1
|
Tapia G, Fuenzalida S, Rivera C, Apablaza P, Silva M, Jaimovich E, Juretić N. L-Arginine Activates the Neuregulin-1/ErbB Receptor Signaling Pathway and Increases Utrophin mRNA Levels in C2C12 Cells. Biochem Res Int 2025; 2025:2171745. [PMID: 40224962 PMCID: PMC11991828 DOI: 10.1155/bri/2171745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 04/15/2025] Open
Abstract
L-arginine induces the expression of utrophin in skeletal muscle cells, so it has been proposed as a pharmacological treatment to attenuate the symptoms of Duchenne muscular dystrophy (DMD). On the other hand, it has been described that one of the pathways that participates in the expression of utrophin in muscle is the Neuregulin-1 (NRG-1)/ErbB receptors pathway. Several studies have postulated that disintegrin and metalloprotease-17 (ADAM17) causes the proteolytic processing of NRG of transmembrane, allowing the release of NRG to the medium, which when joining its ErbB receptor activates the signaling pathway that triggers utrophin transcription. The aim of this study was to evaluate the effect of L-arginine in the activation of NRG-1/ErbB pathway and utrophin mRNA levels in C2C12 cells, and the participation of ADAM17 in this process. Our results indicate that L-arginine induces phosphorylation of ErbB2 and increases utrophin mRNA levels in C2C12 myotubes, with a maximum increase of 2-fold at 4 h post-stimulation. This effect is not observed when the myotubes are stimulated in the presence of GM6001 (general metalloprotease inhibitor) or PD-158780 (specific inhibitor of ErbB receptor phosphorylation). Experiments performed by flow cytometry suggest that L-arginine stimulates ADAM17 activation in our study model. Furthermore, immunofluorescence analysis supports our findings that L-arginine stimulates ADAM17 increase in treated myotubes. However, our results using pharmacological inhibitors suggest that ADAM17 does not participate in utrophin expression in C2C12 cells treated with L-arginine. The results obtained help to clarify the mechanism of action of L-arginine in the expression of utrophin in muscle cells, which will contribute to the design of new therapeutic strategies in pathologies such as DMD.
Collapse
Affiliation(s)
- Gladys Tapia
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Sebastián Fuenzalida
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Constanza Rivera
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Pía Apablaza
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Mónica Silva
- Centro de Estudios de Ejercicio, Metabolismo y Cáncer, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Enrique Jaimovich
- Centro de Estudios de Ejercicio, Metabolismo y Cáncer, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| | - Nevenka Juretić
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
| |
Collapse
|
2
|
Anderson MJM, Hayward AN, Smiley AT, Shi K, Pawlak MR, Aird EJ, Grant E, Greenberg L, Aihara H, Evans RL, Ulens C, Gordon WR. Molecular basis of proteolytic cleavage regulation by the extracellular matrix receptor dystroglycan. Structure 2024; 32:1984-1996.e5. [PMID: 39305901 PMCID: PMC11560575 DOI: 10.1016/j.str.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Abstract
The dystrophin-glycoprotein-complex (DGC), anchored by the transmembrane protein dystroglycan, functions to mechanically link the extracellular matrix and actin cytoskeleton. Breaking this connection is associated with diseases such as muscular dystrophy, yet cleavage of dystroglycan by matrix-metalloproteinases (MMPs) remains an understudied mechanism to disrupt the DGC. We determined the crystal structure of the membrane-adjacent domain (amino acids 491-722) of E. coli expressed human dystroglycan to understand MMP cleavage regulation. The structural model includes tandem immunoglobulin-like (IGL) and sperm/enterokinase/agrin-like (SEAL) domains, which support proteolysis in diverse receptors to facilitate mechanotransduction, membrane protection, and viral entry. The structure reveals a C-terminal extension that buries the MMP site by packing into a hydrophobic pocket, a unique mechanism of MMP cleavage regulation. We further demonstrate structure-guided and disease-associated mutations disrupt proteolytic regulation using a cell-surface proteolysis assay. Thus disrupted proteolysis is a potentially relevant mechanism for "breaking" the DGC link to contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Michael J M Anderson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Adam T Smiley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Matthew R Pawlak
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Eric J Aird
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; Currently at Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Eva Grant
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Lauren Greenberg
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Robert L Evans
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Christopher Ulens
- Department of Cellular and Molecular Medicine, Karolinksa University Leuven, 3000 Leuven, Belgium
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Robertson R, Li S, Filippelli RL, Chang NC. Muscle stem cell dysfunction in rhabdomyosarcoma and muscular dystrophy. Curr Top Dev Biol 2024; 158:83-121. [PMID: 38670717 DOI: 10.1016/bs.ctdb.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Muscle stem cells (MuSCs) are crucial to the repair and homeostasis of mature skeletal muscle. MuSC dysfunction and dysregulation of the myogenic program can contribute to the development of pathology ranging from cancers like rhabdomyosarcoma (RMS) or muscle degenerative diseases such as Duchenne muscular dystrophy (DMD). Both diseases exhibit dysregulation at nearly all steps of myogenesis. For instance, MuSC self-renewal processes are altered. In RMS, this leads to the creation of tumor propagating cells. In DMD, impaired asymmetric stem cell division creates a bias towards producing self-renewing stem cells instead of committing to differentiation. Hyperproliferation of these cells contribute to tumorigenesis in RMS and symmetric expansion of the self-renewing MuSC population in DMD. Both diseases also exhibit a repression of factors involved in terminal differentiation, halting RMS cells in the proliferative stage and thus driving tumor growth. Conversely, the MuSCs in DMD exhibit impaired differentiation and fuse prematurely, affecting myonuclei maturation and the integrity of the dystrophic muscle fiber. Finally, both disease states cause alterations to the MuSC niche. Various elements of the niche such as inflammatory and migratory signaling that impact MuSC behavior are dysregulated. Here we show how these seemingly distantly related diseases indeed have similarities in MuSC dysfunction, underlying the importance of considering MuSCs when studying the pathophysiology of muscle diseases.
Collapse
Affiliation(s)
- Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada.
| |
Collapse
|
4
|
Norris AM, Appu AB, Johnson CD, Zhou LY, McKellar DW, Renault MA, Hammers D, Cosgrove BD, Kopinke D. Hedgehog signaling via its ligand DHH acts as cell fate determinant during skeletal muscle regeneration. Nat Commun 2023; 14:3766. [PMID: 37355632 PMCID: PMC10290686 DOI: 10.1038/s41467-023-39506-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Successful muscle regeneration relies on the interplay of multiple cell populations. However, the signals required for this coordinated intercellular crosstalk remain largely unknown. Here, we describe how the Hedgehog (Hh) signaling pathway controls the fate of fibro/adipogenic progenitors (FAPs), the cellular origin of intramuscular fat (IMAT) and fibrotic scar tissue. Using conditional mutagenesis and pharmacological Hh modulators in vivo and in vitro, we identify DHH as the key ligand that acts as a potent adipogenic brake by preventing the adipogenic differentiation of FAPs. Hh signaling also impacts muscle regeneration, albeit indirectly through induction of myogenic factors in FAPs. Our results also indicate that ectopic and sustained Hh activation forces FAPs to adopt a fibrogenic fate resulting in widespread fibrosis. In this work, we reveal crucial post-developmental functions of Hh signaling in balancing tissue regeneration and fatty fibrosis. Moreover, they provide the exciting possibility that mis-regulation of the Hh pathway with age and disease could be a major driver of pathological IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Ambili Bai Appu
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Connor D Johnson
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Lylybell Y Zhou
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marie-Ange Renault
- Biology of Cardiovascular Diseases, INSERM, University of Bordeaux, Pessac, France
| | - David Hammers
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Bobadilla Muñoz M, Orbe J, Abizanda G, Machado FJD, Vilas A, Ullate-Agote A, Extramiana L, Baraibar Churio A, Aranguren XL, Cantero G, Sáinz Amillo N, Rodríguez JA, Ramos García L, Romero Riojas JP, Vallejo-Illarramendi A, Paradas C, López de Munain A, Páramo JA, Prósper F, Pérez-Ruiz A. Loss of the matrix metalloproteinase-10 causes premature features of aging in satellite cells. Front Cell Dev Biol 2023; 11:1128534. [PMID: 37228645 PMCID: PMC10203875 DOI: 10.3389/fcell.2023.1128534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Aged muscles accumulate satellite cells with a striking decline response to damage. Although intrinsic defects in satellite cells themselves are the major contributors to aging-associated stem cell dysfunction, increasing evidence suggests that changes in the muscle-stem cell local microenvironment also contribute to aging. Here, we demonstrate that loss of the matrix metalloproteinase-10 (MMP-10) in young mice alters the composition of the muscle extracellular matrix (ECM), and specifically disrupts the extracellular matrix of the satellite cell niche. This situation causes premature features of aging in the satellite cells, contributing to their functional decline and a predisposition to enter senescence under proliferative pressure. Similarly, reduction of MMP-10 levels in young satellite cells from wild type animals induces a senescence response, while addition of the protease delays this program. Significantly, the effect of MMP-10 on satellite cell aging can be extended to another context of muscle wasting, muscular dystrophy. Systemic treatment of mdx dystrophic mice with MMP-10 prevents the muscle deterioration phenotype and reduces cellular damage in the satellite cells, which are normally under replicative pressure. Most importantly, MMP-10 conserves its protective effect in the satellite cell-derived myoblasts isolated from a Duchenne muscular dystrophy patient by decreasing the accumulation of damaged DNA. Hence, MMP-10 provides a previously unrecognized therapeutic opportunity to delay satellite cell aging and overcome satellite cell dysfunction in dystrophic muscles.
