thus provide new targets for tailored therapeutic interventions.
Collapse
|
2
|
Incretin-induced changes in the transcriptome of skeletal muscles of fa/fa Zucker rat (ZFR) with obesity, without diabetes. Int J Obes (Lond) 2022; 46:1311-1318. [PMID: 35383269 DOI: 10.1038/s41366-022-01114-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Glucagon-like peptide-1 receptor agonists (GLP-1ra) are increasingly used in treating type 2 diabetes and obesity. Exendin-4 (Ex-4), a long acting GLP-1ra, was previously reported to decrease oxidative stress in hepatocytes, adipocytes and skeletal muscle cells in obese nondiabetic fa/fa Zucker rats (ZFR), thereby improving insulin resistance. AIM We aimed first to identify Ex-4-induced changes in the transcriptome of skeletal muscle cells in ZFR. RESULTS Ontology analysis of differentially expressed genes (DEGs) in ZFR versus lean animals (LR) showed that the extracellular matrix (ECM) is the first most affected cellular compartment, followed by myofibrils and endoplasmic reticulum (ER). Interestingly, among 15 genes regulated in ZFR versus LR, 14 of them were inversely regulated by Ex-4, as further confirmed by RT-qPCR. Picro-Sirius red histological staining showed that decreased ECM fiber area in ZFR is partially restored by Ex-4. Ontology analysis of the myofibril compartment revealed that decreased muscle contractile function in ZFR is partially restored by Ex-4, as confirmed by Phalloidin histological staining that showed a partial restoration by Ex-4 of altered contractile apparatus in ZFR. Ontology analysis of ER DEGs in ZFR versus LR showed that some of them are related to the AMP-activated protein kinase (AMPK) signaling pathway. Phosphorylated AMPK levels were strongly increased in Ex-4-treated ZFR. CONCLUSION Altogether, our results suggest that GLP-1ra strongly restructure ECM and reinforce contractile capabilities in ZFR, while optimizing the cellular metabolism through AMPK.
Collapse
|
3
|
Brightwell CR, Kulkarni AS, Paredes W, Zhang K, Perkins JB, Gatlin KJ, Custodio M, Farooq H, Zaidi B, Pai R, Buttar RS, Tang Y, Melamed ML, Hostetter TH, Pessin JE, Hawkins M, Fry CS, Abramowitz MK. Muscle fibrosis and maladaptation occur progressively in CKD and are rescued by dialysis. JCI Insight 2021; 6:150112. [PMID: 34784301 PMCID: PMC8783691 DOI: 10.1172/jci.insight.150112] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Skeletal muscle maladaptation accompanies chronic kidney disease (CKD) and negatively impacts physical function. Emphasis in CKD has historically been placed on muscle fiber intrinsic deficits, such as altered protein metabolism and atrophy. However, targeted treatment of fiber intrinsic dysfunction has produced limited improvement, whereas alterations within the fiber extrinsic environment have scarcely been examined. METHODS We investigated alterations to the skeletal muscle interstitial environment with deep cellular phenotyping of biopsies from patients with CKD compared to age-matched control participants and performed transcriptome profiling to define the molecular underpinnings of CKD-associated muscle impairments. We further examined changes in the observed muscle maladaptation following initiation of dialysis therapy for kidney failure. RESULTS Patients with CKD exhibited a progressive fibrotic muscle phenotype, which was associated with impaired regenerative capacity and lower vascular density. The severity of these deficits was strongly associated with the degree of kidney dysfunction. Consistent with these profound deficits, CKD was associated with broad alterations to the muscle transcriptome, including altered extracellular matrix organization, downregulated angiogenesis, and altered expression of pathways related to stem cell self-renewal. Remarkably, despite the seemingly advanced nature of this fibrotic transformation, dialysis treatment rescued these deficits, restoring a healthier muscle phenotype. Furthermore, after accounting for muscle atrophy, strength and endurance improved after dialysis initiation. CONCLUSION These data identify a dialysis-responsive muscle fibrotic phenotype in CKD and suggest that the early dialysis window presents a unique opportunity of improved muscle regenerative capacity during which targeted interventions may achieve maximal impact. TRIAL REGISTRATION NCT01452412FUNDING. NIH.
Collapse
Affiliation(s)
- Camille R Brightwell
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, United States of America
| | - Ameya S Kulkarni
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - William Paredes
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Kehao Zhang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Jaclyn B Perkins
- Department of Nutrition and Metabolism, The University of Texas Medical Branch, Galveston, United States of America
| | - Knubian J Gatlin
- Department of Nutrition and Metabolism, The University of Texas Medical Branch, Galveston, United States of America
| | - Matthew Custodio
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Hina Farooq
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Bushra Zaidi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Rima Pai
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Rupinder S Buttar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Yan Tang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Michal L Melamed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Thomas H Hostetter
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, United States of America
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Meredith Hawkins
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | | | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| |
Collapse
|
4
|
Millward DJ. Interactions between Growth of Muscle and Stature: Mechanisms Involved and Their Nutritional Sensitivity to Dietary Protein: The Protein-Stat Revisited. Nutrients 2021; 13:729. [PMID: 33668846 PMCID: PMC7996181 DOI: 10.3390/nu13030729] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Childhood growth and its sensitivity to dietary protein is reviewed within a Protein-Stat model of growth regulation. The coordination of growth of muscle and stature is a combination of genetic programming, and of two-way mechanical interactions involving the mechanotransduction of muscle growth through stretching by bone length growth, the core Protein-Stat feature, and the strengthening of bone through muscle contraction via the mechanostat. Thus, growth in bone length is the initiating event and this is always observed. Endocrine and cellular mechanisms of growth in stature are reviewed in terms of the growth hormone-insulin like growth factor-1 (GH-IGF-1) and thyroid axes and the sex hormones, which together mediate endochondral ossification in the growth plate and bone lengthening. Cellular mechanisms of muscle growth during development are then reviewed identifying (a) the difficulties posed by the need to maintain its ultrastructure during myofibre hypertrophy within the extracellular matrix and the concept of muscle as concentric "bags" allowing growth