1
|
Bock-Pereda A, Cruz-Soca M, Gallardo FS, Córdova-Casanova A, Gutierréz-Rojas C, Faundez-Contreras J, Chun J, Casar JC, Brandan E. Involvement of lysophosphatidic acid-LPA 1-YAP signaling in healthy and pathological FAPs migration. Matrix Biol 2024; 133:103-115. [PMID: 39153517 DOI: 10.1016/j.matbio.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/09/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Skeletal muscle fibrosis is defined as the excessive accumulation of extracellular matrix (ECM) components and is a hallmark of muscular dystrophies. Fibro-adipogenic progenitors (FAPs) are the main source of ECM, and thus have been strongly implicated in fibrogenesis. In skeletal muscle fibrotic models, including muscular dystrophies, FAPs undergo dysregulations in terms of proliferation, differentiation, and apoptosis, however few studies have explored the impact of FAPs migration. Here, we studied fibroblast and FAPs migration and identified lysophosphatidic acid (LPA), a signaling lipid central to skeletal muscle fibrogenesis, as a significant migration inductor. We identified LPA receptor 1 (LPA1) mediated signaling as crucial for this effect through a mechanism dependent on the Hippo pathway, another pathway implicated in fibrosis across diverse tissues. This cross-talk favors the activation of the Yes-associated protein 1 (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), leading to increased expression of fibrosis-associated genes. This study reveals the role of YAP in LPA-mediated fibrotic responses as inhibition of YAP transcriptional coactivator activity hinders LPA-induced migration in fibroblasts and FAPs. Moreover, we found that FAPs derived from the mdx4cv mice, a murine model of Duchenne muscular dystrophy, display a heightened migratory phenotype due to enhanced LPA signaling compared to wild-type FAPs. Remarkably, we found that the inhibition of LPA1 or YAP transcriptional coactivator activity in mdx4cv FAPs reverts this phenotype. In summary, the identified LPA-LPA1-YAP pathway emerges as a critical driver of skeletal muscle FAPs migration and provides insights into potential novel targets to mitigate fibrosis in muscular dystrophies.
Collapse
Affiliation(s)
- Alexia Bock-Pereda
- Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Meilyn Cruz-Soca
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Felipe S Gallardo
- Facultad de Ciencias Biológicas, Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago 8330025, Chile; Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | | | - Cristian Gutierréz-Rojas
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile; Escuela de Kinesiología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340025, Chile; Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Jennifer Faundez-Contreras
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile; Facultad de Medicina y Ciencia, Fundación Ciencia y Vida, Universidad San Sebastián, Avenida del Valle Norte 725 Huechuraba, Santiago 7510602, Chile
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Juan Carlos Casar
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330077, Chile
| | - Enrique Brandan
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile; Facultad de Medicina y Ciencia, Fundación Ciencia y Vida, Universidad San Sebastián, Avenida del Valle Norte 725 Huechuraba, Santiago 7510602, Chile.
| |
Collapse
|
2
|
Liang CC, Huang WC, Shaw SW, Huang YH, Lee TH. Human amniotic fluid stem cells can alleviate detrusor dysfunction caused by bladder outlet obstruction in rats. Sci Rep 2022; 12:6679. [PMID: 35461349 PMCID: PMC9035144 DOI: 10.1038/s41598-022-10640-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/21/2022] [Indexed: 11/09/2022] Open
Abstract
The present study examined whether bladder detrusor dysfunction due to partial bladder outlet obstruction (pBOO) could be improved after the treatment of human amniotic fluid stem cells (hAFSCs). 72 female rats were grouped into sham operation, pBOO, and pBOO with hAFSCs treatment (pBOO + hAFSCs) for in vitro and in vivo studies. Bladder weight, bladder wall thickness, the ratio of collagen to smooth muscle and the levels of positive CD11b/c and HIS48 cells was significantly increased after pBOO but improved after hAFSCs treatment. Cystometries showed impaired bladder function after pBOO. Protein and mRNA levels of hypoxia inducible factor-1α, CCL2, interleukin-1β, transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), α-smooth muscle actin, collagen I and collagen III were increased at 2 and/or 6 weeks, but proteins and mRNA expressions of protein gene product 9.5 were decreased at 2 and 6 weeks after pBOO. These abnormalities were improved after hAFSCs treatment. The expressions of TGF-β1 and CTGF in cultured detrusor cells of pBOO rats were increased but were improved after hAFSCs treatment. The present results showed hAFSCs treatment could improve bladder detrusor dysfunction in pBOO rats, which may be related to the reduction of inflammatory and pro-fibrotic markers in detrusor muscle cells.
Collapse
Affiliation(s)
- Ching-Chung Liang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Chu Huang
- Division of Urogynecology, Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Nursing, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Steven W Shaw
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Obstetrics, Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK
| | - Yung-Hsin Huang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, No. 5, Fu-Hsing Street, Kweishan, 33333, Taoyuan, Taiwan.
| |
Collapse
|
3
|
Franzoso M, Dokshokova L, Vitiello L, Zaglia T, Mongillo M. Tuning the Consonance of Microscopic Neuro-Cardiac Interactions Allows the Heart Beats to Play Countless Genres. Front Physiol 2022; 13:841740. [PMID: 35273522 PMCID: PMC8902305 DOI: 10.3389/fphys.2022.841740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Different from skeletal muscle, the heart autonomously generates rhythmic contraction independently from neuronal inputs. However, speed and strength of the heartbeats are continuously modulated by environmental, physical or emotional inputs, delivered by cardiac innervating sympathetic neurons, which tune cardiomyocyte (CM) function, through activation of β-adrenoceptors (β-ARs). Given the centrality of such mechanism in heart regulation, β-AR signaling has been subject of intense research, which has reconciled the molecular details of the transduction pathway and the fine architecture of cAMP signaling in subcellular nanodomains, with its final effects on CM function. The importance of mechanisms keeping the elements of β-AR/cAMP signaling in good order emerges in pathology, when the loss of proper organization of the transduction pathway leads to detuned β-AR/cAMP signaling, with detrimental consequences on CM function. Despite the compelling advancements in decoding cardiac β-AR/cAMP signaling, most discoveries on the subject were obtained in isolated cells, somehow neglecting that complexity may encompass the means in which receptors are activated in the intact heart. Here, we outline a set of data indicating that, in the context of the whole myocardium, the heart orchestra (CMs) is directed by a closely interacting and continuously attentive conductor, represented by SNs. After a roundup of literature on CM cAMP regulation, we focus on the unexpected complexity and roles of cardiac sympathetic innervation, and present the recently discovered Neuro-Cardiac Junction, as the election site of "SN-CM" interaction. We further discuss how neuro-cardiac communication is based on the combination of extra- and intra-cellular signaling micro/nano-domains, implicating neuronal neurotransmitter exocytosis, β-ARs and elements of cAMP homeostasis in CMs, and speculate on how their dysregulation may reflect on dysfunctional neurogenic control of the heart in pathology.
Collapse
Affiliation(s)
- Mauro Franzoso
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lolita Dokshokova
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
4
|
Treviño-Villarreal JH, Reynolds JS, Langston PK, Thompson A, Mitchell JR, Franco RA. Down-Regulation of a Profibrotic Transforming Growth Factor-β1/Cellular Communication Network Factor 2/Matrix Metalloprotease 9 Axis by Triamcinolone Improves Idiopathic Subglottic Stenosis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1412-1430. [PMID: 34111429 DOI: 10.1016/j.ajpath.2021.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
Idiopathic subglottic stenosis (iSGS) is a progressive fibrotic disease characterized by life-threatening airway narrowing. Although the molecular underpinnings are unknown, previous reports showing that subglottic serial intralesional steroid injections (SILSIs) improve clinical outcomes suggest a steroid-sensitive pathway in iSGS. Herein, a prospective study was conducted to determine the changes in profibrotic markers during SILSI to identify steroid-sensitive profibrotic drivers. Seven newly diagnosed patients with iSGS were recruited for SILSI. Subglottic biopsies before and after SILSI treatments were evaluated for histologic and molecular markers by confocal microscopy and RT-qPCR. At baseline, iSGS subglottises contained abundant vimentin-positive/α-smooth muscle actin-negative fibroblasts, intermingled with a matrix of fibronectin and types I and VI collagen. Transforming growth factor (TGF)-β1 was up-regulated primarily in glandular epithelium. Cellular communication network factor 2 (CCN2) was mainly up-regulated in stromal fibroblasts surrounding TGF-β1-positive glandular structures. SILSI improved iSGS by reducing fibroblast infiltration and increasing matrix remodeling. Mechanistically, SILSI counteracted the effects of TGF-β1 by inducing matrix metalloprotease 9 (MMP9) expression while repressing CCN2 expression, without affecting TGFβ1 levels. Treatment of primary iSGS-derived fibroblasts with TGF-β1 recapitulated aspects of the disease in vivo, demonstrating that the induction in CCN2 and repression of MMP9 are caused by changes in histone acetylation induced by TGF-β1. Triamcinolone counteracted the coregulation of these genes by impairing SMAD2/3 binding to promoter regions, and not through histone acetylation. In conclusion, this study shows that SILSI counteracts a dysregulated TGF-β1/CCN2/MMP9 axis involved in iSGS development.