Collapse
Affiliation(s)
- Miriam Bobadilla Muñoz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Josune Orbe
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS)-Ictus, Instituto de Salud Carlos III, Madrid, Spain
| | - Gloria Abizanda
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Florencio J. D. Machado
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Amaia Vilas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Asier Ullate-Agote
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leire Extramiana
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Arantxa Baraibar Churio
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier L. Aranguren
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gloria Cantero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Neira Sáinz Amillo
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Centre for Nutrition Research, Universidad de Navarra, Pamplona, Spain
| | - José Antonio Rodríguez
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Ramos García
- Radiology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Radiology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - Juan Pablo Romero Riojas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Carmen Paradas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Adolfo López de Munain
- CIBERNED-Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
- Neurology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - José Antonio Páramo
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Ana Pérez-Ruiz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
6
|
Boccanegra B, Cappellari O, Mantuano P, Trisciuzzi D, Mele A, Tulimiero L, De Bellis M, Cirmi S, Sanarica F, Cerchiara AG, Conte E, Meanti R, Rizzi L, Bresciani E, Denoyelle S, Fehrentz JA, Cruciani G, Nicolotti O, Liantonio A, Torsello A, De Luca A. Growth hormone secretagogues modulate inflammation and fibrosis in mdx mouse model of Duchenne muscular dystrophy. Front Immunol 2023; 14:1119888. [PMID: 37122711 PMCID: PMC10130389 DOI: 10.3389/fimmu.2023.1119888] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Growth hormone secretagogues (GHSs) exert multiple actions, being able to activate GHS-receptor 1a, control inflammation and metabolism, to enhance GH/insulin-like growth factor-1 (IGF-1)-mediated myogenesis, and to inhibit angiotensin-converting enzyme. These mechanisms are of interest for potentially targeting multiple steps of pathogenic cascade in Duchenne muscular dystrophy (DMD). Methods Here, we aimed to provide preclinical evidence for potential benefits of GHSs in DMD, via a multidisciplinary in vivo and ex vivo comparison in mdx mice, of two ad hoc synthesized compounds (EP80317 and JMV2894), with a wide but different profile. 4-week-old mdx mice were treated for 8 weeks with EP80317 or JMV2894 (320 µg/kg/d, s.c.). Results In vivo, both GHSs increased mice forelimb force (recovery score, RS towards WT: 20% for EP80317 and 32% for JMV2894 at week 8). In parallel, GHSs also reduced diaphragm (DIA) and gastrocnemius (GC) ultrasound echodensity, a fibrosis-related parameter (RS: ranging between 26% and 75%). Ex vivo, both drugs ameliorated DIA isometric force and calcium-related indices (e.g., RS: 40% for tetanic force). Histological analysis highlighted a relevant reduction of fibrosis in GC and DIA muscles of treated mice, paralleled by a decrease in gene expression of TGF-β1 and Col1a1. Also, decreased levels of pro-inflammatory genes (IL-6, CD68), accompanied by an increment in Sirt-1, PGC-1α and MEF2c expression, were observed in response to treatments, suggesting an overall improvement of myofiber metabolism. No detectable transcript levels of GHS receptor-1a, nor an increase of circulating IGF-1 were found, suggesting the presence of a novel receptor-independent mechanism in skeletal muscle. Preliminary docking studies revealed a potential binding capability of JMV2894 on metalloproteases involved in extracellular matrix remodeling and cytokine production, such as ADAMTS-5 and MMP-9, overactivated in DMD. Discussion Our results support the interest of GHSs as modulators of pathology progression in mdx mice, disclosing a direct anti-fibrotic action that may prove beneficial to contrast pathological remodeling.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ornella Cappellari
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Mantuano
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Daniela Trisciuzzi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Lisamaura Tulimiero
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Michela De Bellis
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Santa Cirmi
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Francesca Sanarica
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Elena Conte
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Orazio Nicolotti
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milan-BICOCCA, Milan, Italy
| | - Annamaria De Luca
- Department of Pharmacy – Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
7
|
Shibabaw T, Teferi B, Ayelign B. The role of Th-17 cells and IL-17 in the metastatic spread of breast cancer: As a means of prognosis and therapeutic target. Front Immunol 2023; 14:1094823. [PMID: 36993955 PMCID: PMC10040566 DOI: 10.3389/fimmu.2023.1094823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Metastatic breast cancer is one of the most common and well-known causes of death for women worldwide. The inflammatory tumor cell and other cancer hallmarks dictate the metastatic form and dissemination of breast cancer. Taking these into account, from various components of the tumor microenvironment, a pro-inflammatory infiltrative cell known as Th-17 plays an immense role in breast cancer proliferation, invasiveness, and metastasis. It has been demonstrated that IL-17, a pleiotropic pro-inflammatory cytokine generated by Th-17, is upregulated in a metastatic form of breast cancer. Recent research updates stated that chronic inflammation and mediators like cytokines and chemokines are causative hallmarks in many human cancers, including breast cancer. Therefore, IL-17 and its multiple downward signaling molecules are the centers of research attention to develop potent treatment options for cancer. They provide information on the role of IL-17-activated MAPK, which results in tumor cell proliferation and metastasis via NF-kB-mediated expression of MMP signaling. Overall, this review article emphasizes IL-17A and its intermediate signaling molecules, such as ERK1/2, NF-kB, MMPs, and VEGF, as potential molecular targets for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
- Research School of Biology, College of Science, Australian National University, Canberra, ACT, Australia
- *Correspondence: Birhanu Ayelign,
| |
Collapse
|
8
|
Goncharuk O, Savosko S, Tykhomyrov A, Guzyk M, Medvediev V, Tsymbaliuk V, Chaikovsky Y. Matrix Metalloproteinase-9 is Involved in the Fibrotic Process in Denervated Muscles after Sciatic Nerve Trauma and Recovery. J Neurol Surg A Cent Eur Neurosurg 2023; 84:116-122. [PMID: 34496416 DOI: 10.1055/s-0041-1731750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Fibrosis of the injured muscles is a problem of recovery from trauma and denervation. The aim of the work was to investigate the interconnection of matrix metalloproteinase-9 (ММР-9) activity in denervated muscles with fibrosis and to estimate its role in nerve restoration by the epineurial suture, fibrin-based glue, and polyethylene glycol hydrogel. The activity of matrix metalloproteinases was estimated by gelatin zymography. Collagen density in muscles was determined histochemically. An increased level of the active MMP-9 is associated with the fibrous changes in the denervated skeletal muscles and after an epineurial suture. The use of fibrin glue and polyethylene glycol hydrogel resulted in a lower level of collagen and ММР-9 activity, which may be a therapeutic target in the treatment of neuromuscular lesions, and has value in fibrosis analysis following microsurgical intervention for peripheral nerve reconstruction.
Collapse
Affiliation(s)
- Oleksii Goncharuk
- Department of Neurosurgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Serhii Savosko
- Department of Histology and Embryology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Artem Tykhomyrov
- Department of Neurosurgery, Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine (NASU), Kyiv, Ukraine
| | - Mykhailo Guzyk
- Department of Neurosurgery, Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine (NASU), Kyiv, Ukraine
| | - Volodymyr Medvediev
- Department of Neurosurgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Vitaliy Tsymbaliuk
- Department of Neurosurgery, Bogomolets National Medical University, Kyiv, Ukraine
| | - Yuri Chaikovsky
- Department of Histology and Embryology, Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
9
|
Muraine L, Bensalah M, Butler-Browne G, Bigot A, Trollet C, Mouly V, Negroni E. Update on anti-fibrotic pharmacotherapies in skeletal muscle disease. Curr Opin Pharmacol 2023; 68:102332. [PMID: 36566666 DOI: 10.1016/j.coph.2022.102332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
Fibrosis, defined as an excessive accumulation of extracellular matrix, is the end point of a defective regenerative process, unresolved inflammation and/or chronic damage. Numerous muscle disorders (MD) are characterized by high levels of fibrosis associated with muscle wasting and weakness. Fibrosis alters muscle homeostasis/regeneration and fiber environment and may interfere with gene and cell therapies. Slowing down or reversing fibrosis is a crucial therapeutic goal to maintain muscle identity in the context of therapies. Several pathways are implicated in the modulation of the fibrotic progression and multiple therapeutic compounds targeting fibrogenic signals have been tested in MDs, mostly in the context of Duchenne Muscular Dystrophy. In this review, we present an up-to-date overview of pharmacotherapies that have been tested to reduce fibrosis in the skeletal muscle.