to be conceived as bag enlargement and filling, (b) the cellular and molecular mechanisms involved in the mechanotransduction of satellite and mesenchymal stromal cells, to enable both connective tissue remodelling and provision of new myonuclei to aid myofibre hypertrophy and (c) the implications of myofibre hypertrophy for protein turnover within the myonuclear domain. Experimental data from rodent and avian animal models illustrate likely changes in DNA domain size and protein turnover during developmental and stretch-induced muscle growth and between different muscle fibre types. Growth of muscle in male rats during adulthood suggests that "bag enlargement" is achieved mainly through the action of mesenchymal stromal cells. Current understanding of the nutritional regulation of protein deposition in muscle, deriving from experimental studies in animals and human adults, is reviewed, identifying regulation by amino acids, insulin and myofibre volume changes acting to increase both ribosomal capacity and efficiency of muscle protein synthesis via the mechanistic target of rapamycin complex 1 (mTORC1) and the phenomenon of a "bag-full" inhibitory signal has been identified in human skeletal muscle. The final section deals with the nutritional sensitivity of growth of muscle and stature to dietary protein in children. Growth in length/height as a function of dietary protein intake is described in the context of the breastfed child as the normative growth model, and the "Early Protein Hypothesis" linking high protein intakes in infancy to later adiposity. The extensive paediatric studies on serum IGF-1 and child growth are reviewed but their clinical relevance is of limited value for understanding growth regulation; a role in energy metabolism and homeostasis, acting with insulin to mediate adiposity, is probably more important. Information on the influence of dietary protein on muscle mass per se as opposed to lean body mass is limited but suggests that increased protein intake in children is unable to promote muscle growth in excess of that linked to genotypic growth in length/height. One possible exception is milk protein intake, which cohort and cross-cultural studies suggest can increase height and associated muscle growth, although such effects have yet to be demonstrated by randomised controlled trials.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
5
|
Gabay Yehezkely R, Zaffryar-Eilot S, Kaganovsky A, Fainshtain Malka N, Aviram R, Livneh I, Hasson P. Intracellular Role for the Matrix-Modifying Enzyme Lox in Regulating Transcription Factor Subcellular Localization and Activity in Muscle Regeneration. Dev Cell 2020; 53:406-417.e5. [DOI: 10.1016/j.devcel.2020.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 02/23/2020] [Accepted: 04/03/2020] [Indexed: 12/31/2022]
|
6
|
Ding R, Horie M, Nagasaka S, Ohsumi S, Shimizu K, Honda H, Nagamori E, Fujita H, Kawamoto T. Effect of cell-extracellular matrix interaction on myogenic characteristics and artificial skeletal muscle tissue. J Biosci Bioeng 2020; 130:98-105. [PMID: 32278672 DOI: 10.1016/j.jbiosc.2020.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 01/23/2023]
Abstract
Although various types of artificial skeletal muscle tissue have been reported, the contractile forces generated by tissue-engineered artificial skeletal muscles remain to be improved for biological model and clinical applications. In this study, we investigated the effects of extracellular matrix (ECM) and supplementation of a small molecule, which has been reported to enhance α7β1 integrin expression (SU9516), on cell migration speed, cell fusion rate, myoblast (mouse C2C12 cells) differentiation and contractile force generation of tissue-engineered artificial skeletal muscles. When cells were cultured on varying ECM coated-surfaces, we observed significant enhancement in the migration speed, while the myotube formation (differentiation ratio) decreased in all except for cells cultured on Matrigel coated-surfaces. In contrast, SU9516 supplementation resulted in an increase in both the myotube width and differentiation ratio. Following combined culture with a Matrigel-coated surface and SU9516 supplementation, myotube width was further increased. Additionally, contractile forces produced by the tissue-engineered artificial skeletal muscles was augmented following combined culture. These findings indicate that regulation of the cell-ECM interaction is a promising approach to improve the function of tissue-engineered artificial skeletal muscles.
Collapse
Affiliation(s)
- Ran Ding
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanobu Horie
- Division of Biochemical Engineering, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Sumire Nagasaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Saki Ohsumi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan; Innovative Research Center for Preventive Medical Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Eiji Nagamori
- Department of Biomedical Engineering, Osaka Institute of Technology, 5-16-1 Omiya, Asahi-ku, Osaka 535-8585, Japan
| | - Hideaki Fujita
- WPI, Immunology Frontier Research Center, Institute of Scientific and Industrial Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takuo Kawamoto
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan; Division of Biological Chemistry, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
7
|
Liu C, Li L, Ge M, Gu L, Wang M, Zhang K, Su Y, Zhang Y, Liu C, Lan M, Yu Y, Wang T, Li Q, Zhao Y, Yu Z, Li N, Meng Q. Overexpression of miR-29 Leads to Myopathy that Resemble Pathology of Ullrich Congenital Muscular Dystrophy. Cells 2019; 8:cells8050459. [PMID: 31096686 PMCID: PMC6562860 DOI: 10.3390/cells8050459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022] Open
Abstract
Ullrich congenital muscular dystrophy (UCMD) bring heavy burden to patients’ families and society. Because the incidence of this disease is very low, studies in patients are extremely limited. Animal models of this disease are indispensable. UCMD belongs to extracellular matrix-related diseases. However, the disease models constructed by knocking out some pathogenic genes of human, such as the Col6a1, Col6a2, or Col6a3 gene, of mice could not mimic UCMD. The purpose of this study is to construct a mouse model which can resemble the pathology of UCMD. miR-29 is closely related to extracellular matrix deposition of tissues and organs. To address this issue, we developed a mouse model for overexpression miR-29 using Tet-on system. In the muscle-specific miR-29ab1 cluster transgenic mice model, we found that mice exhibited dyskinesia, dyspnea, and spinal anomaly. The skeletal muscle was damaged and regenerated. At the same time, we clarify the molecular mechanism of the role of miR-29 in this process. Different from human, Col4a1 and Col4a2, target genes of miR-29, are the key pathogenic genes associating with these phenotypes. This mouse model simulates the human clinical and pathological characteristics of UCMD patients and is helpful for the subsequent research and treatment of UCMD.