Collapse
Affiliation(s)
| | - Justin S Reynolds
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - P Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts
| | - Andrew Thompson
- Dana Farber Cancer Institute/Harvard Medical School Rodent Histopathology Core Facility, Harvard Medical School, Boston, Massachusetts
| | - James R Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ramon A Franco
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Division of Laryngology, Department of Otolaryngology, Mass Eye and Ear and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
5
|
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: 7.3] [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
|
6
|
Petrosino JM, Leask A, Accornero F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle. FASEB J 2019; 33:2047-2057. [PMID: 30216109 PMCID: PMC6338641 DOI: 10.1096/fj.201800622rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023]
Abstract
In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Andrew Leask
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Gonzalez D, Brandan E. CTGF/CCN2 from Skeletal Muscle to Nervous System: Impact on Neurodegenerative Diseases. Mol Neurobiol 2019; 56:5911-5916. [PMID: 30689195 DOI: 10.1007/s12035-019-1490-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022]
Abstract
Connective tissue growth factor (CTGF/CCN2) is a matricellular protein that belongs to the CCN family of proteins. Since its discovery, it has been linked to cellular processes such as cell proliferation, differentiation, adhesion, migration, and synthesis of extracellular matrix (ECM) components, among others. The pro-fibrotic role of CTGF/CCN2 has been well-studied in several pathologies characterized by the development of fibrosis. Reduction of CTGF/CCN2 levels in mdx mice, a murine model for Duchenne muscular dystrophy (DMD), decreases fibrosis and improves skeletal muscle phenotype and function. Recently, it has been shown that skeletal muscle of symptomatic hSOD1G93A mice, a model for Amyotrophic lateral sclerosis (ALS), shows up-regulation of CTGF/CCN2 accompanied by excessive deposition ECM molecules. Elevated levels of CTGF/CCN2 in spinal cord from ALS patients have been previously reported. However, there is no evidence regarding the role of CTGF/CCN2 in neurodegenerative diseases such as ALS, in which alterations in skeletal muscle seem to be the consequence of early pathological denervation. In this regard, the emerging evidence shows that CTGF/CCN2 also exerts non-fibrotic roles in the central nervous system (CNS), specifically impairing oligodendrocyte maturation and regeneration, and inhibiting axon myelination. Despite these striking observations, there is no evidence showing the role of CTGF/CCN2 in peripheral nerves. Therefore, even though more studies are needed to elucidate its precise role, CTGF/CCN2 is starting to emerge as a novel therapeutic target for the treatment of neurodegenerative diseases where demyelination and axonal degeneration occurs.
Collapse
Affiliation(s)
- David Gonzalez
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Libertador Bernardo O'Higgins 340, 8331150, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Libertador Bernardo O'Higgins 340, 8331150, Santiago, Chile.
| |
Collapse
|
8
|
Adedeji TG, Olapade-Olaopa EO. Dietary macronutrient content affects inflammatory and fibrotic factors in normal and obstructed bladders. Life Sci 2018; 210:192-200. [DOI: 10.1016/j.lfs.2018.08.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/23/2018] [Accepted: 08/30/2018] [Indexed: 10/28/2022]
|
9
|
Hara M, Yokota K, Saito T, Kobayakawa K, Kijima K, Yoshizaki S, Okazaki K, Yoshida S, Matsumoto Y, Harimaya K, Nakashima Y, Okada S. Periostin Promotes Fibroblast Migration and Inhibits Muscle Repair After Skeletal Muscle Injury. J Bone Joint Surg Am 2018; 100:e108. [PMID: 30106825 DOI: 10.2106/jbjs.17.01230] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Skeletal muscle injury (SMI) can cause physical disability due to insufficient recovery of the muscle. The development of muscle fibrosis after SMI has been widely regarded as a principal cause of this failure to recover. Periostin (Postn) exacerbates tissue fibrosis in various organs. We investigated whether Postn is involved in the pathophysiology after SMI. METHODS Partial laceration injuries of the gastrocnemius were created in wild-type (WT) and Postn knockout (Postn) mice. We examined the expression of the Postn gene before and after SMI. Regeneration and fibrosis of skeletal muscle were evaluated by histological analyses, and recovery of muscle strength was measured by physiological testing. Immunohistochemistry was used to examine the number and proliferative potential of infiltrating fibroblasts in injured muscle. A trans-well migration assay was used to assess the migration capability of fibroblasts. Control immunoglobulin G (IgG) or Postn-neutralizing antibody (Postn-nAb) was injected into injured muscle at 7 and 14 days after injury (dpi). We evaluated the effects of Postn-nAb on muscle repair after SMI. RESULTS The expression of Postn was dramatically upregulated after SMI. Compared with WT mice, Postn mice had improved muscle recovery and attenuated fibrosis as well as a significantly reduced number of infiltrating fibroblasts. The proliferative potential of these fibroblasts in WT and Postn mice was comparable at 14 dpi; however, the migration capability of fibroblasts was significantly enhanced in the presence of Postn (mean, 258%; 95% confidence interval, 183% to 334%). Moreover, the administration of Postn-nAb inhibited fibroblast infiltration and promoted muscle repair after SMI. CONCLUSIONS Postn exacerbates fibrotic scar formation through the promotion of fibroblast migration into injured muscle after SMI. Treatment with Postn-nAb is effective for attenuating fibrosis and improving muscle recovery after SMI. CLINICAL RELEVANCE Our findings may provide a potential therapeutic strategy to enhance muscle repair and functional recovery after SMI.