Collapse
Affiliation(s)
- Laura Muraine
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Mona Bensalah
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Capucine Trollet
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France.
| | - Elisa Negroni
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France.
| |
Collapse
|
10
|
Molina T, Fabre P, Dumont NA. Fibro-adipogenic progenitors in skeletal muscle homeostasis, regeneration and diseases. Open Biol 2021; 11:210110. [PMID: 34875199 PMCID: PMC8651418 DOI: 10.1098/rsob.210110] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle possesses a remarkable regenerative capacity that relies on the activity of muscle stem cells, also known as satellite cells. The presence of non-myogenic cells also plays a key role in the coordination of skeletal muscle regeneration. Particularly, fibro-adipogenic progenitors (FAPs) emerged as master regulators of muscle stem cell function and skeletal muscle regeneration. This population of muscle resident mesenchymal stromal cells has been initially characterized based on its bi-potent ability to differentiate into fibroblasts or adipocytes. New technologies such as single-cell RNAseq revealed the cellular heterogeneity of FAPs and their complex regulatory network during muscle regeneration. In acute injury, FAPs rapidly enter the cell cycle and secrete trophic factors that support the myogenic activity of muscle stem cells. Conversely, deregulation of FAP cell activity is associated with the accumulation of fibrofatty tissue in pathological conditions such as muscular dystrophies and ageing. Considering their central role in skeletal muscle pathophysiology, the regulatory mechanisms of FAPs and their cellular and molecular crosstalk with muscle stem cells are highly investigated in the field. In this review, we summarize the current knowledge on FAP cell characteristics, heterogeneity and the cellular crosstalk during skeletal muscle homeostasis and regeneration. We further describe their role in muscular disorders, as well as different therapeutic strategies targeting these cells to restore muscle regeneration.
Collapse
Affiliation(s)
- Thomas Molina
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Paul Fabre
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas A. Dumont
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Wilburn D, Ismaeel A, Machek S, Fletcher E, Koutakis P. Shared and distinct mechanisms of skeletal muscle atrophy: A narrative review. Ageing Res Rev 2021; 71:101463. [PMID: 34534682 DOI: 10.1016/j.arr.2021.101463] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022]
Abstract
Maintenance of skeletal muscle mass and function is an incredibly nuanced balance of anabolism and catabolism that can become distorted within different pathological conditions. In this paper we intend to discuss the distinct intracellular signaling events that regulate muscle protein atrophy for a given clinical occurrence. Aside from the common outcome of muscle deterioration, several conditions have at least one or more distinct mechanisms that creates unique intracellular environments that facilitate muscle loss. The subtle individuality to each of these given pathologies can provide both researchers and clinicians with specific targets of interest to further identify and increase the efficacy of medical treatments and interventions.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76706, USA
| | - Steven Machek
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Emma Fletcher
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; Department of Biology, Baylor University, Waco, TX 76706, USA
| | | |
Collapse
|
12
|
Marine T, Marielle S, Graziella M, Fabio RMV. Macrophages in Skeletal Muscle Dystrophies, An Entangled Partner. J Neuromuscul Dis 2021; 9:1-23. [PMID: 34542080 PMCID: PMC8842758 DOI: 10.3233/jnd-210737] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While skeletal muscle remodeling happens throughout life, diseases that result in its dysfunction are accountable for many deaths. Indeed, skeletal muscle is exceptionally capable to respond to stimuli modifying its homeostasis, such as in atrophy, hypertrophy, regeneration and repair. In particular conditions such as genetic diseases (muscular dystrophies), skeletal muscle’s capacity to remodel is strongly affected and undergoes continuous cycles of chronic damage. This induces scarring, fatty infiltration, as well as loss of contractibility and of the ability to generate force. In this context, inflammation, primarily mediated by macrophages, plays a central pathogenic role. Macrophages contribute as the primary regulators of inflammation during skeletal muscle regeneration, affecting tissue-resident cells such as myogenic cells and endothelial cells, but also fibro-adipogenic progenitors, which are the main source of the fibro fatty scar. During skeletal muscle regeneration their function is tightly orchestrated, while in dystrophies their fate is strongly disturbed, resulting in chronic inflammation. In this review, we will discuss the latest findings on the role of macrophages in skeletal muscle diseases, and how they are regulated.
Collapse
Affiliation(s)
- Theret Marine
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia, Vancouver BC, Canada
| | - Saclier Marielle
- Department of Biosciences, University of Milan, via Celoria, Milan, Italy
| | - Messina Graziella
- Department of Biosciences, University of Milan, via Celoria, Milan, Italy
| | - Rossi M V Fabio
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia, Vancouver BC, Canada
| |
Collapse
|
13
|
Asano T, Tsujii M, Iino T, Odake K, Sudo A. Pathological features of reinnervated skeletal muscles after crush injury of the sciatic nerve in ob/ob mice. Muscle Nerve 2021; 64:365-373. [PMID: 34212392 DOI: 10.1002/mus.27365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 06/21/2021] [Accepted: 06/27/2021] [Indexed: 11/11/2022]
Abstract
INTRODUCTION/AIMS Obesity is a factor contributing to suboptimal improvement of motor function in peripheral nerve disorders. In this study we aimed to evaluate the skeletal muscles during denervation and reinnervation after nerve crush injury in leptin-deficient (ob/ob) mice. METHODS Experiments were performed on the skeletal muscles of the hindlimbs in 20 male ob/ob mice and controls. Characteristics of the gastrocnemius muscles were evaluated by histological analysis, immunohistological analysis, and Sircol-collagen assay after measurement of body weight and wet weight of the skeletal muscles, and by walking track analysis. The sciatic nerve was denervated by crushing with smooth forceps and reinnervation was evaluated. RESULTS Gastrocnemius wet weight was significantly lower in the ob/ob mice than in the control mice. A smaller cross-sectional area of type II fibers and increase of type I fiber grouping of the skeletal muscles was demonstrated in the ob/ob mice. After nerve injury, motor function recovery was equal between the groups but the cross-sectional area of type II fibers was significantly smaller in the ob/ob mice than in control mice at 4 weeks. The denervated muscles showed an increase in collagen deposition in the interstitial space; predominant in the ob/ob mice after nerve injury. DISCUSSION The results of this study suggest that fibrosis in the skeletal muscle of obese patients after nerve injury is prominent, which may impair improvement of muscle function after treatment of peripheral nerve disorders.
Collapse
Affiliation(s)
- Takahiro Asano
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masaya Tsujii
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Kazuya Odake
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Tsu, Japan
| |
Collapse
|
14
|
Zhao L, Liu X, Zhang J, Dong G, Xiao W, Xu X. Hydrogen Sulfide Alleviates Skeletal Muscle Fibrosis via Attenuating Inflammation and Oxidative Stress. Front Physiol 2020; 11:533690. [PMID: 33071808 PMCID: PMC7530892 DOI: 10.3389/fphys.2020.533690] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study was to investigate the effect of exogenous hydrogen sulfide (H2S) treatment on skeletal muscle contusion. We established a skeletal muscle contusion model (S group) and an H2S treated of skeletal muscle contusion model (H2S group). Gastrocnemius muscles (GMs) were collected at day 1, day 5, day 10, and day 15 after injury, and comprehensive morphological and genetic analyses was conducted. H2S treatment reduced M1 macrophage (CD68), profibrotic cytokines (TGF-β), pro-inflammatory cytokines (TNF-α, IFN-γ, IL-1β, and IL-6), chemokines (CCL2, CCR2, CCL3, CCL5, CXCL12, and CXCR4), matrix metalloproteinases (MMP-1, MMP-2, MMP-9, and MMP-14) and oxidative stress factor (gp91phox) expression levels, improved M2 macrophage (CD206) level. Thus, exogenous H2S treatment reduced inflammation and oxidative stress, attenuated skeletal muscle fibrosis, and partly improved skeletal muscle injury.
Collapse
Affiliation(s)
- Linlin Zhao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jing Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Gaoyang Dong
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
15
|
Cappellari O, Mantuano P, De Luca A. "The Social Network" and Muscular Dystrophies: The Lesson Learnt about the Niche Environment as a Target for Therapeutic Strategies. Cells 2020; 9:cells9071659. [PMID: 32660168 PMCID: PMC7407800 DOI: 10.3390/cells9071659] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
The muscle stem cells niche is essential in neuromuscular disorders. Muscle injury and myofiber death are the main triggers of muscle regeneration via satellite cell activation. However, in degenerative diseases such as muscular dystrophy, regeneration still keep elusive. In these pathologies, stem cell loss occurs over time, and missing signals limiting damaged tissue from activating the regenerative process can be envisaged. It is unclear what comes first: the lack of regeneration due to satellite cell defects, their pool exhaustion for degeneration/regeneration cycles, or the inhibitory mechanisms caused by muscle damage and fibrosis mediators. Herein, Duchenne muscular dystrophy has been taken as a paradigm, as several drugs have been tested at the preclinical and clinical levels, targeting secondary events in the complex pathogenesis derived from lack of dystrophin. We focused on the crucial roles that pro-inflammatory and pro-fibrotic cytokines play in triggering muscle necrosis after damage and stimulating satellite cell activation and self-renewal, along with growth and mechanical factors. These processes contribute to regeneration and niche maintenance. We review the main effects of drugs on regeneration biomarkers to assess whether targeting pathogenic events can help to protect niche homeostasis and enhance regeneration efficiency other than protecting newly formed fibers from further damage.