Collapse
Affiliation(s)
- Chuncheng Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
- The Institute of Bioengineering and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China.
| | - Lei Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Mengxu Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Lijie Gu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Meng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Kuo Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yang Su
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yuying Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Chang Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Miaomiao Lan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yingying Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Tongtong Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Qiuyan Li
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yaofeng Zhao
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Zhengquan Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Ning Li
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Qingyong Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
D'Andrea P, Sciancalepore M, Veltruska K, Lorenzon P, Bandiera A. Epidermal Growth Factor - based adhesion substrates elicit myoblast scattering, proliferation, differentiation and promote satellite cell myogenic activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:504-517. [PMID: 30343052 DOI: 10.1016/j.bbamcr.2018.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022]
Abstract
The biochemical properties of muscle extracellular matrix are essential for stem cell adhesion, motility, proliferation and myogenic development. Recombinant elastin-like polypeptides are synthetic polypeptides that, besides maintaining some properties of the native protein, can be tailored by fusing bioactive sequences to their C-terminal. Our laboratory synthesized several Human Elastin-Like Polypeptides (HELP) derived from the sequence of human tropoelastin. Here, we developed a novel HELP family member by fusing the elastin-like backbone to the sequence of human Epidermal Growth Factor. We employed this synthetic protein, named HEGF, either alone or in combination with other proteins of the HELP family carrying RGD-integrin binding sites, as adhesion substrate for C2C12 myoblasts and satellite cells primary cultures. Adhesion of myoblasts to HEGF-based substrates induced scattering, decreased adhesion and cytoskeleton assembly; the concomitant presence of the RGD motifs potentiated all these effects. Recombinant substrates induced myoblasts proliferation, differentiation and the development of multinucleated myotubes, thus favoring myoblasts expansion and preserving their myogenic potential. The effects induced by adhesion substrates were inhibited by AG82 (Tyrphostin 25) and herbimycin A, indicating their dependence on the activation of both the EGF receptor and the tyrosine kinase c-src. Finally, HEGF increased the number of muscle stem cells (satellite cells) derived from isolated muscle fibers in culture, thus highlighting its potential as a novel substrate for skeletal muscle regeneration strategies.
Collapse
Affiliation(s)
- Paola D'Andrea
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy.
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy; Centre for Neuroscience B.R.A.I.N., University of Trieste, I-34127 Trieste, Italy
| | - Katerina Veltruska
- Department of Surface and Plasma Science, Faculty of Mathematics and Physics, Charles University in Prague V Holešovičkách 747/2, Praha 8, Czech Republic
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy; Centre for Neuroscience B.R.A.I.N., University of Trieste, I-34127 Trieste, Italy
| | - Antonella Bandiera
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy
| |
Collapse
|
9
|
Abramowitz MK, Paredes W, Zhang K, Brightwell CR, Newsom JN, Kwon HJ, Custodio M, Buttar RS, Farooq H, Zaidi B, Pai R, Pessin JE, Hawkins M, Fry CS. Skeletal muscle fibrosis is associated with decreased muscle inflammation and weakness in patients with chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1658-F1669. [PMID: 30280599 DOI: 10.1152/ajprenal.00314.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Muscle dysfunction is an important cause of morbidity among patients with chronic kidney disease (CKD). Although muscle fibrosis is present in a CKD rodent model, its existence in humans and its impact on physical function are currently unknown. We examined isometric leg extension strength and measures of skeletal muscle fibrosis and inflammation in vastus lateralis muscle from CKD patients ( n = 10) and healthy, sedentary controls ( n = 10). Histochemistry and immunohistochemistry were used to assess muscle collagen and macrophage and fibro/adipogenic progenitor (FAP) cell populations, and RT-qPCR was used to assess muscle-specific inflammatory marker expression. Muscle collagen content was significantly greater in CKD compared with control (18.8 ± 2.1 vs. 11.7 ± 0.7% collagen area, P = 0.008), as was staining for collagen I, pro-collagen I, and a novel collagen-hybridizing peptide that binds remodeling collagen. Muscle collagen was inversely associated with leg extension strength in CKD ( r = -0.74, P = 0.01). FAP abundance was increased in CKD, was highly correlated with muscle collagen ( r = 0.84, P < 0.001), and was inversely associated with TNF-α expression ( r = -0.65, P = 0.003). TNF-α, CD68, CCL2, and CCL5 mRNA were significantly lower in CKD than control, despite higher serum TNF-α and IL-6. Immunohistochemistry confirmed fewer CD68+ and CD11b+ macrophages in CKD muscle. In conclusion, skeletal muscle collagen content is increased in humans with CKD and is associated with functional parameters. Muscle fibrosis correlated with increased FAP abundance, which may be due to insufficient macrophage-mediated TNF-α secretion. These data provide a foundation for future research elucidating the mechanisms responsible for this newly identified human muscle pathology.
Collapse
Affiliation(s)
| | - William Paredes
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Kehao Zhang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Camille R Brightwell
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| | - Julia N Newsom
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| | - Hyok-Joon Kwon
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers-The State University of New Jersey , New Brunswick, New Jersey
| | - Matthew Custodio
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rupinder S Buttar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Hina Farooq
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Bushra Zaidi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rima Pai
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Meredith Hawkins
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch , Galveston, Texas
| |
Collapse
|
10
|
Lee EJ, Nam JH, Choi I. Fibromodulin modulates myoblast differentiation by controlling calcium channel. Biochem Biophys Res Commun 2018; 503:580-585. [PMID: 29913145 DOI: 10.1016/j.bbrc.2018.06.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/10/2018] [Indexed: 10/28/2022]
Abstract
Fibromodulin (FMOD) is a proteoglycan present in extracellular matrix (ECM). Based on our previous findings that FMOD controls myoblast differentiation by regulating the gene expressions of collagen type I alpha 1 (COL1α1) and integral membrane protein 2 A (Itm2a), we undertook this study to investigate relationships between FMOD and calcium channels and to understand further the mechanism by which they control myoblast differentiation. Gene expression studies and luciferase reporter assays showed FMOD affected calcium channel gene expressions by regulating calcium channel gene promoter, and patch-clamp experiments showed both L- and T-type calcium channel currents were almost undetectable in FMOD knocked down cells. In addition, gene knock-down studies demonstrated the COL1α1 and Itm2a genes both regulate the expressions of calcium channel genes. Studies using a cardiotoxin-induced mouse muscle injury model demonstrated calcium channels play important roles in the regeneration of muscle tissue, possibly by promoting the differentiation of muscle stem cells (MSCs). Summarizing, the study demonstrates ECM components secreted by myoblasts during differentiation provide an essential environment for muscle differentiation and regeneration.