Collapse
Affiliation(s)
- Masamitsu Hara
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Shigeo Yoshida
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsumi Harimaya
- Department of Orthopaedic Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yasuharu Nakashima
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Riquelme-Guzmán C, Contreras O, Brandan E. Expression of CTGF/CCN2 in response to LPA is stimulated by fibrotic extracellular matrix via the integrin/FAK axis. Am J Physiol Cell Physiol 2017; 314:C415-C427. [PMID: 29351412 DOI: 10.1152/ajpcell.00013.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fibrosis is a common feature of several chronic diseases and is characterized by exacerbated accumulation of ECM. An understanding of the cellular and molecular mechanisms involved in the development of this condition is crucial for designing efficient treatments for those pathologies. Connective tissue growth factor (CTGF/CCN2) is a pleiotropic protein with strong profibrotic activity. In this report, we present experimental evidence showing that ECM stimulates the synthesis of CTGF in response to lysophosphatidic acid (LPA).The integrin/focal adhesion kinase (FAK) signaling pathway mediates this effect, since CTGF expression is abolished by the use of the Arg-Gly-Asp-Ser peptide and also by an inhibitor of FAK autophosphorylation at tyrosine 397. Cilengitide, a specific inhibitor of αv integrins, inhibits the expression of CTGF mediated by LPA or transforming growth factor β1. We show that ECM obtained from decellularized myofibroblast cultures or derived from activated fibroblasts from muscles of the Duchenne muscular dystrophy mouse model ( mdx) induces the expression of CTGF. This effect is dependent on FAK phosphorylation in response to its activation by integrin. We also found that the fibrotic ECM inhibits skeletal muscle differentiation. This novel regulatory mechanism of CTGF expression could be acting as a positive profibrotic feedback between the ECM and CTGF, revealing a novel concept in the control of fibrosis under chronic damage.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- Centro de Envejecimiento y Regeneración, CARE Chile UC, and Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Osvaldo Contreras
- Centro de Envejecimiento y Regeneración, CARE Chile UC, and Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, and Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
11
|
Nio Y, Tanaka M, Hirozane Y, Muraki Y, Okawara M, Hazama M, Matsuo T. Phosphodiesterase 4 inhibitor and phosphodiesterase 5 inhibitor combination therapy has antifibrotic and anti‐inflammatory effects in mdx mice with Duchenne muscular dystrophy. FASEB J 2017; 31:5307-5320. [DOI: 10.1096/fj.201700249r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Yasunori Nio
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masayuki Tanaka
- Inflammation Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yoshihiko Hirozane
- Biomolecular Research LaboratoriesPharmaceutical Research DivisionTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yo Muraki
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Mitsugi Okawara
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masatoshi Hazama
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Takanori Matsuo
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| |
Collapse
|
12
|
Song Y, Yao S, Liu Y, Long L, Yang H, Li Q, Liang J, Li X, Lu Y, Zhu H, Zhang N. Expression levels of TGF-β1 and CTGF are associated with the severity of Duchenne muscular dystrophy. Exp Ther Med 2017; 13:1209-1214. [PMID: 28413459 PMCID: PMC5377242 DOI: 10.3892/etm.2017.4105] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/26/2016] [Indexed: 12/20/2022] Open
Abstract
The present study aimed to analyze the association of transforming growth factor-β1 (TGF-β1) and connective tissue growth factor (CTGF) expression levels in skeletal muscle with the clinical manifestation of Duchenne muscular dystrophy (DMD). A total of 18 cases of DMD, which were confirmed by routine pathological diagnosis were recruited into the present study, along with 8 subjects who suffered from acute trauma but did not present any neuromuscular diseases and were enrolled as the healthy controls. Immunohistochemical staining was used to detect the expression levels of CTGF and TGF-β1 in muscle biopsy specimens. Furthermore, Spearman rank correlation analysis was conducted among the expression levels of CTGF and TGF-β1, age, clinical severity and pathological severity in DMD patients. The immunohistochemical staining results revealed that the expression levels of CTGF and TGF-β1 were significantly increased in the DMD group compared with those in the control group (P<0.05). These levels were not found to be significantly correlated with the onset age (P>0.05), but there was a significant correlation with the degree of pathology and clinical severity (P<0.05). In conclusion, an upregulated expression of CTGF and TGF-β1 was revealed in the skeletal muscle of DMD patients, which were in positive correlation with the degree of pathology and clinical severity. These two factors may be involved in the pathophysiology of fibrosis in DMD.
Collapse
Affiliation(s)
- Yanmin Song
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China.,Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuai Yao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China.,Department of Rehabilitation Medicine, Mental Health Centre of Wuxi, Wuxi, Jiangsu 214151, P.R. China
| | - Yunhai Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qiuxiang Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jinghui Liang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xinxin Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yuling Lu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Haoran Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ning Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
13
|
Goetsch KP, Niesler CU. The extracellular matrix regulates the effect of decorin and transforming growth factor beta-2 (TGF-β2) on myoblast migration. Biochem Biophys Res Commun 2016; 479:351-357. [PMID: 27644884 DOI: 10.1016/j.bbrc.2016.09.079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/15/2016] [Indexed: 12/31/2022]
Abstract
Muscular injuries that destroy the basal lamina result in poor functional recovery of skeletal muscle. This is due, in part, to the deposition of structural fibrotic proteins such as fibronectin and collagen by fibroblasts and other cells. Transforming growth factor-β (TGF-β) promotes fibrosis, whereas the proteoglycan decorin is known to act as an anti-fibrotic agent, in part via the binding and neutralization of TGF-β. We have previously established that decorin can alter the migratory response of skeletal muscle myoblasts to the extracellular matrix (ECM) factor collagen, but not fibronectin. We have also shown that TGF-β reduces myoblast migration. In the current study we demonstrate that decorin can dramatically alter the inhibitory role of TGF-β on human myoblast migration and go on to shown that the extracellular matrix can significantly modify this effect. Decorin and TGF-β2 in combination were observed to significantly increase the rate of human myoblast migration, despite the inhibitory effect of TGF-β2 on its own. Furthermore, in the presence of fibronectin, TGF-β2 and decorin no longer acted synergistically to promote migration; while in the presence of collagen I, TGF-β2 failed to inhibit migration. These studies show, for the first time, that decorin can alter the bioactivity of TGF-β2 on human myoblast migration and emphasize the crucial regulatory role of the extracellular matrix in determining the response of skeletal muscle myoblasts to migratory cues.
Collapse
Affiliation(s)
- K P Goetsch
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - C U Niesler
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa.
| |
Collapse
|
14
|
Contreras O, Rebolledo DL, Oyarzún JE, Olguín HC, Brandan E. Connective tissue cells expressing fibro/adipogenic progenitor markers increase under chronic damage: relevance in fibroblast-myofibroblast differentiation and skeletal muscle fibrosis. Cell Tissue Res 2016; 364:647-660. [DOI: 10.1007/s00441-015-2343-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
|
15
|
Gutiérrez J, Droppelmann CA, Contreras O, Takahashi C, Brandan E. RECK-Mediated β1-Integrin Regulation by TGF-β1 Is Critical for Wound Contraction in Mice. PLoS One 2015; 10:e0135005. [PMID: 26247610 PMCID: PMC4527692 DOI: 10.1371/journal.pone.0135005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/16/2015] [Indexed: 11/18/2022] Open
Abstract
Fibroblasts are critical for wound contraction; a pivotal step in wound healing. They produce and modify the extracellular matrix (ECM) required for the proper tissue remodeling. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK) is a key regulator of ECM homeostasis and turnover. However, its role in wound contraction is presently unknown. Here we describe that Transforming growth factor type β1 (TGF-β1), one of the main pro-fibrotic wound-healing promoting factors, decreases RECK expression in fibroblasts through the Smad and JNK dependent pathways. This TGF-β1 dependent downregulation of RECK occurs with the concomitant increase of β1-integrin, which is required for fibroblasts adhesion and wound contraction through the activation of focal adhesion kinase (FAK). Loss and gain RECK expression experiments performed in different types of fibroblasts indicate that RECK downregulation mediates TGF-β1 dependent β1-integrin expression. Also, reduced levels of RECK potentiate TGF-β1 effects over fibroblasts FAK-dependent contraction, without affecting its cognate signaling. The above results were confirmed on fibroblasts derived from the Reck+/- mice compared to wild type-derived fibroblasts. We observed that Reck+/- mice heal dermal wounds more efficiently than wild type mice. Our results reveal a critical role for RECK in skin wound contraction as a key mediator in the axis: TGF-β1—RECK- β1-integrin.
Collapse
Affiliation(s)
- Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail: (JG); (EB)
| | - Cristian A. Droppelmann
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Molecular Medicine Group, Robarts Research Institute, Western University, London, Ontario, Canada
| | - Osvaldo Contreras
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Chiaki Takahashi
- Oncology and Molecular Biology, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Enrique Brandan
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail: (JG); (EB)
| |
Collapse
|
16
|
Gutpell KM, Hoffman LM. VEGF induces stress fiber formation in fibroblasts isolated from dystrophic muscle. J Cell Commun Signal 2015. [PMID: 26219981 DOI: 10.1007/s12079-015-0300-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Treatment with vascular endothelial growth factor (VEGF) to reduce ischemia and enhance both endogenous muscle repair and regenerative cell therapy in Duchenne muscular dystrophy (DMD) has been widely proposed in recent years. However, the interaction between angiogenesis and fibrosis, a hallmark feature of DMD, remains unclear. To date, it has not been determined whether VEGF exerts a pro-fibrotic effect on DMD-derived fibroblasts, which may contribute to further disease progression. Thus, the purpose of this study was to investigate the effect of exogenous VEGF on fibroblast cultures established from a murine model of DMD. Primary fibroblast cultures were established from gastrocnemius and diaphragm muscles of 10 week-old mdx/utrn+/- mice. Quantitative polymerase chain reaction (qPCR) was employed to assess changes in transcript expression of alpha-smooth muscle actin (Acta2), type-1 collagen (Col1a1), connective tissue growth factor (Ctgf/ccn2) and fibronectin (Fn1). Immunofluorescence and Western blot analysis was further employed to visualize changes in protein expression of alpha-smooth muscle actin (α-SMA), CTGF/CCN2 and fibronectin. mRNA levels of Col1a1, Ctgf/ccn2, and FN did not increase following treatment with VEGF in fibroblasts derived from either diaphragm or gastrocnemius muscles. Acta2 expression increased significantly in diaphragm-derived fibroblasts following treatment with VEGF. Morphological assessment revealed increased stress fiber formation in VEGF-treated fibroblasts compared to the untreated control fibroblasts. The findings from this study suggest that further investigation into the effect of VEGF on fibroblast function is required prior to the utilization of the growth factor as a treatment for DMD.