Collapse
|
16
|
Anti-Inflammatory and General Glucocorticoid Physiology in Skeletal Muscles Affected by Duchenne Muscular Dystrophy: Exploration of Steroid-Sparing Agents. Int J Mol Sci 2020; 21:ijms21134596. [PMID: 32605223 PMCID: PMC7369834 DOI: 10.3390/ijms21134596] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/13/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), the activation of proinflammatory and metabolic cellular pathways in skeletal muscle cells is an inherent characteristic. Synthetic glucocorticoid intake counteracts the majority of these mechanisms. However, glucocorticoids induce burdensome secondary effects, including hypertension, arrhythmias, hyperglycemia, osteoporosis, weight gain, growth delay, skin thinning, cushingoid appearance, and tissue-specific glucocorticoid resistance. Hence, lowering the glucocorticoid dosage could be beneficial for DMD patients. A more profound insight into the major cellular pathways that are stabilized after synthetic glucocorticoid administration in DMD is needed when searching for the molecules able to achieve similar pathway stabilization. This review provides a concise overview of the major anti-inflammatory pathways, as well as the metabolic effects of glucocorticoids in the skeletal muscle affected in DMD. The known drugs able to stabilize these pathways, and which could potentially be combined with glucocorticoid therapy as steroid-sparing agents, are described. This could create new opportunities for testing in DMD animal models and/or clinical trials, possibly leading to smaller glucocorticoids dosage regimens for DMD patients.
Collapse
|
17
|
Heterogenetic parabiosis between healthy and dystrophic mice improve the histopathology in muscular dystrophy. Sci Rep 2020; 10:7075. [PMID: 32341395 PMCID: PMC7184587 DOI: 10.1038/s41598-020-64042-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/09/2020] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease, characterized by mutations in the X-linked dystrophin, that has several therapeutic options but no curative treatment. Transplantation of muscle progenitor cells for treatment of DMD has been widely investigated; however, its application is hindered by limited cell survival due to the harmful dystrophic microenvironment. An alternative approach to utilize progenitor cells and circulatory factors and to improve the dystrophic muscle pathology and microenvironment is through parabiotic pairing, where mice are surgically sutured to create a joint circulatory system. Parabiotic mice were generated by surgically joining wild type (WT) mice expressing green fluorescent protein (GFP) with mdx mice. These mice developed a common circulation (approximately 50% green cells in the blood of mdx mice) 2-weeks after parabiotic pairing. We observed significantly improved dystrophic muscle pathology, including decreased inflammation, necrotic fibers and fibrosis in heterogenetic parabionts. Importantly, the GFP + cells isolated from the mdx mice (paired with GFP mice) underwent myogenic differentiation in vitro and expressed markers of mesenchymal stem cells and macrophages, which may potentially be involved in the improvement of dystrophic muscle pathology. These observations suggest that changing the dystrophic microenvironment can be a new approach to treat DMD.
Collapse
|
18
|
Adhipandito CF, Ludji DPKS, Aprilianto E, Jenie RI, Al-Najjar B, Hariono M. Matrix metalloproteinase9 as the protein target in anti-breast cancer drug discovery: an approach by targeting hemopexin domain. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2019. [DOI: 10.1186/s43094-019-0001-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
19
|
Hayward AN, Aird EJ, Gordon WR. A toolkit for studying cell surface shedding of diverse transmembrane receptors. eLife 2019; 8:e46983. [PMID: 31172946 PMCID: PMC6586460 DOI: 10.7554/elife.46983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022] Open
Abstract
Proteolysis of transmembrane receptors is a critical cellular communication mechanism dysregulated in disease, yet decoding proteolytic regulation mechanisms of hundreds of shed receptors is hindered by difficulties controlling stimuli and unknown fates of cleavage products. Notch proteolytic regulation is a notable exception, where intercellular forces drive exposure of a cryptic protease site within a juxtamembrane proteolytic switch domain to activate transcriptional programs. We created a Synthetic Notch Assay for Proteolytic Switches (SNAPS) that exploits the modularity and unequivocal input/response of Notch proteolysis to screen surface receptors for other putative proteolytic switches. We identify several new proteolytic switches among receptors with structural homology to Notch. We demonstrate SNAPS can detect shedding in chimeras of diverse cell surface receptors, leading to new, testable hypotheses. Finally, we establish the assay can be used to measure modulation of proteolysis by potential therapeutics and offer new mechanistic insights into how DECMA-1 disrupts cell adhesion.
Collapse
Affiliation(s)
- Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaMinneapolisUnited States
| | - Eric J Aird
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaMinneapolisUnited States
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaMinneapolisUnited States
| |
Collapse
|
20
|
Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun 2019; 10:2430. [PMID: 31160583 PMCID: PMC6547715 DOI: 10.1038/s41467-019-10438-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/10/2019] [Indexed: 12/30/2022] Open
Abstract
Muscle loss due to fibrotic or adipogenic replacement of myofibers is common in muscle diseases and muscle-resident fibro/adipogenic precursors (FAPs) are implicated in this process. While FAP-mediated muscle fibrosis is widely studied in muscle diseases, the role of FAPs in adipogenic muscle loss is not well understood. Adipogenic muscle loss is a feature of limb girdle muscular dystrophy 2B (LGMD2B) - a disease caused by mutations in dysferlin. Here we show that FAPs cause the adipogenic loss of dysferlin deficient muscle. Progressive accumulation of Annexin A2 (AnxA2) in the myofiber matrix causes FAP differentiation into adipocytes. Lack of AnxA2 prevents FAP adipogenesis, protecting against adipogenic loss of dysferlinopathic muscle while exogenous AnxA2 enhances muscle loss. Pharmacological inhibition of FAP adipogenesis arrests adipogenic replacement and degeneration of dysferlin-deficient muscle. These results demonstrate the pathogenic role of FAPs in LGMD2B and establish these cells as therapeutic targets to ameliorate muscle loss in patients.
Collapse
|
21
|
Liu X, Zeng Z, Zhao L, Chen P, Xiao W. Impaired Skeletal Muscle Regeneration Induced by Macrophage Depletion Could Be Partly Ameliorated by MGF Injection. Front Physiol 2019; 10:601. [PMID: 31164836 PMCID: PMC6534059 DOI: 10.3389/fphys.2019.00601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle injury is one of the most common injuries in sports medicine. Our previous study found that macrophage depletion impairs muscle regeneration and that mechano growth factor (MGF) may play an important role in this process. However, whether injection of MGF protects against impaired muscle regeneration after macrophage depletion has not been explored. Therefore, we generated a muscle contusion and macrophage depletion mouse model and injected MGF into the damaged muscle. Comprehensive morphological and genetic analyses were performed on the injured skeletal muscle after macrophage depletion and MGF injection. The results showed that injection of MGF did not exert a protective effect on muscle fiber regeneration; however, it did decrease fibrosis in the contused skeletal muscle after macrophage depletion. Moreover, MGF injection decreased the expression of muscle inflammatory cytokines (TNF-α, IFN-γ, IL-1β, and TGF-β), chemokines (CCL2, CCL5, and CXCR4), oxidative stress factors (gp91phox) and matrix metalloproteinases (MMP-1, MMP-2, MMP-9, MMP-10, and MMP-14). These results suggest that the impairment of skeletal muscle regeneration induced by macrophage depletion could be partly ameliorated by MGF injection and that inflammatory cytokines, oxidative stress factors, chemokines, and MMP may be involved in this process.
Collapse
Affiliation(s)
- Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Zhigang Zeng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,College of Physical Education, Jinggangshan University, Jiangxi, China
| | - Linlin Zhao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
22
|
Gallot YS, Straughn AR, Bohnert KR, Xiong G, Hindi SM, Kumar A. MyD88 is required for satellite cell-mediated myofiber regeneration in dystrophin-deficient mdx mice. Hum Mol Genet 2019; 27:3449-3463. [PMID: 30010933 DOI: 10.1093/hmg/ddy258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, leads to severe muscle wasting and eventual death of the afflicted individuals, primarily due to respiratory failure. Deficit in myofiber regeneration, potentially due to an exhaustion of satellite cells, is one of the major pathological features of DMD. Myeloid differentiation primary response 88 (MyD88) is an adaptor protein that mediates activation of various inflammatory pathways in response to signaling from Toll-like receptors and interleukin-1 receptor. MyD88 also regulates cellular survival, proliferation and differentiation in a cell-autonomous manner. However, the role of MyD88 in satellite stem cell homeostasis and function in dystrophic muscle remains unknown. In this study, we demonstrate that tamoxifen-inducible deletion of MyD88 in satellite cells causes loss of skeletal muscle mass and strength in the mdx mouse model of DMD. Satellite cell-specific deletion of MyD88 inhibits myofiber regeneration and stimulates fibrogenesis in dystrophic muscle of mdx mice. Deletion of MyD88 also reduces the number of satellite cells and inhibits their fusion with injured myofibers in dystrophic muscle of mdx mice. Ablation of MyD88 in satellite cells increases the markers of M2 macrophages without having any significant effect on M1 macrophages and expression of inflammatory cytokines. Finally, we found that satellite cell-specific deletion of MyD88 leads to aberrant activation of Notch and Wnt signaling in skeletal muscle of mdx mice. Collectively, our results demonstrate that MyD88-mediated signaling in satellite cells is essential for the regeneration of injured myofibers in dystrophic muscle of mdx mice.