Collapse
Affiliation(s)
- Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University, College of Medicine, Gyeongju, 38067, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
11
|
Implication of SPARC in the modulation of the extracellular matrix and mitochondrial function in muscle cells. PLoS One 2018; 13:e0192714. [PMID: 29420632 PMCID: PMC5805355 DOI: 10.1371/journal.pone.0192714] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/29/2018] [Indexed: 01/16/2023] Open
Abstract
Secreted protein, acidic and rich in cysteine (SPARC) is differentially associated with cell proliferation and extracellular matrix (ECM) assembly. We show here the effect of exogenous SPARC inhibition/induction on ECM and mitochondrial proteins expression and on the differentiation of C2C12 cells. The cells were cultured in growth medium (GM) supplemented with different experimental conditions. The differentiation of myoblasts was studied for 5 days, the expressions of ECM and mitochondrial proteins were measured and the formation of the myotubes was quantified after exogenous induction/inhibition of SPARC. The results indicate that the addition of recombinant SPARC protein (rSPARC) in cell culture medium increased the differentiation of C2C12 myoblasts and myogenin expression during the myotube formation. However, the treatment with antibody specific for SPARC (anti-SPARC) prevented the differentiation and decreased myogenin expression. The induction of SPARC in the proliferating and differentiating C2C12 cells increased collagen 1a1 protein expression, whereas the inhibition decreased it. The effects on fibronectin protein expression were opposite. Furthermore, the addition of rSPARC in C2C12 myoblast increased the expression of mitochondrial proteins, ubiquinol-cytochrome c reductase core protein II (UQCRC2) and succinate dehydrogenase iron-sulfur subunit (SDHB), whereas the anti-SPARC decreased them. During the differentiation, only the anti-SPARC had the effects on mitochondrial proteins, NADH dehydrogenase ubiquinone 1 beta subcomplex subunit 8 (NADHB8), SDHB and cytochrome c oxidase 1 (MTCO1). Thus, SPARC plays a crucial role in the proliferation and differentiation of C2C12 and may be involved in the link between the ECM remodeling and mitochondrial function.
Collapse
|
12
|
Goldman SM, Corona BT. Co-delivery of micronized urinary bladder matrix damps regenerative capacity of minced muscle grafts in the treatment of volumetric muscle loss injuries. PLoS One 2017; 12:e0186593. [PMID: 29040321 PMCID: PMC5645132 DOI: 10.1371/journal.pone.0186593] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
Minced muscle grafts (MG) promote de novo muscle fiber regeneration and neuromuscular strength recovery in small and large animal models of volumetric muscle loss. The most noteworthy limitation of this approach is its reliance on a finite supply of donor tissue. To address this shortcoming, this study sought to evaluate micronized acellular urinary bladder matrix (UBM) as a scaffolding to promote in vivo expansion of this MG therapy in a rat model. Rats received volumetric muscle loss injuries to the tibialis anterior muscle of their left hind limb which were either left untreated or repaired with minced muscle graft at dosages of 50% and 100% of the defect mass, urinary bladder matrix in isolation, or a with an expansion product consisting of a combination of the two putative therapies in which the minced graft is delivered at a dosage of 50% of the defect mass. Rats survived to 2 and 8 weeks post injury before functional (in vivo neuromuscular strength), histological, morphological, and biochemical analyses were performed. Rats treated with the expansion product exhibited improved neuromuscular function relative to untreated VML after an 8 week time period following injury. This improvement in functional capacity, however, was accompanied with a concomitant reduction in graft mediated regeneration, as evidenced cell lineage tracing enable by a transgenic GFP expressing donor, and a mixed histological outcome indicating coincident fibrous matrix deposition with interspersed islands of nascent muscle fibers. Furthermore, quantitative immunofluorescence and transcriptional analysis following the 2 week time point suggests an exacerbated immune response to the UBM as a possible nidus for the observed suboptimal regenerative outcome. Moving forward, efforts related to the development of a MG expansion product should carefully consider the effects of the host immune response to candidate biomaterials in order to avoid undesirable dysregulation of pro-regenerative cross talk between the immune system and myogenic processes.
Collapse
Affiliation(s)
- Stephen M. Goldman
- United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States of America
| | - Benjamin T. Corona
- United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States of America
| |
Collapse
|
13
|
Wong TY, Chang CH, Yu CH, Huang LLH. Hyaluronan keeps mesenchymal stem cells quiescent and maintains the differentiation potential over time. Aging Cell 2017; 16:451-460. [PMID: 28474484 PMCID: PMC5418204 DOI: 10.1111/acel.12567] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hyaluronan (HA), an abundant polysaccharide found in human bodies, plays a role in the mesenchymal stem cells (MSCs) maintenance. We had previously found that HA prolonged the lifespan, and prevented the cellular aging of murine adipose-derived stromal cells. Recently, we had also summarized the potential pathways associated with HA regulation in human MSCs. In this study, we used the human placenta-derived MSCs (PDMSC) to investigate the effectiveness of HA in maintaining the PDMSC. We found that coating the culture surface coated with 30 μg cm-2 of HA (C) led to cluster growth of PDMSC, and maintained a higher number of PDMSC in quiescence compared to those grown on the normal tissue culture surface (T). PDMSC were treated for either 4 (short-term) or 19 (long-term) consecutive passages. PDMSC which were treated with HA for 19 consecutive passages had reduced cell enlargement, preserved MSCs biomarker expressions and osteogenic potential when compared to those grown only on T. The PDMSC transferred to T condition after long-term HA treatment showed preserved replicative capability compared to those on only T. The telomerase activity of the HA-treated PDMSC was also higher than that of untreated PDMSC. These data suggested a connection between HA and MSC maintenance. We suggest that HA might be regulating the distribution of cytoskeletal proteins on cell spreading in the event of quiescence to preserve MSC stemness. Maintenance of MSCs stemness delayed cellular aging, leading to the anti-aging phenotype of PDMSC.
Collapse
Affiliation(s)
- Tzyy Yue Wong
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
| | - Chiung-Hsin Chang
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Chen-Hsiang Yu
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Lynn L. H. Huang
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Department of Biotechnology and Bioindustry Sciences; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Institute of Clinical Medicine; College of Medicine; National Cheng Kung University; Tainan Taiwan
- Research Center of Excellence in Regenerative Medicine; National Cheng Kung University; Tainan Taiwan
- Advanced Optoelectronic Technology Center; National Cheng Kung University; Tainan Taiwan
| |
Collapse
|
14
|
Lund DK, McAnulty P, Siegel AL, Cornelison D. Methods for Observing and Quantifying Muscle Satellite Cell Motility and Invasion In Vitro. Methods Mol Biol 2017; 1556:303-315. [PMID: 28247357 DOI: 10.1007/978-1-4939-6771-1_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Motility and/or chemotaxis of satellite cells has been suggested or observed in multiple in vitro and in vivo contexts. Satellite cell motility also affects the efficiency of muscle regeneration, particularly in the context of engrafted exogenous cells. Consequently, there is keen interest in determining what cell-autonomous and environmental factors influence satellite cell motility and chemotaxis in vitro and in vivo. In addition, the ability of activated satellite cells to relocate in vivo would suggest that they must be able to invade and transit through the extracellular matrix (ECM), which is supported by studies in which alteration or addition of matrix metalloprotease (MMP) activity enhanced the spread of engrafted satellite cells. However, despite its potential importance, analysis of satellite cell motility or invasion quantitatively even in an in vitro setting can be difficult; one of the most powerful techniques for overcoming these difficulties is timelapse microscopy. Identification and longitudinal evaluation of individual cells over time permits not only quantification of variations in motility due to intrinsic or extrinsic factors, it permits observation and analysis of other (frequently unsuspected) cellular activities as well. We describe here three protocols developed in our group for quantitatively analyzing satellite cell motility over time in two dimensions on purified ECM substrates, in three dimensions on a living myofiber, and in three dimensions through an artificial matrix.