Collapse
Affiliation(s)
- Kelly M Gutpell
- Lawson Health Research Institute, London, Ontario, Canada. .,Department of Anatomy and Cell Biology, Western University, London, Ontario, Canada. .,Department of Medical Biophysics, Western University, London, Ontario, Canada.
| | - Lisa M Hoffman
- Lawson Health Research Institute, London, Ontario, Canada. .,Department of Anatomy and Cell Biology, Western University, London, Ontario, Canada. .,Department of Medical Biophysics, Western University, London, Ontario, Canada. .,Imaging Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario, Canada, N6A 4V2.
| |
Collapse
|
17
|
Cofre C, Acuña MJ, Contreras O, Morales MG, Riquelme C, Cabello-Verrugio C, Brandan E. Transforming growth factor type-β inhibits Mas receptor expression in fibroblasts but not in myoblasts or differentiated myotubes; Relevance to fibrosis associated to muscular dystrophies. Biofactors 2015; 41:111-20. [PMID: 25809912 DOI: 10.1002/biof.1208] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/26/2015] [Indexed: 12/28/2022]
Abstract
Duchenne muscular dystrophy is a genetic disorder characterized by myofiber degeneration, muscle weakness, and increased fibrosis. Transforming growth factor type-β (TGF-β), a central mediator of fibrosis, is upregulated in fibrotic diseases. Angiotensin-(1-7) [Ang-(1-7)] is a peptide with actions that oppose those of angiotensin-II (Ang II). Ang-(1-7) effects are mediated by the Mas receptor. Treatment with Ang-(1-7) produce positive effects in the mdx mouse, normalizing skeletal muscle architecture, decreasing local fibrosis, and fibroblasts, and improving muscle function. Mdx mice deficient for the Mas receptor showed the opposite effects. To identify the cell type(s) responsible for Mas receptor expression, and to characterize whether profibrotic effectors had any effect on its expression, we determined the effect of profibrotic agents on Mas expression. TGF-β, but not connective tissue growth factor or Ang-II, reduced the expression of Mas receptor in fibroblasts isolated from skeletal muscle cells and fibroblasts from two established cell lines. In contrast, no effects were observed in myoblasts and differentiated myotubes. This inhibition was mediated by the Smad-dependent (canonical) and the PI3K and MEK1/2 (noncanonical) TGF-β signaling pathways. When both canonical and noncanonical inhibitors of the TGF-β-dependent pathways were added together, the inhibitory effect of TGF-β on Mas expression was lost. The decrease in Mas receptor induced by TGF-β in fibroblasts reduced the Ang-(1-7) mediated stimulation of phosphorylation of AKT pathway proteins. These results suggest that reduction of Mas receptor in fibroblasts, by TGF-β, could increase the fibrotic phenotype observed in dystrophic skeletal muscle decreasing the beneficial effect of Ang-(1-7).
Collapse
MESH Headings
- Angiotensin I/pharmacology
- Angiotensin II/pharmacology
- Animals
- Cell Line
- Disease Models, Animal
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Fibrosis
- Gene Expression Regulation
- MAP Kinase Kinase 1/genetics
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/genetics
- MAP Kinase Kinase 2/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts/drug effects
- Myoblasts/metabolism
- Myoblasts/pathology
- Organ Specificity
- Peptide Fragments/pharmacology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation/drug effects
- Primary Cell Culture
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Catalina Cofre
- Center for Aging and Regeneration, CARE Chile-UC and Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
18
|
Steinberger M, Föller M, Vogelgesang S, Krautwald M, Landsberger M, Winkler CK, Kasch J, Füchtbauer EM, Kuhl D, Voelkl J, Lang F, Brinkmeier H. Lack of the serum- and glucocorticoid-inducible kinase SGK1 improves muscle force characteristics and attenuates fibrosis in dystrophic mdx mouse muscle. Pflugers Arch 2014; 467:1965-74. [DOI: 10.1007/s00424-014-1645-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 10/10/2014] [Accepted: 10/31/2014] [Indexed: 02/06/2023]
|
19
|
Song YS, Li H, Balcos MC, Yun HY, Baek KJ, Kwon NS, Choi HR, Park KC, Kim DS. Fucoidan promotes the reconstruction of skin equivalents. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:327-31. [PMID: 25177165 PMCID: PMC4146635 DOI: 10.4196/kjpp.2014.18.4.327] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/20/2014] [Accepted: 06/27/2014] [Indexed: 11/15/2022]
Abstract
In this study we investigated the effects of fucoidan on the proliferation of fibroblasts and the reconstruction of a skin equivalent (SE). Fucoidan significantly stimulated the proliferation of CCD-25Sk human fibroblasts and Western blot analysis demonstrated that fucoidan markedly increased the expression of cyclin D1 and decreased the expression of p27. Fucoidan was used to reconstruct SE. Immunohistochemical staining showed that the addition of fucoidan to dermal equivalents increased expression of proliferating cell nuclear antigen (PCNA) and p63. In addition, expression of α6-integrin was significantly increased by fucoidan, whereas expression of β1-integrin, type 1 collagen, elastin, fibronectin did not markedly change. These results suggest that fucoidan has positive effects on epidermal reconstruction and will therefore be beneficial in the reconstruction of SE.
Collapse
Affiliation(s)
- Yu Seok Song
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Hailan Li
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Marie Carmel Balcos
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Hye-Young Yun
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Kwang Jin Baek
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Nyoun Soo Kwon
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Hye-Ryung Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Kyoung-Chan Park
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Dong-Seok Kim
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| |
Collapse
|
20
|
Cabrera D, Gutiérrez J, Cabello-Verrugio C, Morales MG, Mezzano S, Fadic R, Casar JC, Hancke JL, Brandan E. Andrographolide attenuates skeletal muscle dystrophy in mdx mice and increases efficiency of cell therapy by reducing fibrosis. Skelet Muscle 2014; 4:6. [PMID: 24655808 PMCID: PMC4021597 DOI: 10.1186/2044-5040-4-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/26/2014] [Indexed: 02/06/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is characterized by the absence of the cytoskeletal protein dystrophin, muscle wasting, increased transforming growth factor type beta (TGF-β) signaling, and fibrosis. At the present time, the only clinically validated treatments for DMD are glucocorticoids. These drugs prolong muscle strength and ambulation of patients for a short term only and have severe adverse effects. Andrographolide, a bicyclic diterpenoid lactone, has traditionally been used for the treatment of colds, fever, laryngitis, and other infections with no or minimal side effects. We determined whether andrographolide treatment of mdx mice, an animal model for DMD, affects muscle damage, physiology, fibrosis, and efficiency of cell therapy. Methods mdx mice were treated with andrographolide for three months and skeletal muscle histology, creatine kinase activity, and permeability of muscle fibers were evaluated. Fibrosis and TGF-β signaling were evaluated by indirect immunofluorescence and Western blot analyses. Muscle strength was determined in isolated skeletal muscles and by a running test. Efficiency of cell therapy was determined by grafting isolated skeletal muscle satellite cells onto the tibialis anterior of mdx mice. Results mdx mice treated with andrographolide exhibited less severe muscular dystrophy than untreated dystrophic mice. They performed better in an exercise endurance test and had improved muscle strength in isolated muscles, reduced skeletal muscle impairment, diminished fibrosis and a significant reduction in TGF-β signaling. Moreover, andrographolide treatment of mdx mice improved grafting efficiency upon intramuscular injection of dystrophin-positive satellite cells. Conclusions These results suggest that andrographolide could be used to improve quality of life in individuals with DMD.