Collapse
Affiliation(s)
- Yann S Gallot
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Alex R Straughn
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kyle R Bohnert
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Guangyan Xiong
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
23
|
Liu X, Zheng L, Zhou Y, Chen Y, Chen P, Xiao W. BMSC Transplantation Aggravates Inflammation, Oxidative Stress, and Fibrosis and Impairs Skeletal Muscle Regeneration. Front Physiol 2019; 10:87. [PMID: 30814953 PMCID: PMC6382023 DOI: 10.3389/fphys.2019.00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/24/2019] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle contusion is one of the most common muscle injuries in sports medicine and traumatology. Bone marrow mesenchymal stem cell (BMSC) transplantation has been proposed as a promising strategy to promote skeletal muscle regeneration. However, the roles and underlying mechanisms of BMSCs in the regulation of skeletal muscle regeneration are still not completely clear. Here, we investigated the role of BMSC transplantation after muscle contusion. BMSCs were immediately transplanted into gastrocnemius muscles (GMs) following direct contusion. Comprehensive morphological and genetic analyses were performed after BMSC transplantation. BMSC transplantation exacerbated muscle fibrosis and inflammation, as evidenced by increased leukocyte and macrophage infiltration, increased inflammatory cytokines and chemokines, and increased matrix metalloproteinases. BMSC transplantation also increased muscle oxidative stress. Overall, BMSC transplantation aggravated inflammation, oxidative stress and fibrosis and impaired skeletal muscle regeneration. These results, shed new light on the role of BMSCs in regenerative medicine and indicate that caution is needed in the application of BMSCs for muscle injury.
Collapse
Affiliation(s)
- Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lifang Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yongzhan Zhou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yingjie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
24
|
Soslow JH, Xu M, Slaughter JC, Crum K, Chew JD, Burnette WB, Su YR, Tomasek K, Parra DA, Markham LW. The Role of Matrix Metalloproteinases and Tissue Inhibitors of Metalloproteinases in Duchenne Muscular Dystrophy Cardiomyopathy. J Card Fail 2019; 25:259-267. [PMID: 30763738 DOI: 10.1016/j.cardfail.2019.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD). Standard cardiac biomarkers are poor indicators of DMD cardiovascular disease. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) regulate collagen turnover. Given the cardiac fibrosis seen in DMD, we hypothesized that MMPs and TIMPs correlate with severity of DMD cardiomyopathy. METHODS AND RESULTS Prospectively enrolled DMD subjects (n = 42) underwent cardiac magnetic resonance imaging for function and late gadolinium enhancement (LGE), including LGE severity from 0 (no LGE) to 4 (severe). Serum from DMD and healthy male control subjects (n = 15) analyzed for MMPs 1, 2, 3, 7, 9, and 10 and TIMPs 1-4. MMP1, MMP7, and MMP10 were higher in DMD than in control (respectively, median 5080 pg/mL vs 2120 pg/mL [P = .007], 2170 pg/mL vs 1420 pg/mL [P < .001], and 216 pg/mL vs 140pg/mL [P = .040]); TIMP4 was lower in DMD (124 pg/mL vs 263 pg/mL; P = .046). Within DMD, MMP7 correlated inversely with left ventricular ejection fraction (r = -0.40; P = .012) and directly with strain (r = 0.54; P = .001) and LGE severity (r = 0.47; P = .003). MMP7 was higher in DMD patients with LGE compared with those without LGE and control subjects (P < .001). CONCLUSIONS Multiple MMPs are elevated in DMD compared with control subjects. MMP7 is related to DMD cardiac dysfunction and myocardial fibrosis, possibly through remodeling of the extracellular matrix.
Collapse
Affiliation(s)
- Jonathan H Soslow
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kimberly Crum
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua D Chew
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - W Bryan Burnette
- Division of Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yan Ru Su
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kelsey Tomasek
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David A Parra
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Larry W Markham
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
25
|
Alameddine HS, Morgan JE. Matrix Metalloproteinases and Tissue Inhibitor of Metalloproteinases in Inflammation and Fibrosis of Skeletal Muscles. J Neuromuscul Dis 2018; 3:455-473. [PMID: 27911334 PMCID: PMC5240616 DOI: 10.3233/jnd-160183] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In skeletal muscles, levels and activity of Matrix MetalloProteinases (MMPs) and Tissue Inhibitors of MetalloProteinases (TIMPs) have been involved in myoblast migration, fusion and various physiological and pathological remodeling situations including neuromuscular diseases. This has opened perspectives for the use of MMPs' overexpression to improve the efficiency of cell therapy in muscular dystrophies and resolve fibrosis. Alternatively, inhibition of individual MMPs in animal models of muscular dystrophies has provided evidence of beneficial, dual or adverse effects on muscle morphology or function. We review here the role played by MMPs/TIMPs in skeletal muscle inflammation and fibrosis, two major hurdles that limit the success of cell and gene therapy. We report and analyze the consequences of genetic or pharmacological modulation of MMP levels on the inflammation of skeletal muscles and their repair in light of experimental findings. We further discuss how the interplay between MMPs/TIMPs levels, cytokines/chemokines, growth factors and permanent low-grade inflammation favor cellular and molecular modifications resulting in fibrosis.
Collapse
Affiliation(s)
- Hala S Alameddine
- Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, boulevard de l'Hôpital, 75651 Paris Cedex 13, France
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, UK
| |
Collapse
|
26
|
Perandini LA, Chimin P, Lutkemeyer DDS, Câmara NOS. Chronic inflammation in skeletal muscle impairs satellite cells function during regeneration: can physical exercise restore the satellite cell niche? FEBS J 2018; 285:1973-1984. [PMID: 29473995 DOI: 10.1111/febs.14417] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/27/2018] [Accepted: 02/19/2018] [Indexed: 12/23/2022]
Abstract
Chronic inflammation impairs skeletal muscle regeneration. Although many cells are involved in chronic inflammation, macrophages seem to play an important role in impaired muscle regeneration since these cells are associated with skeletal muscle stem cell (namely, satellite cells) activation and fibro-adipogenic progenitor cell (FAP) survival. Specifically, an imbalance of M1 and M2 macrophages seems to lead to impaired satellite cell activation, and these are the main cells that function during skeletal muscle regeneration, after muscle damage. Additionally, this imbalance leads to the accumulation of FAPs in skeletal muscle, with aberrant production of pro-fibrotic factors (e.g., extracellular matrix components), impairing the niche for proper satellite cell activation and differentiation. Treatments aiming to block the inflammatory pro-fibrotic response are partially effective due to their side effects. Therefore, strategies reverting chronic inflammation into a pro-regenerative pattern are required. In this review, we first describe skeletal muscle resident macrophage ontogeny and homeostasis, and explain how macrophages are replenished after muscle injury. We next discuss the potential role of chronic physical activity and exercise in restoring the M1 and M2 macrophage balance and consequently, the satellite cell niche to improve skeletal muscle regeneration after injury.
Collapse
Affiliation(s)
- Luiz Augusto Perandini
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Patricia Chimin
- Department of Physical Education, Physical Education and Sports Center, Londrina State University, Brazil
| | - Diego da Silva Lutkemeyer
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil.,Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Department of Medicine, Federal University of Sao Paulo, Brazil
| |
Collapse
|
27
|
[Molecular mechanism of sarcopenia]. Nihon Ronen Igakkai Zasshi 2018; 55:13-24. [PMID: 29503355 DOI: 10.3143/geriatrics.55.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
|
28
|
de Carvalho SC, Hindi SM, Kumar A, Marques MJ. Effects of omega-3 on matrix metalloproteinase-9, myoblast transplantation and satellite cell activation in dystrophin-deficient muscle fibers. Cell Tissue Res 2017. [PMID: 28623422 DOI: 10.1007/s00441-017-2640-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In Duchenne muscular dystrophy (DMD), lack of dystrophin leads to progressive muscle degeneration, with DMD patients suffering from cardiorespiratory failure. Cell therapy is an alternative to life-long corticoid therapy. Satellite cells, the stem cells of skeletal muscles, do not completely compensate for the muscle damage in dystrophic muscles. Elevated levels of proinflammatory and profibrotic factors, such as metalloproteinase 9 (MMP-9), impair muscle regeneration, leading to extensive fibrosis and poor results with myoblast transplantation therapies. Omega-3 is an anti-inflammatory drug that protects against muscle degeneration in the mdx mouse model of DMD. In the present study, we test our hypothesis that omega-3 affects MMP-9 and thereby benefits muscle regeneration and myoblast transplantation in the mdx mouse. We observe that omega-3 reduces MMP-9 gene expression and improves myoblast engraftment, satellite cell activation, and muscle regeneration by mechanisms involving, at least in part, the regulation of macrophages, as shown here with the fluorescence-activated cell sorting technique. The present study demonstrates the benefits of omega-3 on satellite cell survival and muscle regeneration, further supporting its use in clinical trials and cell therapies in DMD.