Collapse
Affiliation(s)
- Dane K Lund
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO, 65211 7310, USA
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Patrick McAnulty
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO, 65211 7310, USA
- The Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ashley L Siegel
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO, 65211 7310, USA
- Elemental Enzymes, St. Louis, MO, USA
| | - Ddw Cornelison
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, MO, 65211 7310, USA.
| |
Collapse
|
15
|
Corona BT, Greising SM. Challenges to acellular biological scaffold mediated skeletal muscle tissue regeneration. Biomaterials 2016; 104:238-46. [DOI: 10.1016/j.biomaterials.2016.07.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/11/2016] [Accepted: 07/16/2016] [Indexed: 02/08/2023]
|
16
|
Ikeda K, Ito A, Sato M, Kawabe Y, Kamihira M. Improved contractile force generation of tissue-engineered skeletal muscle constructs by IGF-I and Bcl-2 gene transfer with electrical pulse stimulation. Regen Ther 2016; 3:38-44. [PMID: 31245471 PMCID: PMC6581813 DOI: 10.1016/j.reth.2015.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 12/21/2015] [Accepted: 12/24/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Tissue-engineered skeletal muscle constructs should be designed to generate contractile force with directional movement. Because electrical impulses from a somatic nervous system are crucial for in vivo skeletal muscle development, electrical pulse stimulation (EPS) culture as an artificial exercise is essential to fabricate functional skeletal muscle tissues in vitro. To further improve muscle functions, the activation of cell-signaling pathways from myogenic growth factors, such as insulin-like growth factor (IGF)-I, is also important. Because tissue-engineered skeletal muscle constructs should maintain a high cell-dense structure, the expression of an anti-apoptotic factor, such as B-cell lymphoma 2 (Bcl-2), could be effective in preventing cell death. Methods In the present study, myoblasts were genetically modified with inducible expression units of IGF-I and Bcl-2 genes, and the tissue-engineered skeletal muscle constructs fabricated from the myoblasts were cultured under continuous EPS. Results Overexpression of IGF-I gene induced muscular hypertrophy in the muscle tissue constructs, and Bcl-2-overexpressing myoblasts formed significantly cell-dense and viable muscle tissue constructs. Furthermore, the combination of IGF-I and Bcl-2 gene transfer with EPS culture highly improved the force generation of the tissue-engineered skeletal muscle constructs. Conclusions This approach has the potential to yield functional skeletal muscle substitutes with high force generation ability.
Collapse
Affiliation(s)
- Kazushi Ikeda
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masanori Sato
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masamichi Kamihira
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
17
|
Smith LR, Hammers DW, Sweeney HL, Barton ER. Increased collagen cross-linking is a signature of dystrophin-deficient muscle. Muscle Nerve 2016; 54:71-8. [PMID: 26616495 PMCID: PMC5067682 DOI: 10.1002/mus.24998] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2015] [Indexed: 01/18/2023]
Abstract
Introduction Collagen cross‐linking is a key parameter in extracellular matrix (ECM) maturation, turnover, and stiffness. We examined aspects of collagen cross‐linking in dystrophin‐deficient murine, canine, and human skeletal muscle. Methods DMD patient biopsies and samples from mdx mice and golden retriever muscular dystrophy dog samples (with appropriate controls) were analyzed. Collagen cross‐linking was evaluated using solubility and hydroxyproline assays. Expression of the cross‐linking enzyme lysyl oxidase (LOX) was determined by real‐time polymerase chain reaction, immunoblotting, and immunofluorescence. Results LOX protein levels are increased in dystrophic muscle from all species evaluated. Dystrophic mice and dogs had significantly higher cross‐linked collagen than controls, especially in the diaphragm. Distribution of intramuscular LOX was heterogeneous in all samples, but it increased in frequency and intensity in dystrophic muscle. Conclusion These findings implicate elevated collagen cross‐linking as an important component of the disrupted ECM in dystrophic muscles, and heightened cross‐linking is evident in mouse, dog, and man. Muscle Nerve54: 71–78, 2016
Collapse
Affiliation(s)
- Lucas R Smith
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David W Hammers
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA.,Myology Institute, University of Florida, Gainesville, Florida, USA
| | - H Lee Sweeney
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, Florida, USA.,Myology Institute, University of Florida, Gainesville, Florida, USA
| | - Elisabeth R Barton
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Myology Institute, University of Florida, Gainesville, Florida, USA.,Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, 1864 Stadium Road, 124 Florida Gym, Gainesville, Florida, 32611, USA
| |
Collapse
|
18
|
Riederer I, Bonomo AC, Mouly V, Savino W. Laminin therapy for the promotion of muscle regeneration. FEBS Lett 2015; 589:3449-53. [DOI: 10.1016/j.febslet.2015.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/25/2015] [Accepted: 10/06/2015] [Indexed: 12/18/2022]
|
19
|
Holland A, Murphy S, Dowling P, Ohlendieck K. Pathoproteomic profiling of the skeletal muscle matrisome in dystrophinopathy associated myofibrosis. Proteomics 2015; 16:345-66. [PMID: 26256116 DOI: 10.1002/pmic.201500158] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/12/2015] [Accepted: 07/24/2015] [Indexed: 12/14/2022]
Abstract
The gradual accumulation of collagen and associated proteins of the extracellular matrix is a crucial myopathological parameter of many neuromuscular disorders. Progressive tissue damage and fibrosis play a key pathobiochemical role in the dysregulation of contractile functions and often correlates with poor motor outcome in muscular dystrophies. Following a brief introduction into the role of the extracellular matrix in skeletal muscles, we review here the proteomic profiling of myofibrosis and its intrinsic role in X-linked muscular dystrophy. Although Duchenne muscular dystrophy is primarily a disease of the membrane cytoskeleton, one of its most striking histopathological features is a hyperactive connective tissue and tissue scarring. We outline the identification of novel factors involved in the modulation of the extracellular matrix in muscular dystrophy, such as matricellular proteins. The establishment of novel proteomic markers will be helpful in improving the diagnosis, prognosis, and therapy monitoring in relation to fibrotic substitution of contractile tissue. In the future, the prevention of fibrosis will be crucial for providing optimum conditions to apply novel pharmacological treatments, as well as establish cell-based approaches or gene therapeutic interventions. The elimination of secondary abnormalities in the matrisome promises to reduce tissue scarring and the loss of skeletal muscle elasticity.