Collapse
Affiliation(s)
- Daniel Cabrera
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Laboratorio de Diferenciación Celular y Patología, Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile ; Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Jaime Gutiérrez
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Laboratorio de Diferenciación Celular y Patología, Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Maria Gabriela Morales
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Laboratorio de Diferenciación Celular y Patología, Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Sergio Mezzano
- División de Nefrología, Escuela de Medicina, Universidad Austral, Valdivia, Chile
| | - Ricardo Fadic
- Departamento de Neurología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Casar
- Departamento de Neurología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan L Hancke
- Instituto de Farmacología, Universidad Austral de Chile, Valdivia, Chile
| | - Enrique Brandan
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Laboratorio de Diferenciación Celular y Patología, Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| |
Collapse
|
21
|
Wnt signaling in skeletal muscle dynamics: myogenesis, neuromuscular synapse and fibrosis. Mol Neurobiol 2013; 49:574-89. [PMID: 24014138 DOI: 10.1007/s12035-013-8540-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/15/2013] [Indexed: 12/21/2022]
Abstract
The signaling pathways activated by Wnt ligands are related to a wide range of critical cell functions, such as cell division, migration, and synaptogenesis. Here, we summarize compelling evidence on the role of Wnt signaling on several features of skeletal muscle physiology. We briefly review the role of Wnt pathways on the formation of muscle fibers during prenatal and postnatal myogenesis, highlighting its role on the activation of stem cells of the adult muscles. We also discuss how Wnt signaling regulates the precise formation of neuromuscular synapses, by modulating the differentiation of presynaptic and postsynaptic components, particularly regarding the clustering of acetylcholine receptors on the muscle membrane. In addition, based on previous evidence showing that Wnt pathways are linked to several diseases, such as Alzheimer's and cancer, we address recent studies indicating that Wnt signaling plays a key role in skeletal muscle fibrosis, a disease characterized by an increase in the extracellular matrix components leading to failure in muscle regeneration, tissue disorganization and loss of muscle activity. In this context, we also discuss the possible cross-talk between the Wnt/β-catenin pathway with two other critical profibrotic pathways, transforming growth factor β and connective tissue growth factor, which are potent stimulators of the accumulation of connective tissue, an effect characteristic of the fibrotic condition. As it has emerged in other pathological conditions, we suggests that muscle fibrosis may be a consequence of alterations of Wnt signaling activity.
Collapse
|
22
|
Morales MG, Gutierrez J, Cabello-Verrugio C, Cabrera D, Lipson KE, Goldschmeding R, Brandan E. Reducing CTGF/CCN2 slows down mdx muscle dystrophy and improves cell therapy. Hum Mol Genet 2013; 22:4938-51. [PMID: 23904456 DOI: 10.1093/hmg/ddt352] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD) and the mdx mouse model, the absence of the cytoskeletal protein dystrophin causes defective anchoring of myofibres to the basal lamina. The resultant myofibre degeneration and necrosis lead to a progressive loss of muscle mass, increased fibrosis and ultimately fatal weakness. Connective tissue growth factor (CTGF/CCN-2) is critically involved in several chronic fibro-degenerative diseases. In DMD, the role of CTGF might extend well beyond replacement fibrosis secondary to loss of muscle fibres, since its overexpression in skeletal muscle could by itself induce a dystrophic phenotype. Using two independent approaches, we here show that mdx mice with reduced CTGF availability do indeed have less severe muscular dystrophy. Mdx mice with hemizygous CTGF deletion (mdx-Ctgf+/-), and mdx mice treated with a neutralizing anti-CTGF monoclonal antibody (FG-3019), performed better in an exercise endurance test, had better muscle strength in isolated muscles and reduced skeletal muscle impairment, apoptotic damage and fibrosis. Transforming growth factor type-β (TGF-β), pERK1/2 and p38 signalling remained unaffected during CTGF suppression. Moreover, both mdx-Ctgf+/- and FG-3019 treated mdx mice had improved grafting upon intramuscular injection of dystrophin-positive satellite cells. These findings reveal the potential of targeting CTGF to reduce disease progression and to improve cell therapy in DMD.
Collapse
Affiliation(s)
- Maria Gabriela Morales
- Laboratorio de Diferenciación Celular y Patología, Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
23
|
Painemal P, Acuña MJ, Riquelme C, Brandan E, Cabello-Verrugio C. Transforming growth factor type beta 1 increases the expression of angiotensin II receptor type 2 by a SMAD- and p38 MAPK-dependent mechanism in skeletal muscle. Biofactors 2013; 39:467-75. [PMID: 23460581 DOI: 10.1002/biof.1087] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 12/13/2012] [Indexed: 01/13/2023]
Abstract
Excessive deposition of extracellular matrix (ECM) proteins, a condition known as fibrosis, is a hallmark of Duchenne muscular dystrophy. Among the factors that trigger muscle fibrosis are transforming growth factor beta (TGF-β) and angiotensin II (Ang-II). Ang-II belongs to the renin-angiotensin system, and its biological effects are exerted by Ang-II receptors type 1 and type 2 (AT-1 and AT-2, respectively). This study aims to determine the effect of TGF-β1 on the expression of AT-1 and AT-2 receptor in skeletal muscle. C2 C12 myoblasts exposed to TGF-β1 showed a dose-dependent increase in AT-2 expression but with no effect on AT-1 levels. Injection of TGF-β1 in the skeletal muscle of mice increased the levels of AT-2 and ECM protein but unchanged AT-1 levels. We also detected higher expression levels of AT-2 receptor in dystrophic skeletal muscle of mdx mice than in normal mice. The induction of AT-2 was mediated by the canonical TGF-β pathway because under the inhibitory conditions of the kinase activity of TGFβ receptor I or the knockdown of Smad2/3 levels, TGF-β-induced AT-2 receptor increase was strongly inhibited. Furthermore, we demonstrated that p38MAPK activity in response to TGF-β is also required for AT-2 increase as evaluated by a p38MAPK inhibitor. Our results show that the levels of AT-2 but not AT-1 receptor are modulated by the pro-fibrotic factor TGF-β1 in myoblasts and mouse skeletal muscle. This finding suggests that AT-2 might be involved in the physiopathology of fibrosis in dystrophic skeletal muscle.
Collapse
MESH Headings
- Animals
- Cell Line
- Gene Expression
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Renin-Angiotensin System
- Signal Transduction
- Smad Proteins/metabolism
- Transforming Growth Factor beta1/pharmacology
- Transforming Growth Factor beta1/physiology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Paula Painemal
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
24
|
Brandan E, Gutierrez J. Role of proteoglycans in the regulation of the skeletal muscle fibrotic response. FEBS J 2013; 280:4109-17. [PMID: 23560928 DOI: 10.1111/febs.12278] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/30/2013] [Accepted: 04/03/2013] [Indexed: 02/06/2023]
Abstract
Myogenesis consists of a highly organized and regulated sequence of cellular processes aimed at forming or repairing muscle tissue. Several processes occur during myogenesis, including cell proliferation, migration, and differentiation. Cytokines, proteinases, cell adhesion molecules and growth factors are involved, either activating or inhibiting these events, and are modulated by a group of molecules called proteoglycans (PGs), which play critical roles in skeletal muscle physiology. Particularly interesting are some of the factors responsible for the fibrotic response associated with skeletal muscular dystrophies. Transforming growth factor-β and connective tissue growth factor have gained great attention as factors participating in the fibrotic response in skeletal muscle. This review is focused on the advances achieved in understanding the roles of proteoglycans as modulators of profibrotic growth factors in fibrosis associated with diseases such as skeletal muscle dystrophies.