Collapse
Affiliation(s)
- Samara Camaçari de Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, CEP 1083-970, Brazil
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Maria Julia Marques
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, SP, CEP 1083-970, Brazil.
| |
Collapse
|
29
|
Capogrosso RF, Mantuano P, Cozzoli A, Sanarica F, Massari AM, Conte E, Fonzino A, Giustino A, Rolland JF, Quaranta A, De Bellis M, Camerino GM, Grange RW, De Luca A. Contractile efficiency of dystrophic mdx mouse muscle: in vivo and ex vivo assessment of adaptation to exercise of functional end points. J Appl Physiol (1985) 2017; 122:828-843. [PMID: 28057817 DOI: 10.1152/japplphysiol.00776.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 11/22/2022] Open
Abstract
Progressive weakness is a typical feature of Duchenne muscular dystrophy (DMD) patients and is exacerbated in the benign mdx mouse model by in vivo treadmill exercise. We hypothesized a different threshold for functional adaptation of mdx muscles in response to the duration of the exercise protocol. In vivo weakness was confirmed by grip strength after 4, 8, and 12 wk of exercise in mdx mice. Torque measurements revealed that exercise-related weakness in mdx mice correlated with the duration of the protocol, while wild-type (WT) mice were stronger. Twitch and tetanic forces of isolated diaphragm and extensor digitorum longus (EDL) muscles were lower in mdx compared with WT mice. In mdx, both muscle types exhibited greater weakness after a single exercise bout, but only in EDL after a long exercise protocol. As opposite to WT muscles, mdx EDL ones did not show any exercise-induced adaptations against eccentric contraction force drop. qRT-PCR analysis confirmed the maladaptation of genes involved in metabolic and structural remodeling, while damage-related genes remained significantly upregulated and angiogenesis impaired. Phosphorylated AMP kinase level increased only in exercised WT muscle. The severe histopathology and the high levels of muscular TGF-β1 and of plasma matrix metalloproteinase-9 confirmed the persistence of muscle damage in mdx mice. Therefore, dystrophic muscles showed a partial degree of functional adaptation to chronic exercise, although not sufficient to overcome weakness nor signs of damage. The improved understanding of the complex mechanisms underlying maladaptation of dystrophic muscle paves the way to a better managment of DMD patients.NEW & NOTEWORTHY We focused on the adaptation/maladaptation of dystrophic mdx mouse muscles to a standard protocol of exercise to provide guidance in the development of more effective drug and physical therapies in Duchenne muscular dystrophy. The mdx muscles showed a modest functional adaptation to chronic exercise, but it was not sufficient to overcome the progressive in vivo weakness, nor to counter signs of muscle damage. Therefore, a complex involvement of multiple systems underlies the maladaptive response of dystrophic muscle.
Collapse
Affiliation(s)
- Roberta Francesca Capogrosso
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy.,Department of Chemical, Toxicological and Pharmacological Drug Studies, Catholic University "Our Lady of Good Counsel," Tirana, Albany
| | - Paola Mantuano
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Anna Cozzoli
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Francesca Sanarica
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Ada Maria Massari
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Elena Conte
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Adriano Fonzino
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Arcangela Giustino
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Jean-Francois Rolland
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Angelo Quaranta
- Department of Veterinary Medicine, University of Bari "Aldo Moro," Valenzano (BA), Italy
| | - Michela De Bellis
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Giulia Maria Camerino
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech University, Blacksburg, Virginia; and
| | - Annamaria De Luca
- Section of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari "Aldo Moro," Bari, Italy;
| |
Collapse
|
30
|
Arvanitidis A, Henriksen K, Karsdal M, Nedergaard A. Neo-epitope Peptides as Biomarkers of Disease Progression for Muscular Dystrophies and Other Myopathies. J Neuromuscul Dis 2016; 3:333-346. [PMID: 27854226 PMCID: PMC5123625 DOI: 10.3233/jnd-160150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For several decades, serological biomarkers of neuromuscular diseases as dystrophies, myopathies and myositis have been limited to routine clinical biochemistry panels. Gauging the pathological progression is a prerequisite for proper treatment and therefore identifying accessible, easy to monitor biomarkers that can predict the disease progression would be an important advancement. Most muscle diseases involve accelerated muscle fiber degradation, inflammation, fatty tissue substitution and/or fibrosis. All these pathological traits have been shown to give rise to serological peptide biomarkers in other tissues, underlining the potential application of existing biomarkers of such traits in muscle disorders. A significant quantity of tissue is involved in these pathological mechanisms alongside with qualitative changes in protein turnover in myofibrillar, extra-cellular matrix and immunological cell protein fractions accompanied by alterations in body fluids. We propose that protein and peptides can leak out of the afflicted muscles and can be of use in diagnosis, prediction of pathology trajectory and treatment efficacy. Proteolytic cleavage systems are especially modulated during a range of muscle pathologies, thereby giving rise to peptides that are differentially released during disease manifestation. Therefore, we believe that pathology-specific post-translational modifications like cleavages can give rise to neoepitope peptides that may represent a promising class of peptides for discovery of biomarkers pertaining to neuromuscular diseases.
Collapse
Affiliation(s)
- A. Arvanitidis
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - K. Henriksen
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - M.A. Karsdal
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - A. Nedergaard
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| |
Collapse
|
31
|
Barbin ICC, Pereira JA, Bersan Rovere M, de Oliveira Moreira D, Marques MJ, Santo Neto H. Diaphragm degeneration and cardiac structure in mdx mouse: potential clinical implications for Duchenne muscular dystrophy. J Anat 2016; 228:784-91. [PMID: 26822140 DOI: 10.1111/joa.12443] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 02/03/2023] Open
Abstract
We examined the effects of exercise on diaphragm degeneration and cardiomyopathy in dystrophin-deficient mdx mice. Mdx mice (11 months of age) were exercised (swimming) for 2 months to worsen diaphragm degeneration. Control mdx mice were kept sedentary. Morphological evaluation demonstrated increased fibrosis in the diaphragm of exercised mdx mice (33.3 ± 6.0% area of fibrosis) compared with control mdx mice (20.9 ± 1.7% area of fibrosis). Increased (26%) activity of MMP-2, a marker of fibrosis, was detected in the diaphragms from exercised mdx mice. Morphological evaluation of the heart demonstrated a 45% increase in fibrosis in the right ventricle (8.3 ± 0.6% in sedentary vs. 12.0 ± 0.6% of fibrosis in exercised) and in the left ventricle (35% increase) in the exercised mdx mice. The density of inflammatory cells-degenerating cardiomyocytes increased 95% in the right ventricle (2.3 ± 0.6 in sedentary vs. 4.5 ± 0.8 in exercised) and 71% in the left ventricle (1.4 ± 0.6 sedentary vs. 2.4 ± 0.5 exercised). The levels of both active MMP-2 and the pro-fibrotic factor transforming growth factor beta were elevated in the hearts of exercised compared with sedentary mdx mice. The wall thickness to lumen diameter ratio of the pulmonary trunk was significantly increased in the exercised mdx mice (0.11 ± 0.04 in sedentary vs. 0.28 ± 0.12 in exercised), as was the thickness of the right ventricle wall, which suggests the occurrence of pulmonary hypertension in those animals. It is suggested that diaphragm degeneration is a main contributor to right ventricle dystrophic pathology. These findings may be relevant for future interventional studies for Duchenne muscular dystrophy-associated cardiomyopathy.
Collapse
Affiliation(s)
- Isabel Cristina Chagas Barbin
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Juliano Alves Pereira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Matheus Bersan Rovere
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Drielen de Oliveira Moreira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Maria Julia Marques
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Humberto Santo Neto
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
32
|
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: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
|
33
|
Holland A, Murphy S, Dowling P, Ohlendieck K. Pathoproteomic profiling of the skeletal muscle matrisome in dystrophinopathy associated myofibrosis. Proteomics 2015; 16:345-66. [PMID: 26256116 DOI: 10.1002/pmic.201500158] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/12/2015] [Accepted: 07/24/2015] [Indexed: 12/14/2022]
Abstract
The gradual accumulation of collagen and associated proteins of the extracellular matrix is a crucial myopathological parameter of many neuromuscular disorders. Progressive tissue damage and fibrosis play a key pathobiochemical role in the dysregulation of contractile functions and often correlates with poor motor outcome in muscular dystrophies. Following a brief introduction into the role of the extracellular matrix in skeletal muscles, we review here the proteomic profiling of myofibrosis and its intrinsic role in X-linked muscular dystrophy. Although Duchenne muscular dystrophy is primarily a disease of the membrane cytoskeleton, one of its most striking histopathological features is a hyperactive connective tissue and tissue scarring. We outline the identification of novel factors involved in the modulation of the extracellular matrix in muscular dystrophy, such as matricellular proteins. The establishment of novel proteomic markers will be helpful in improving the diagnosis, prognosis, and therapy monitoring in relation to fibrotic substitution of contractile tissue. In the future, the prevention of fibrosis will be crucial for providing optimum conditions to apply novel pharmacological treatments, as well as establish cell-based approaches or gene therapeutic interventions. The elimination of secondary abnormalities in the matrisome promises to reduce tissue scarring and the loss of skeletal muscle elasticity.