Collapse
Affiliation(s)
- Ashling Holland
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
20
|
Dahlquist DT, Dieter BP, Koehle MS. Plausible ergogenic effects of vitamin D on athletic performance and recovery. J Int Soc Sports Nutr 2015; 12:33. [PMID: 26288575 PMCID: PMC4539891 DOI: 10.1186/s12970-015-0093-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/09/2015] [Indexed: 01/06/2023] Open
Abstract
The purpose of this review is to examine vitamin D in the context of sport nutrition and its potential role in optimizing athletic performance. Vitamin D receptors (VDR) and vitamin D response elements (VDREs) are located in almost every tissue within the human body including skeletal muscle. The hormonally-active form of vitamin D, 1,25-dihydroxyvitamin D, has been shown to play critical roles in the human body and regulates over 900 gene variants. Based on the literature presented, it is plausible that vitamin D levels above the normal reference range (up to 100 nmol/L) might increase skeletal muscle function, decrease recovery time from training, increase both force and power production, and increase testosterone production, each of which could potentiate athletic performance. Therefore, maintaining higher levels of vitamin D could prove beneficial for athletic performance. Despite this situation, large portions of athletic populations are vitamin D deficient. Currently, the research is inconclusive with regards to the optimal intake of vitamin D, the specific forms of vitamin D one should ingest, and the distinct nutrient-nutrient interactions of vitamin D with vitamin K that affect arterial calcification and hypervitaminosis. Furthermore, it is possible that dosages exceeding the recommendations for vitamin D (i.e. dosages up to 4000-5000 IU/day), in combination with 50 to 1000 mcg/day of vitamin K1 and K2 could aid athletic performance. This review will investigate these topics, and specifically their relevance to athletic performance.
Collapse
Affiliation(s)
- Dylan T Dahlquist
- UBC Environmental Physiology Laboratory, School of Kinesiology, University of British Columbia, Vancouver, BC Canada
| | - Brad P Dieter
- Providence Medical Research Center, Providence Sacred Heart Medical Center and Children's Hospital, Research Discovery Lab, Spokane, WA 99204 USA
| | - Michael S Koehle
- Simon Fraser University, Biomedical Physiology and Kinesiology (BPK), 8888 University Drive - Burnaby, Vancouver, BC V5A 1S6 Canada
| |
Collapse
|
21
|
Hjorth M, Norheim F, Meen AJ, Pourteymour S, Lee S, Holen T, Jensen J, Birkeland KI, Martinov VN, Langleite TM, Eckardt K, Drevon CA, Kolset SO. The effect of acute and long-term physical activity on extracellular matrix and serglycin in human skeletal muscle. Physiol Rep 2015; 3:e12473. [PMID: 26290530 PMCID: PMC4562559 DOI: 10.14814/phy2.12473] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 12/20/2022] Open
Abstract
Remodeling of extracellular matrix (ECM), including regulation of proteoglycans in skeletal muscle can be important for physiological adaptation to exercise. To investigate the effects of acute and long-term exercise on the expression of ECM-related genes and proteoglycans in particular, 26 middle-aged, sedentary men underwent a 12 weeks supervised endurance and strength training intervention and two acute, 45 min bicycle tests (70% VO2max), one at baseline and one after 12 weeks of training. Total gene expression in biopsies from m. vastus lateralis was measured with deep mRNA sequencing. After 45 min of bicycling approximately 550 gene transcripts were >50% upregulated. Of these, 28 genes (5%) were directly related to ECM. In response to long-term exercise of 12 weeks 289 genes exhibited enhanced expression (>50%) and 20% of them were ECM related. Further analyses of proteoglycan mRNA expression revealed that more than half of the proteoglycans expressed in muscle were significantly enhanced after 12 weeks intervention. The proteoglycan serglycin (SRGN) has not been studied in skeletal muscle and was one of few proteoglycans that showed increased expression after acute (2.2-fold, P < 0.001) as well as long-term exercise (1.4-fold, P < 0.001). Cultured, primary human skeletal muscle cells expressed and secreted SRGN. When the expression of SRGN was knocked down, the expression and secretion of serpin E1 (SERPINE1) increased. In conclusion, acute and especially long-term exercise promotes enhanced expression of several ECM components and proteoglycans. SRGN is a novel exercise-regulated proteoglycan in skeletal muscle with a potential role in exercise adaptation.
Collapse
Affiliation(s)
- Marit Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Astri J Meen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Shirin Pourteymour
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torgeir Holen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Kåre I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital and Institute of Clinical Medicine University of Oslo, Oslo, Norway
| | - Vladimir N Martinov
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torgrim M Langleite
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital and Institute of Clinical Medicine University of Oslo, Oslo, Norway
| | - Kristin Eckardt
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Svein O Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Guo B, Greenwood PL, Cafe LM, Zhou G, Zhang W, Dalrymple BP. Transcriptome analysis of cattle muscle identifies potential markers for skeletal muscle growth rate and major cell types. BMC Genomics 2015; 16:177. [PMID: 25887672 PMCID: PMC4364331 DOI: 10.1186/s12864-015-1403-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/24/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to identify markers for muscle growth rate and the different cellular contributors to cattle muscle and to link the muscle growth rate markers to specific cell types. RESULTS The expression of two groups of genes in the longissimus muscle (LM) of 48 Brahman steers of similar age, significantly enriched for "cell cycle" and "ECM (extracellular matrix) organization" Gene Ontology (GO) terms was correlated with average daily gain/kg liveweight (ADG/kg) of the animals. However, expression of the same genes was only partly related to growth rate across a time course of postnatal LM development in two cattle genotypes, Piedmontese x Hereford (high muscling) and Wagyu x Hereford (high marbling). The deposition of intramuscular fat (IMF) altered the relationship between the expression of these genes and growth rate. K-means clustering across the development time course with a large set of genes (5,596) with similar expression profiles to the ECM genes was undertaken. The locations in the clusters of published markers of different cell types in muscle were identified and used to link clusters of genes to the cell type most likely to be expressing them. Overall correspondence between published cell type expression of markers and predicted major cell types of expression in cattle LM was high. However, some exceptions were identified: expression of SOX8 previously attributed to muscle satellite cells was correlated with angiogenesis. Analysis of the clusters and cell types suggested that the "cell cycle" and "ECM" signals were from the fibro/adipogenic lineage. Significant contributions to these signals from the muscle satellite cells, angiogenic cells and adipocytes themselves were not as strongly supported. Based on the clusters and cell type markers, sets of five genes predicted to be representative of fibro/adipogenic precursors (FAPs) and endothelial cells, and/or ECM remodelling and angiogenesis were identified. CONCLUSIONS Gene sets and gene markers for the analysis of many of the major processes/cell populations contributing to muscle composition and growth have been proposed, enabling a consistent interpretation of gene expression datasets from cattle LM. The same gene sets are likely to be applicable in other cattle muscles and in other species.