Collapse
Affiliation(s)
- Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile.
| | | |
Collapse
|
25
|
Mosqueira M, Zeiger U, Förderer M, Brinkmeier H, Fink RHA. Cardiac and respiratory dysfunction in Duchenne muscular dystrophy and the role of second messengers. Med Res Rev 2013; 33:1174-213. [PMID: 23633235 DOI: 10.1002/med.21279] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) affects young boys and is characterized by the absence of dystrophin, a large cytoskeletal protein present in skeletal and cardiac muscle cells and neurons. The heart and diaphragm become necrotic in DMD patients and animal models of DMD, resulting in cardiorespiratory failure as the leading cause of death. The major consequences of the absence of dystrophin are high levels of intracellular Ca(2+) and the unbalanced production of NO that can finally trigger protein degradation and cell death. Cytoplasmic increase in Ca(2+) concentration directly and indirectly triggers different processes such as necrosis, fibrosis, and activation of macrophages. The absence of the neuronal isoform of nitric oxide synthase (nNOS) and the overproduction of NO by the inducible isoform (iNOS) further increase the intracellular Ca(2+) via a hypernitrosylation of the ryanodine receptor. NO overproduction, which further induces the expression of iNOS but decreases the expression of the endothelial isoform (eNOS), deregulates the muscle tissue blood flow creating an ischemic situation. The high levels of Ca(2+) in dystrophic muscles and the ischemic state of the muscle tissue would culminate in a positive feedback loop. While efforts continue toward optimizing cardiac and respiratory care of DMD patients, both Ca(2+) and NO in cardiac and respiratory muscle pathways have been shown to be important to the etiology of the disease. Understanding the mechanisms behind the fine regulation of Ca(2+) -NO may be important for a noninterventional and noninvasive supportive approach to treat DMD patients, improving the quality of life and natural history of DMD patients.
Collapse
Affiliation(s)
- Matias Mosqueira
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, INF326, Heidelberg University, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
26
|
The involvement of collagen triple helix repeat containing 1 in muscular dystrophies. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:905-16. [PMID: 23274062 DOI: 10.1016/j.ajpath.2012.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 10/15/2012] [Accepted: 11/01/2012] [Indexed: 11/20/2022]
Abstract
Fibrosis is the main complication of muscular dystrophies. We identified collagen triple helix repeat containing 1 (Cthrc1) in skeletal and cardiac muscles of mice, representing Duchenne and congenital muscle dystrophies (DMD and CMD, respectively), and dysferlinopathy. In all of the mice, Cthrc1 was associated with high collagen type I levels; no Cthrc1 or collagen was observed in muscles of control mice. High levels of Cthrc1 were also observed in biopsy specimens from patients with DMD, in whom they were reversibly correlated with that of β-dystroglycan, whereas collagen type I levels were elevated in all patients with DMD. At the muscle sites where collagen and Cthrc1 were adjacent, collagen fibers appeared smaller, suggesting involvement of Cthrc1 in collagen turnover. Halofuginone, an inhibitor of Smad3 phosphorylation downstream of the transforming growth factor-β signaling, reduced Cthrc1 levels in skeletal and cardiac muscles of mice, representing DMD, CMD, and dysferlinopathy. The myofibroblasts infiltrating the dystrophic muscles of the murine models of DMD, CMD, and dysferlinopathy were the source of Cthrc1. Transforming growth factor-β did not affect Cthrc1 levels in the mdx fibroblasts but decreased them in the control fibroblasts, in association with increased migration of mdx fibroblasts and dystrophic muscle invasion by myofibroblasts. To our knowledge, this is the first demonstration of Cthrc1 as a marker of the severity of the disease progression in the dystrophic muscles, and as a possible target for therapy.
Collapse
|
27
|
Morales MG, Vazquez Y, Acuña MJ, Rivera JC, Simon F, Salas JD, Alvarez Ruf J, Brandan E, Cabello-Verrugio C. Angiotensin II-induced pro-fibrotic effects require p38MAPK activity and transforming growth factor beta 1 expression in skeletal muscle cells. Int J Biochem Cell Biol 2012; 44:1993-2002. [PMID: 22964022 DOI: 10.1016/j.biocel.2012.07.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
Fibrotic disorders are typically characterised by excessive connective tissue and extracellular matrix (ECM) deposition that preclude the normal healing of different tissues. Several skeletal muscle dystrophies are characterised by extensive fibrosis. Among the factors involved in skeletal muscle fibrosis is angiotensin II (Ang-II), a key protein of the renin-angiotensin system (RAS). We previously demonstrated that myoblasts responded to Ang-II by increasing the ECM protein levels mediated by AT-1 receptors, implicating an Ang-II-induced reactive oxygen species (ROS) by a NAD(P)H oxidase-dependent mechanism. In this paper, we show that in myoblasts, Ang-II induced the increase of transforming growth factor beta 1 (TGF-β1) and connective tissue growth factor (CTGF) expression through its AT-1 receptor. This effect is dependent of the NAD(P)H oxidase (NOX)-induced ROS, as indicated by a decrease of the expression of both pro-fibrotic factors when the ROS production was inhibited via the NOX inhibitor apocynin. The increase in pro-fibrotic factors levels was paralleled by enhanced p38MAPK and ERK1/2 phosphorylation in response to Ang-II. However, only the p38MAPK activity was critical for the Ang-II-induced fibrotic effects, as indicated by the decrease in the Ang-II-induced TGF-β1 and CTGF expression and fibronectin levels by SB-203580, an inhibitor of the p38MAPK, but not by U0126, an inhibitor of ERK1/2 phosphorylation. Furthermore, we showed that the Ang-II-dependent p38MAPK activation, but not the ERK1/2 phosphorylation, was necessary for the NOX-derived ROS. In addition, we demonstrated that TGF-β1 expression was required for the Ang-II-induced pro-fibrotic effects evaluated by using SB-431542, an inhibitor of TGF-βRI kinase activity, and by knocking down TGF-β1 levels by shRNA technique. These results strongly suggest that the fibrotic response to Ang-II is mediated by the AT-1 receptor and requires the p38MAPK phosphorylation, NOX-induced ROS, and TGF-β1 expression increase mediated by Ang-II in skeletal muscle cells.
Collapse
Affiliation(s)
- María Gabriela Morales
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bo Li Z, Zhang J, Wagner KR. Inhibition of myostatin reverses muscle fibrosis through apoptosis. J Cell Sci 2012; 125:3957-65. [PMID: 22685331 DOI: 10.1242/jcs.090365] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Skeletal muscle fibrosis is a defining feature of the muscular dystrophies in which contractile myofibers are replaced by fibroblasts, adipocytes and extracellular matrix. This maladaptive response of muscle to repetitive injury is progressive, self-perpetuating and thus far, has been considered irreversible. We have previously shown that myostatin, a known endogenous modulator of muscle growth, stimulates normal muscle fibroblasts to proliferate. Here, we demonstrate that myostatin also regulates the proliferation of dystrophic muscle fibroblasts, and increases resistance of fibroblasts to apoptosis through Smad and MAPK signaling. Inhibition of myostatin signaling pathways with a soluble activin IIB receptor (ActRIIB.Fc) reduces resistance of muscle fibroblasts to apoptosis in vitro. Systemic administration of ActRIIB.Fc in senescent mdx mice, a model of muscular dystrophy, significantly increases the number of muscle fibroblasts undergoing apoptosis. This leads to the reversal of pre-existing muscle fibrosis as determined by histological, biochemical and radiographical criteria. These results demonstrate that skeletal muscle fibrosis can be pharmacologically reversed through induction of fibroblast apoptosis.
Collapse
Affiliation(s)
- Zhao Bo Li
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, 707 North Broadway, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
29
|
Verhaart IEC, Heemskerk H, Karnaoukh TG, Kolfschoten IGM, Vroon A, van Ommen GJB, van Deutekom JCT, Aartsma-Rus A. Prednisolone treatment does not interfere with 2'-O-methyl phosphorothioate antisense-mediated exon skipping in Duchenne muscular dystrophy. Hum Gene Ther 2012; 23:262-73. [PMID: 22017442 DOI: 10.1089/hum.2011.127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In Duchenne muscular dystrophy (DMD), dystrophin deficiency leading to progressive muscular degeneration is caused by frame-shifting mutations in the DMD gene. Antisense oligonucleotides (AONs) aim to restore the reading frame by skipping of a specific exon(s), thereby allowing the production of a shorter, but semifunctional protein, as is found in the mostly more mildly affected patients with Becker muscular dystrophy. AONs are currently being investigated in phase 3 placebo-controlled clinical trials. Most of the participating patients are treated symptomatically with corticosteroids (mainly predniso[lo]ne) to stabilize the muscle fibers, which might affect the uptake and/or efficiency of AONs. Therefore the effect of prednisolone on 2'-O-methyl phosphorothioate AON efficacy in patient-derived cultured muscle cells and the mdx mouse model (after local and systemic AON treatment) was assessed in this study. Both in vitro and in vivo skip efficiency and biomarker expression were comparable between saline- and prednisolone-cotreated cells and mice. After systemic exon 23-specific AON (23AON) treatment for 8 weeks, dystrophin was detectable in all treated mice. Western blot analyses indicated slightly higher dystrophin levels in prednisolone-treated mice, which might be explained by better muscle condition and consequently more target dystrophin pre-mRNA. In addition, fibrotic and regeneration biomarkers were normalized to some extent in prednisolone- and/or 23AON-treated mice. Overall these results show that the use of prednisone forms no barrier to participation in clinical trials with AONs.