Collapse
Affiliation(s)
- Ashling Holland
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
34
|
Differential roles of MMP-9 in early and late stages of dystrophic muscles in a mouse model of Duchenne muscular dystrophy. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2170-82. [PMID: 26170062 DOI: 10.1016/j.bbadis.2015.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/05/2015] [Accepted: 07/08/2015] [Indexed: 01/08/2023]
Abstract
Matrix metalloprotease (MMP)-9 is an endopeptidase associated with the pathogenesis of Duchenne muscular dystrophy (DMD). The precise function of MMP-9 in DMD has not been elucidated to date. We investigated the effect of genetic ablation of MMP-9 in the mdx mouse model (mdx/Mmp9(-/-)). At the early disease stage, the muscles of mdx/Mmp9(-/-) mice showed reduced necrosis and neutrophil invasion, accompanied by down-regulation of chemokine MIP-2. In addition, muscle regeneration was enhanced, which coincided with increased macrophage infiltration and upregulation of MCP-1, and resulted in increased muscle strength. The mdx/Mmp9(-/-) mice also displayed accelerated upregulation of osteopontin expression in skeletal muscle at the acute onset phase of dystrophy. However, at a later disease stage, the mice exhibited muscle growth impairment through altered expression of myogenic factors, and increased fibroadipose tissue. These results showed that MMP-9 might have multiple functions during disease progression. Therapy targeting MMP-9 may improve muscle pathology and function at the early disease stage, but continuous inhibition of this protein may result in the accumulation of fibroadipose tissues and reduced muscle strength at the late disease stage.
Collapse
|
35
|
MicroRNA-431 accelerates muscle regeneration and ameliorates muscular dystrophy by targeting Pax7 in mice. Nat Commun 2015; 6:7713. [PMID: 26151913 DOI: 10.1038/ncomms8713] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 06/02/2015] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle stem cells, called satellite cells, are a quiescent heterogeneous population. Their heterogeneity is influenced by Pax7, a well-defined transcriptional regulator of satellite cell functions that defines two subpopulations: Pax7(Hi) and Pax7(Lo). However, the mechanisms by which these subpopulations are established and maintained during myogenesis are not completely understood. Here we show that miR-431, which is predominantly expressed in the skeletal muscle, mediates satellite cell heterogeneity by fine-tuning Pax7 levels during muscle development and regeneration. In miR-431 transgenic mice, the Pax7(Lo) subpopulation is enriched, enhances myogenic differentiation and accelerates muscle regeneration. Notably, miR-431 attenuates the muscular dystrophic phenotype in mdx mice and may be a potential therapeutic target in muscular diseases. miR-431 transgenic mice are a unique genetic model for investigating the cellular features and biological functions of Pax7(Lo) satellite cells during muscle development and regeneration.
Collapse
|
36
|
Devine RD, Bicer S, Reiser PJ, Velten M, Wold LE. Metalloproteinase expression is altered in cardiac and skeletal muscle in cancer cachexia. Am J Physiol Heart Circ Physiol 2015; 309:H685-91. [PMID: 26092976 DOI: 10.1152/ajpheart.00106.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/05/2015] [Indexed: 02/08/2023]
Abstract
Cardiac and skeletal muscle dysfunction is a recognized effect of cancer-induced cachexia, with alterations in heart function leading to heart failure and negatively impacting patient morbidity. Cachexia is a complex and multifaceted disease state with several potential contributors to cardiac and skeletal muscle dysfunction. Matrix metalloproteinases (MMPs) are a family of enzymes capable of degrading components of the extracellular matrix (ECM). Changes to the ECM cause disruption both in the connections between cells at the basement membrane and in cell-to-cell interactions. In the present study, we used a murine model of C26 adenocarcinoma-induced cancer cachexia to determine changes in MMP gene and protein expression in cardiac and skeletal muscle. We analyzed MMP-2, MMP-3, MMP-9, and MMP-14 as they have been shown to contribute to both cardiac and skeletal muscle ECM changes and, thereby, to pathology in models of heart failure and muscular dystrophy. In our model, cardiac and skeletal muscles showed a significant increase in RNA and protein levels of several MMPs and tissue inhibitors of metalloproteinases. Cardiac muscle showed significant protein increases in MMP-2, MMP-3, MMP-9, and MMP-14, whereas skeletal muscles showed increases in MMP-2, MMP-3, and MMP-14. Furthermore, collagen deposition was increased after C26 adenocarcinoma-induced cancer cachexia as indicated by an increased left ventricular picrosirius red-positive-stained area. Increases in serum hydroxyproline suggest increased collagen turnover, implicating skeletal muscle remodeling. Our findings demonstrate that cancer cachexia-associated matrix remodeling results in cardiac fibrosis and possible skeletal muscle remodeling. With these findings, MMPs represent a possible therapeutic target for the treatment of cancer-induced cachexia.
Collapse
Affiliation(s)
- Raymond D Devine
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio
| | - Sabahattin Bicer
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, Ohio
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms-University, University Medical Center, Bonn, Germany; and
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio; College of Nursing, The Ohio State University, Columbus, Ohio
| |
Collapse
|
37
|
Bozzi M, Sciandra F, Brancaccio A. Role of gelatinases in pathological and physiological processes involving the dystrophin–glycoprotein complex. Matrix Biol 2015; 44-46:130-7. [DOI: 10.1016/j.matbio.2015.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 12/16/2022]
|
38
|
Simvastatin reduces fibrosis and protects against muscle weakness after massive rotator cuff tear. J Shoulder Elbow Surg 2015; 24:280-7. [PMID: 25213828 PMCID: PMC4291297 DOI: 10.1016/j.jse.2014.06.048] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/14/2014] [Accepted: 06/27/2014] [Indexed: 02/01/2023]
Abstract
BACKGROUND Chronic rotator cuff tears are a common source of shoulder pain and disability, and patients with chronic cuff tears often have substantial weakness, fibrosis, inflammation, and fat accumulation. Identifying therapies to prevent the development of these pathologic processes will likely have a positive impact on clinical outcomes. Simvastatin is a drug with demonstrated anti-inflammatory and antifibrotic effects in many tissues but had not previously been studied in the context of rotator cuff tears. We hypothesized that after the induction of a massive supraspinatus tear, simvastatin would protect muscles from a loss of force production and fibrosis. METHODS We measured changes in muscle fiber contractility, histology, and biochemical markers of fibrosis and fatty infiltration in rats that received a full-thickness supraspinatus tear and were treated with either carrier alone or simvastatin. RESULTS Compared with vehicle-treated controls, simvastatin did not have an appreciable effect on muscle fiber size, but treatment did increase muscle fiber specific force by 20%. Simvastatin also reduced collagen accumulation by 50% but did not affect triglyceride content of muscles. Several favorable changes in the expression of genes and other markers of inflammation, fibrosis, and regeneration were also observed. CONCLUSIONS Simvastatin partially protected muscles from the weakness that occurs as a result of chronic rotator cuff tear. Fibrosis was also markedly reduced in simvastatin-treated animals. Whereas further studies are necessary, statin medication could potentially help improve outcomes for patients with rotator cuff tears.
Collapse
|
39
|
Hadler-Olsen E, Solli AI, Hafstad A, Winberg JO, Uhlin-Hansen L. Intracellular MMP-2 activity in skeletal muscle is associated with type II fibers. J Cell Physiol 2015; 230:160-9. [PMID: 24905939 DOI: 10.1002/jcp.24694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 05/29/2014] [Indexed: 02/03/2023]
Abstract
Matrix metalloproteinase 2 (MMP-2) is a proteolytic enzyme implicated in motility, differentiation, and regeneration of skeletal muscle fibers through processing of extracellular substrates. Although MMP-2 has been found to be localized intracellularly in cardiomyocytes where the enzyme is thought to contribute to post-ischemic loss of contractility, little is known about intracellular MMP-2 activity in skeletal muscle fibers. In the present study we demonstrate intracellular MMP-2 in normal skeletal muscle by immunohistochemical staining. Immunogold electron microscopic analyses indicated that the enzyme was concentrated in Z-lines of the sarcomers, in the nuclear membrane, and in mitochondria. By use of in situ zymography, we found that gelatinolytic activity in muscle fibers was co-localized with immunofluorecent staining for MMP-2. Staining for MMP-9, the other member of the gelatinase group of the MMPs, was negative. The broad-spectrum metalloprotease inhibitor EDTA and the selective gelatinase inhibitor CTT2, but not the cysteine inhibitor E64, strongly reduced the gelatinolytic activity. The intracellular gelatinolytic activity was much more prominent in fast twitch type II fibers than in slow twitch type I fibers, and there was a decrease in intracellular gelatinolytic activity and MMP-2 expression in muscles from mice exposed to high intensity interval training. Together our results indicate that MMP-2 is part of the intracellular proteolytic network in normal skeletal muscle, especially in fast twitch type II fibers. Further, the results suggest that intracellular MMP-2 in skeletal muscle fibers is active during normal homeostasis, and affected by the level of physical activity.