Collapse
Affiliation(s)
- Bing Guo
- Key Laboratory of Meat Processing and Quality Control, Synergetic Innovation Centre of Food Safety and Nutrition, College of Food Science and Technology, Nanjing Agriculture University, Nanjing, 210095, P. R. China.
- CSIRO Agriculture Flagship, St. Lucia, QLD, 4067, Australia.
| | - Paul L Greenwood
- CSIRO Agriculture Flagship, Armidale, NSW, 2350, Australia.
- NSW Department of Primary Industries, University of New England, Armidale, NSW, 2351, Australia.
| | - Linda M Cafe
- NSW Department of Primary Industries, University of New England, Armidale, NSW, 2351, Australia.
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, Synergetic Innovation Centre of Food Safety and Nutrition, College of Food Science and Technology, Nanjing Agriculture University, Nanjing, 210095, P. R. China.
| | - Wangang Zhang
- Key Laboratory of Meat Processing and Quality Control, Synergetic Innovation Centre of Food Safety and Nutrition, College of Food Science and Technology, Nanjing Agriculture University, Nanjing, 210095, P. R. China.
| | | |
Collapse
|
23
|
Goody MF, Sher RB, Henry CA. Hanging on for the ride: adhesion to the extracellular matrix mediates cellular responses in skeletal muscle morphogenesis and disease. Dev Biol 2015; 401:75-91. [PMID: 25592225 DOI: 10.1016/j.ydbio.2015.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 10/24/2022]
Abstract
Skeletal muscle specification and morphogenesis during early development are critical for normal physiology. In addition to mediating locomotion, skeletal muscle is a secretory organ that contributes to metabolic homeostasis. Muscle is a highly adaptable tissue, as evidenced by the ability to increase muscle cell size and/or number in response to weight bearing exercise. Conversely, muscle wasting can occur during aging (sarcopenia), cancer (cancer cachexia), extended hospital stays (disuse atrophy), and in many genetic diseases collectively known as the muscular dystrophies and myopathies. It is therefore of great interest to understand the cellular and molecular mechanisms that mediate skeletal muscle development and adaptation. Muscle morphogenesis transforms short muscle precursor cells into long, multinucleate myotubes that anchor to tendons via the myotendinous junction. This process requires carefully orchestrated interactions between cells and their extracellular matrix microenvironment. These interactions are dynamic, allowing muscle cells to sense biophysical, structural, organizational, and/or signaling changes within their microenvironment and respond appropriately. In many musculoskeletal diseases, these cell adhesion interactions are disrupted to such a degree that normal cellular adaptive responses are not sufficient to compensate for accumulating damage. Thus, one major focus of current research is to identify the cell adhesion mechanisms that drive muscle morphogenesis, with the hope that understanding how muscle cell adhesion promotes the intrinsic adaptability of muscle tissue during development may provide insight into potential therapeutic approaches for muscle diseases. Our objectives in this review are to highlight recent studies suggesting conserved roles for cell-extracellular matrix adhesion in vertebrate muscle morphogenesis and cellular adaptive responses in animal models of muscle diseases.
Collapse
Affiliation(s)
- Michelle F Goody
- School of Biology and Ecology, University of Maine, Orono, ME 04469, United States
| | - Roger B Sher
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, United States
| | - Clarissa A Henry
- School of Biology and Ecology, University of Maine, Orono, ME 04469, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, United States; Institute for Molecular Biophysics, University of Maine, Orono, ME 04469, United States.
| |
Collapse
|
24
|
Schering L, Hoene M, Kanzleiter T, Jähnert M, Wimmers K, Klaus S, Eckel J, Weigert C, Schürmann A, Maak S, Jonas W, Sell H. Identification of novel putative adipomyokines by a cross-species annotation of secretomes and expression profiles. Arch Physiol Biochem 2015; 121:194-205. [PMID: 26599229 DOI: 10.3109/13813455.2015.1092044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue and skeletal muscle are organs that respond strongly to obesity and physical activity exhibiting high secretory activity. To identify novel putative adipomyokines, comparative expression studies of skeletal muscle and adipose tissue of lean (C57BL/6J) and obese (C57BL/6J on a high-fat diet and NZO) mice, of sedentary and endurance trained C57BL/6J mice and of cattle characterized by different amounts of intramuscular fat were combined with human secretome data and scored. In highly regulated transcripts, we identified 119 myokines, 79 adipokines and 22 adipomyokines. Network analysis of these candidates revealed remodelling of extracellular matrix and tissue fibrosis as relevant functions of several of these candidates. Given the pathophysiogical relevance of fibrosis for adipose-muscle-cross-talk in obesity and type 2 diabetes and its physiological role in exercise adaptation and meat quality of farm animals, they represent interesting candidates for further investigations in different research areas and species.