Collapse
Affiliation(s)
- Ingrid E C Verhaart
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gillies AR, Lieber RL. Structure and function of the skeletal muscle extracellular matrix. Muscle Nerve 2012; 44:318-31. [PMID: 21949456 DOI: 10.1002/mus.22094] [Citation(s) in RCA: 655] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The skeletal muscle extracellular matrix (ECM) plays an important role in muscle fiber force transmission, maintenance, and repair. In both injured and diseased states, ECM adapts dramatically, a property that has clinical manifestations and alters muscle function. Here we review the structure, composition, and mechanical properties of skeletal muscle ECM; describe the cells that contribute to the maintenance of the ECM; and, finally, overview changes that occur with pathology. New scanning electron micrographs of ECM structure are also presented with hypotheses about ECM structure–function relationships. Detailed structure–function relationships of the ECM have yet to be defined and, as a result, we propose areas for future study.
Collapse
Affiliation(s)
- Allison R Gillies
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0863, USA
| | | |
Collapse
|
31
|
Cabello-Verrugio C, Acuña MJ, Morales MG, Becerra A, Simon F, Brandan E. Fibrotic response induced by angiotensin-II requires NAD(P)H oxidase-induced reactive oxygen species (ROS) in skeletal muscle cells. Biochem Biophys Res Commun 2011; 410:665-70. [DOI: 10.1016/j.bbrc.2011.06.051] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/06/2011] [Indexed: 12/20/2022]
|
32
|
Metcalfe PD, Wang J, Jiao H, Huang Y, Hori K, Moore RB, Tredget EE. Bladder outlet obstruction: progression from inflammation to fibrosis. BJU Int 2010; 106:1686-1694. [DOI: 10.1111/j.1464-410x.2010.09445.x] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Study Type – Aetiology (case control)
Level of Evidence 3bOBJECTIVETo investigate the progression of urodynamic changes, as well as histological and biochemical outcomes over a prolonged period of partial bladder outlet obstruction (pBOO) in an animal model with physiologically relevant pBOO.MATERIALS AND METHODSHealthy, adult, female Fischer rats underwent surgical creation of a pBOO for either 2, 4, 8, or 13 weeks and were compared with sham‐operated rats. Urodynamic measurements were used to compare bladder volumes and pressure. Tissue was grossly analysed with light microscopy and bladder weights and thicknesses were compared. Reverse transcription‐polymerase chain reaction for collagen, transforming growth factor β (TGF‐β), connective tissue growth factor (CTGF), hypoxia inducible factor 1α (HIF‐1α), and platelet‐derived growth factor (PDGF‐A) was performed on all samples, as well as immunohistochemistry (IHC) for α‐smooth muscle actin (α‐SMA). Finally, mass spectrometry was used to quantify the collagen content of the bladders as a measure of fibrosis.RESULTSAfter induction of pBOO, all rats remained healthy. Initial urodynamics showed an increase in capacity while maintaining normal pressures, but then deteriorated into small capacity, high‐pressure bladders. Haematoxylin and eosin (H&E) staining showed an initial inflammatory response, and this was confirmed with significantly increased mRNA levels of TGF‐β, CTGF, HIF‐1α, and PDGF. The progression to smooth muscle hypertrophy was evident on H&E and confirmed with increased bladder mass and thickness. IHC for α‐SMA showed a progressive increase associated with the elevated bladder pressures. Masson’s trichrome and mass spectrometry showed a progressive increase in collagen to 13 weeks.CONCLUSIONWith this model, we have effectively replicated the clinical scenario, with significant pathophysiological changes occurring insidiously in otherwise healthy rats. We believe that our observed changes represent distinct phases of bladder decompensation; with an initial inflammatory response to the stress of the pBOO, smooth muscle hypertrophy to overcome the increased urethral resistance, and eventual decompensation to fibrosis. The time course of the inflammatory markers implies the need for early intervention to prevent this cascade. Novel strategies targeting these observed physiological responses could lead to improved preventative strategies, with respect to biochemical pathways and the time course of their initiation.
Collapse
|
33
|
Cabello-Verrugio C, Córdova G, Vial C, Zúñiga LM, Brandan E. Connective tissue growth factor induction by lysophosphatidic acid requires transactivation of transforming growth factor type β receptors and the JNK pathway. Cell Signal 2010; 23:449-57. [PMID: 20965247 DOI: 10.1016/j.cellsig.2010.10.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/06/2010] [Accepted: 10/11/2010] [Indexed: 02/06/2023]
Abstract
Transforming growth factor β (TGF-β) is a very strong pro-fibrotic factor which mediates its action, at least in part, through the expression of connective tissue growth factor (CTGF/CCN2). Along with these cytokines, the involvement of phospholipids in wound healing and the development of fibrosis has been revealed. Among them, lysophosphatidic acid (LPA) is a novel, potent regulator of wound healing and fibrosis that has diverse effects on many types of cells. We decided to evaluate the effect of LPA together with TGF-β on CTGF expression. We found that myoblasts treated with LPA and TGF-β1 produced an additive effect on CTGF expression. In the absence of TGF-β, the induction of CTGF expression by LPA was abolished by a dominant negative form of the TGF-β receptor type II (TGF-βRII) and by the use of SB 431542, a specific inhibitor of the serine/threonine kinase activity of TGF-βRI, suggesting that CTGF induction is dependent on LPA and requires active TGF-βRs. Moreover, we show that LPA requires Smad-2/3 proteins for the induction of CTGF expression, but not their phosphorylation or their nuclear translocation. The requirement of TGF-βRI for LPA mediated-effects is differential, since treatment of myoblasts with LPA in the presence of SB 431542 abolished the induction of stress fibers but not the induction of proliferation. Finally, we demonstrated that CTGF induction in response to LPA requires the activation of JNK, but not ERK, signaling pathways. The JNK requirement is independent of TGF-βRI-mediated activity. These novel results for the mechanism of action of LPA and TGF-β are important for understanding the role of pro-fibrotic growth factors and phospholipids involved in wound healing and related diseases.
Collapse
Affiliation(s)
- Claudio Cabello-Verrugio
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
34
|
Nevo Y, Halevy O, Genin O, Moshe I, Turgeman T, Harel M, Biton E, Reif S, Pines M. Fibrosis inhibition and muscle histopathology improvement in laminin-alpha2-deficient mice. Muscle Nerve 2010; 42:218-29. [PMID: 20589893 DOI: 10.1002/mus.21706] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In muscular dystrophies (MD) the loss of muscle and its ability to function are associated with fibrosis. We evaluated the efficacy of halofuginone in reducing fibrosis in the dy(2J)/dy(2J) mouse model of congenital MD. Mice were injected intraperitoneally with 5 microg of halofuginone 3 times a week for 5 or 15 weeks, starting at the age of 3 weeks. Halofuginone caused a reduction in collagen synthesis in hindlimb muscles. This was associated with reductions in the degenerated area, in cell proliferation, in the number of myofibers with central nuclei, with increased myofiber diameter, and with enhanced motor coordination and balance. Halofuginone caused a reduction in infiltrating fibroblasts that were located close to centrally nucleated myofibers. Our results suggest that halofuginone reduced the deleterious effects of fibrosis, thus improving muscle integrity. Halofuginone meets the criteria for a novel antifibrotic therapy for MD patients.