Collapse
Affiliation(s)
- Lisa Schering
- a Institute for Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology , Dummerstorf , Germany
| | - Miriam Hoene
- b Division of Clinical Chemistry and Pathobiochemistry , Department of Internal Medicine IV, University Hospital Tübingen , Tübingen , Germany
| | - Timo Kanzleiter
- c Department of Experimental Diabetology , German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany and German Center for Diabetes Research (DZD) , Neuherberg , Germany
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
| | - Markus Jähnert
- c Department of Experimental Diabetology , German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany and German Center for Diabetes Research (DZD) , Neuherberg , Germany
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
| | - Klaus Wimmers
- e Institute for Genome Biology, Leibniz Institute for Farm Animal Biology , Dummerstorf , Germany
| | - Susanne Klaus
- f Group of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke , Nuthetal , Germany , and
| | - Jürgen Eckel
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
- g Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center , Düsseldorf , Germany
| | - Cora Weigert
- b Division of Clinical Chemistry and Pathobiochemistry , Department of Internal Medicine IV, University Hospital Tübingen , Tübingen , Germany
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
| | - Annette Schürmann
- c Department of Experimental Diabetology , German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany and German Center for Diabetes Research (DZD) , Neuherberg , Germany
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
| | - Steffen Maak
- a Institute for Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology , Dummerstorf , Germany
| | - Wenke Jonas
- c Department of Experimental Diabetology , German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany and German Center for Diabetes Research (DZD) , Neuherberg , Germany
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
| | - Henrike Sell
- d German Center for Diabetes Research (DZD) , Neuherberg , Germany
- g Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center , Düsseldorf , Germany
| |
Collapse
|
25
|
Effects of type IV collagen on myogenic characteristics of IGF-I gene-engineered myoblasts. J Biosci Bioeng 2014; 119:596-603. [PMID: 25454061 DOI: 10.1016/j.jbiosc.2014.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
Skeletal muscle regeneration requires migration, proliferation and fusion of myoblasts to form multinucleated myotubes. In our previous study, we showed that insulin-like growth factor (IGF)-I gene delivery stimulates the proliferation and differentiation of mouse myoblast C2C12 cells and promotes the contractile force generated by tissue-engineered skeletal muscles. The aim of this study was to investigate the effects of the extracellular matrix on IGF-I gene-engineered C2C12 cells in vitro. Retroviral vectors for doxycycline (Dox)-inducible expression of the IGF-I gene were transduced into C2C12 cells. When cultured on a type IV collagen-coated surface, we observed significant increases in the migration speed and number of IGF-I gene-engineered C2C12 cells with Dox addition, designated as C2C12/IGF (+) cells. Co-culture of C2C12/IGF (+) cells and parental C2C12 cells, which had been cultured in differentiation medium for 3 days, greatly enhanced myotube formation. Moreover, type IV collagen supplementation promoted the fusion of C2C12/IGF (+) cells with differentiated C2C12 cells and increased the number of myotubes with striations. Myotubes formed by C2C12/IGF (+) cells cultured on type IV collagen showed a dynamic contractile activity in response to electrical pulse stimulation. These findings indicate that type IV collagen promotes skeletal muscle regeneration mediated by IGF-I-expressing myoblasts, which may have important clinical implications in the design of myoblast-based therapies.
Collapse
|
26
|
Abstract
Interest in Vitamin D has risen considerably recently with many athletes now advised to take daily vitamin D supplements. The reason for this interest is partly not only attributed to the resurgence of the Vitamin D-deficient disease rickets but also due to the discovery of a Vitamin D receptor in many tissues suggesting a more global role for Vitamin D than previously considered. Unlike the other vitamins that are obtained through the diet, Vitamin D is unique since endogenous synthesis following ultraviolet B (UVB) exposure is the predominant route of entry into systemic circulation. Moreover, Vitamin D could be better classed as a seco-steroid, given that its structure is similar to that of a steroid, and its production is derived from a cholesterol precursor (7-dehydrocholesteol) in the skin. The classification of Vitamin D status is currently subject to considerable debate with many authors opposing governing body recommendations. Regardless of the suggested optimal concentration, there is now growing evidence to suggest that many athletes are in fact Vitamin D deficient, especially in the winter months largely as a consequence of inadequate sun exposure, combined with poor dietary practices, although the consequences of such deficiencies are still unclear in athletic populations. Impaired muscle function and reduced regenerative capacity, impaired immune function, poor bone health and even impaired cardiovascular function have all been associated with low Vitamin D in athletes, however, to date, the majority of studies on Vitamin D have described associations and much more research is now needed examining causation.
Collapse
Affiliation(s)
- Daniel J Owens
- a Research Institute for Sport and Exercise Sciences , Liverpool John Moores University , Liverpool , UK
| | | | | |
Collapse
|
27
|
Lund DK, Mouly V, Cornelison DDW. MMP-14 is necessary but not sufficient for invasion of three-dimensional collagen by human muscle satellite cells. Am J Physiol Cell Physiol 2014; 307:C140-9. [PMID: 24898588 DOI: 10.1152/ajpcell.00032.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The twenty-five known matrix metalloproteases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteases (TIMPs), mediate cell invasion through the extracellular matrix (ECM). In a comparative three-dimensional assay, we analyzed human and mouse satellite cells' competence to invade an artificial ECM (collagen I). We identified a single MMP that 1) is expressed by human muscle satellite cells; 2) is induced at the mRNA/protein level by adhesion to collagen I; and 3) is necessary for invasion into a collagen I matrix. Interestingly, murine satellite cells neither express this MMP, nor invade the collagen matrix. However, exogenous human MMP-14 is not sufficient to induce invasion of a collagen matrix by murine cells, emphasizing species differences.
Collapse
Affiliation(s)
- Dane K Lund
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| | - Vincent Mouly
- Institut de Myologie, Université Pierre et Marie Curie, Paris, France
| | - D D W Cornelison
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
28
|
Lieber RL, Ward SR. Cellular mechanisms of tissue fibrosis. 4. Structural and functional consequences of skeletal muscle fibrosis. Am J Physiol Cell Physiol 2013; 305:C241-52. [PMID: 23761627 DOI: 10.1152/ajpcell.00173.2013] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Skeletal muscle fibrosis can be a devastating clinical problem that arises from many causes, including primary skeletal muscle tissue diseases, as seen in the muscular dystrophies, or it can be secondary to events that include trauma to muscle or brain injury. The cellular source of activated fibroblasts (myofibroblasts) may include resident fibroblasts, adult muscle stem cells, or inflammatory or perivascular cells, depending on the model studied. Even though it is likely that there is no single source for all myofibroblasts, a common mechanism for the production of fibrosis is via the transforming growth factor-β/phosphorylated Smad3 pathway. This pathway and its downstream targets thus provide loci for antifibrotic therapies, as do methods for blocking the transdifferentiation of progenitors into activated fibroblasts. A structural model for the extracellular collagen network of skeletal muscle is needed so that measurements of collagen content, morphology, and gene expression can be related to mechanical properties. Approaches used to study fibrosis in tissues, such as lung, kidney, and liver, need to be applied to studies of skeletal muscle to identify ways to prevent or even cure the devastating maladies of skeletal muscle.
Collapse
Affiliation(s)
- Richard L Lieber
- Department of Orthopaedic Surgery, University of California San Diego, San Diego, California 92093-0863, USA.
| | | |
Collapse
|