Collapse
Affiliation(s)
- Yoram Nevo
- Neuropediatric Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Improved muscle strength and mobility in the dy(2J)/dy(2J) mouse with merosin deficient congenital muscular dystrophy treated with Glatiramer acetate. Neuromuscul Disord 2010; 20:267-72. [PMID: 20304648 DOI: 10.1016/j.nmd.2010.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 01/29/2010] [Accepted: 02/01/2010] [Indexed: 01/01/2023]
Abstract
The therapeutic effect of Glatiramer acetate, an immune modulating agent, was evaluated in the dy(2J)/dy(2J) mouse with merosin deficient congenital muscular dystrophy, which is a milder variant of the dy/dy mouse. The treated mice showed significant improvement in hind limb muscle strength measured by electronic grip strength meter and in motor performance quantified by video detection software. Glatiramer acetate treatment was associated with significantly increased expression of regeneration transcription factors MyoD and myogenin, and attenuation of the fibrosis markers vimentin and fibronectin. No effective treatment is currently available in congenital muscular dystrophy and Glatiramer acetate may present a new potential treatment for this disorder.
Collapse
|
36
|
Osses N, Casar JC, Brandan E. Inhibition of extracellular matrix assembly induces the expression of osteogenic markers in skeletal muscle cells by a BMP-2 independent mechanism. BMC Cell Biol 2009; 10:73. [PMID: 19804635 PMCID: PMC2762961 DOI: 10.1186/1471-2121-10-73] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 10/05/2009] [Indexed: 11/10/2022] Open
Abstract
Background The conversion of one cell type into another has been suggested to be, at the molecular level, the consequence of change(s) in the expression level of key developmental genes. Myoblasts have the ability to differentiate either to skeletal muscle or osteogenic lineage depending of external stimuli. Extracellular matrix (ECM) has been shown to be essential for skeletal muscle differentiation, through its direct interaction with myoblasts' cell receptors. We attempt to address if ECM also plays a role in the osteogenic differentiation of skeletal muscle cells. Results Inhibition of proteoglycan sulfation by sodium chlorate in myoblast cultures strongly affects ECM synthesis and deposition and induces the expression of the osteogenic lineage markers alkaline phosphatase (ALP) and osteocalcin in mononuclear cells. Induction of ALP by sodium chlorate does not affect the expression of specific muscle determination transcription factors, such as MyoD and Myf-5, in the same cells. The osteogenic transcription factor Cbfa-1 expression is also unaffected. Induction of ALP is not inhibited by a soluble form of BMP receptor IA. This suggests that the deviation of the myogenic pathway of C2C12 myoblasts into the osteogenic lineage by inhibitors of proteoglycan sulfation is BMP-2 independent. The increase of osteogenic markers expression can be totally prevented by an exogenous ECM. Interestingly, a similar BMP-2-independent ALP activity induction can be observed in myoblasts cultured on an ECM previously synthesized by BMP-2 treated myoblasts. Under in vivo conditions of increased ECM turn-over and deposition, as in the mdx dystrophic muscle and during skeletal muscle regeneration, an induction and relocalization of ALP is observed in a subpopulation of skeletal muscle fibers, whereas in normal skeletal muscle, ALP expression is restricted to blood vessels and some endomysial mononuclear cells. Conclusion These results suggest that signals arising from the ECM induce the expression of osteogenic markers in muscle cells by a mechanism independent of BMP-2 and without affecting the expression of key muscle or osteogenic determination genes. An induction and relocalization of ALP is also observed in mdx and regenerating skeletal muscles, in vivo conditions of increased muscle ECM deposition or turnover.
Collapse
Affiliation(s)
- Nelson Osses
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | |
Collapse
|
37
|
Immunoneutralization of TGFbeta1 Improves Skeletal Muscle Regeneration: Effects on Myoblast Differentiation and Glycosaminoglycan Content. Int J Cell Biol 2009; 2009:659372. [PMID: 20111627 PMCID: PMC2809363 DOI: 10.1155/2009/659372] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Revised: 02/16/2009] [Accepted: 02/23/2009] [Indexed: 01/26/2023] Open
Abstract
When injured by crushing, the repair of the slow-twitch soleus rat muscle, unlike the fast-twitch EDL, is associated with fibrosis. As TGFβ1, whose activity can be controlled by glycosaminoglycans (GAG), plays a major role in fibrosis, we hypothesized that levels of TGFβ1 and GAG contents could account for this differential quality of regeneration. Here we show that the regeneration of the soleus was accompanied by elevated and more sustained TGFβ1 level than in the EDL. Neutralization of TGFβ1 effects by antibodies to TGFβ1 or its receptor TGFβ-R1 improved muscle repair, especially of the soleus muscle, increased in vitro growth of myoblasts, and accelerated their differentiation. These processes were accompanied by alterations of GAG contents. These results indicate that the control of TGFβ1 activity is important to improve regeneration of injured muscle and accelerate myoblast differentiation, in part through changes in GAG composition of muscle cell environment.
Collapse
|
38
|
Droppelmann CA, Gutiérrez J, Vial C, Brandan E. Matrix metalloproteinase-2-deficient fibroblasts exhibit an alteration in the fibrotic response to connective tissue growth factor/CCN2 because of an increase in the levels of endogenous fibronectin. J Biol Chem 2009; 284:13551-13561. [PMID: 19276073 DOI: 10.1074/jbc.m807352200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Matrix metalloproteinase-2 (MMP-2) is an important extracellular matrix remodeling enzyme, and it has been involved in different fibrotic disorders. The connective tissue growth factor (CTGF/CCN2), which is increased in these pathologies, induces the production of extracellular matrix proteins. To understand the fibrotic process observed in diverse pathologies, we analyzed the fibroblast response to CTGF when MMP-2 activity is inhibited. CTGF increased fibronectin (FN) amount, MMP-2 mRNA expression, and gelatinase activity in 3T3 cells. When MMP-2 activity was inhibited either by the metalloproteinase inhibitor GM-6001 or in MMP-2-deficient fibroblasts, an increase in the basal amount of FN together with a decrease of its levels in response to CTGF was observed. This paradoxical effect could be explained by the fact that the excess of FN could block the access to other ligands, such as CTGF, to integrins. This effect was emulated in fibroblasts by adding exogenous FN or RGDS peptides or using anti-integrin alpha(V) subunit-blocking antibodies. Additionally, in MMP-2-deficient cells CTGF did not induce the formation of stress fibers, focal adhesion sites, and ERK phosphorylation. Anti-integrin alpha(V) subunit-blocking antibodies inhibited ERK phosphorylation in control cells. Finally, in MMP-2-deficient cells, FN mRNA expression was not affected by CTGF, but degradation of (125)I-FN was increased. These results suggest that expression, regulation, and activity of MMP-2 can play an important role in the initial steps of fibrosis and shows that FN levels can regulate the cellular response to CTGF.
Collapse
Affiliation(s)
- Cristian A Droppelmann
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Millenium Institute for Fundamental and Applied Biology, Pontificia Universidad Católica de Chile, Casilla 114-D, Santiago, Chile
| | - Jaime Gutiérrez
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Millenium Institute for Fundamental and Applied Biology, Pontificia Universidad Católica de Chile, Casilla 114-D, Santiago, Chile
| | - Cecilia Vial
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Millenium Institute for Fundamental and Applied Biology, Pontificia Universidad Católica de Chile, Casilla 114-D, Santiago, Chile
| | - Enrique Brandan
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Millenium Institute for Fundamental and Applied Biology, Pontificia Universidad Católica de Chile, Casilla 114-D, Santiago, Chile.
| |
Collapse
|
39
|
TGF-β's delay skeletal muscle progenitor cell differentiation in an isoform-independent manner. Exp Cell Res 2009; 315:373-84. [DOI: 10.1016/j.yexcr.2008.10.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 10/20/2008] [Accepted: 10/28/2008] [Indexed: 11/18/2022]
|
40
|
Novel regulatory mechanisms for the proteoglycans decorin and biglycan during muscle formation and muscular dystrophy. Matrix Biol 2008; 27:700-8. [DOI: 10.1016/j.matbio.2008.07.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 06/24/2008] [Accepted: 07/01/2008] [Indexed: 01/11/2023]
|