Collapse
Affiliation(s)
- Elin Hadler-Olsen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | | | | | | | | |
Collapse
|
40
|
Heier CR, Guerron AD, Korotcov A, Lin S, Gordish-Dressman H, Fricke S, Sze RW, Hoffman EP, Wang P, Nagaraju K. Non-invasive MRI and spectroscopy of mdx mice reveal temporal changes in dystrophic muscle imaging and in energy deficits. PLoS One 2014; 9:e112477. [PMID: 25390038 PMCID: PMC4229202 DOI: 10.1371/journal.pone.0112477] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/04/2014] [Indexed: 01/16/2023] Open
Abstract
In Duchenne muscular dystrophy (DMD), a genetic disruption of dystrophin protein expression results in repeated muscle injury and chronic inflammation. Magnetic resonance imaging shows promise as a surrogate outcome measure in both DMD and rehabilitation medicine that is capable of predicting clinical benefit years in advance of functional outcome measures. The mdx mouse reproduces the dystrophin deficiency that causes DMD and is routinely used for preclinical drug testing. There is a need to develop sensitive, non-invasive outcome measures in the mdx model that can be readily translatable to human clinical trials. Here we report the use of magnetic resonance imaging and spectroscopy techniques for the non-invasive monitoring of muscle damage in mdx mice. Using these techniques, we studied dystrophic mdx muscle in mice from 6 to 12 weeks of age, examining both the peak disease phase and natural recovery phase of the mdx disease course. T2 and fat-suppressed imaging revealed significant levels of tissue with elevated signal intensity in mdx hindlimb muscles at all ages; spectroscopy revealed a significant deficiency of energy metabolites in 6-week-old mdx mice. As the mdx mice progressed from the peak disease stage to the recovery stage of disease, each of these phenotypes was either eliminated or reduced, and the cross-sectional area of the mdx muscle was significantly increased when compared to that of wild-type mice. Histology indicates that hyper-intense MRI foci correspond to areas of dystrophic lesions containing inflammation as well as regenerating, degenerating and hypertrophied myofibers. Statistical sample size calculations provide several robust measures with the ability to detect intervention effects using small numbers of animals. These data establish a framework for further imaging or preclinical studies, and they support the development of MRI as a sensitive, non-invasive outcome measure for muscular dystrophy.
Collapse
Affiliation(s)
- Christopher R. Heier
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
| | - Alfredo D. Guerron
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
| | - Alexandru Korotcov
- Department of Radiology, Howard University College of Medicine, Washington, D.C., United States of America
| | - Stephen Lin
- Department of Radiology, Howard University College of Medicine, Washington, D.C., United States of America
| | - Heather Gordish-Dressman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, D.C., United States of America
| | - Stanley Fricke
- Department of Diagnostic Imaging and Radiology, Children's National Medical Center, Washington, D.C., United States of America
| | - Raymond W. Sze
- Department of Radiology, Children's National Medical Center, Washington, D.C., United States of America
| | - Eric P. Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, D.C., United States of America
| | - Paul Wang
- Department of Radiology, Howard University College of Medicine, Washington, D.C., United States of America
- Department of Electrical Engineering, Fu Jen Catholic University, Taipei, Taiwan
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
41
|
Bobadilla M, Sáinz N, Rodriguez JA, Abizanda G, Orbe J, de Martino A, García Verdugo JM, Páramo JA, Prósper F, Pérez-Ruiz A. MMP-10 is required for efficient muscle regeneration in mouse models of injury and muscular dystrophy. Stem Cells 2014; 32:447-61. [PMID: 24123596 DOI: 10.1002/stem.1553] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/20/2013] [Accepted: 08/23/2013] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs), a family of endopeptidases that are involved in the degradation of extracellular matrix components, have been implicated in skeletal muscle regeneration. Among the MMPs, MMP-2 and MMP-9 are upregulated in Duchenne muscular dystrophy (DMD), a fatal X-linked muscle disorder. However, inhibition or overexpression of specific MMPs in a mouse model of DMD (mdx) has yielded mixed results regarding disease progression, depending on the MMP studied. Here, we have examined the role of MMP-10 in muscle regeneration during injury and muscular dystrophy. We found that skeletal muscle increases MMP-10 protein expression in response to damage (notexin) or disease (mdx mice), suggesting its role in muscle regeneration. In addition, we found that MMP-10-deficient muscles displayed impaired recruitment of endothelial cells, reduced levels of extracellular matrix proteins, diminished collagen deposition, and decreased fiber size, which collectively contributed to delayed muscle regeneration after injury. Also, MMP-10 knockout in mdx mice led to a deteriorated dystrophic phenotype. Moreover, MMP-10 mRNA silencing in injured muscles (wild-type and mdx) reduced muscle regeneration, while addition of recombinant human MMP-10 accelerated muscle repair, suggesting that MMP-10 is required for efficient muscle regeneration. Furthermore, our data suggest that MMP-10-mediated muscle repair is associated with VEGF/Akt signaling. Thus, our findings indicate that MMP-10 is critical for skeletal muscle maintenance and regeneration during injury and disease.
Collapse
Affiliation(s)
- Míriam Bobadilla
- Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pereira JA, Matsumura CY, Minatel E, Marques MJ, Santo Neto H. Understanding the beneficial effects of doxycycline on the dystrophic phenotype of the mdx
mouse. Muscle Nerve 2014; 50:283-6. [DOI: 10.1002/mus.24177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Juliano Alves Pereira
- Departamento de Biologia Estrutural e Funcional; Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP); Campinas São Paulo 13083-865 Brazil
| | - Cintia Yuri Matsumura
- Departamento de Biologia Estrutural e Funcional; Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP); Campinas São Paulo 13083-865 Brazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional; Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP); Campinas São Paulo 13083-865 Brazil
| | - Maria Julia Marques
- Departamento de Biologia Estrutural e Funcional; Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP); Campinas São Paulo 13083-865 Brazil
| | - Humberto Santo Neto
- Departamento de Biologia Estrutural e Funcional; Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP); Campinas São Paulo 13083-865 Brazil
| |
Collapse
|
43
|
Ogura Y, Tajrishi MM, Sato S, Hindi SM, Kumar A. Therapeutic potential of matrix metalloproteinases in Duchenne muscular dystrophy. Front Cell Dev Biol 2014; 2:11. [PMID: 25364719 PMCID: PMC4207008 DOI: 10.3389/fcell.2014.00011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/13/2014] [Indexed: 12/31/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are secreted proteinases that have physiologic roles in degradation and remodeling of extracellular matrix (ECM) in almost all tissues. However, their excessive production in disease conditions leads to many pathological features including tissue breakdown, inflammation, cell death, and fibrosis. Duchenne Muscular dystrophy (DMD) is a devastating genetic muscle disorder caused by partial or complete loss of cytoskeletal protein dystrophin. Progressive muscle wasting in DMD is accompanied by myofiber necrosis followed by cycles of regeneration and degeneration and inflammation that eventually result in replacement of myofiber by connective and adipose tissues. Emerging evidence suggests that gene expression and the activity of various MMPs are aberrantly regulated in muscle biopsies from DMD patients and in skeletal muscle of animal models of DMD. Moreover, a few studies employing genetic mouse models have revealed that different MMPs play distinct roles in disease progression in DMD. Modulation of the activity of MMPs improves myofiber regeneration and enhances the efficacy of transplantation and engraftment of muscle progenitor cells in dystrophic muscle in mouse models of DMD. Furthermore, recent reports also suggest that some MMPs especially MMP-9 can serve as a biomarker for diagnosis and prognosis of DMD. In this article, we provide a succinct overview of the regulation of various MMPs and their therapeutic importance in DMD.
Collapse
Affiliation(s)
- Yuji Ogura
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Marjan M Tajrishi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Shuichi Sato
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| |
Collapse
|
44
|
Matrix metalloproteinase-9 inhibition improves proliferation and engraftment of myogenic cells in dystrophic muscle of mdx mice. PLoS One 2013; 8:e72121. [PMID: 23977226 PMCID: PMC3744489 DOI: 10.1371/journal.pone.0072121] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/09/2013] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) caused by loss of cytoskeletal protein dystrophin is a devastating disorder of skeletal muscle. Primary deficiency of dystrophin leads to several secondary pathological changes including fiber degeneration and regeneration, extracellular matrix breakdown, inflammation, and fibrosis. Matrix metalloproteinases (MMPs) are a group of extracellular proteases that are involved in tissue remodeling, inflammation, and development of interstitial fibrosis in many disease states. We have recently reported that the inhibition of MMP-9 improves myopathy and augments myofiber regeneration in mdx mice (a mouse model of DMD). However, the mechanisms by which MMP-9 regulates disease progression in mdx mice remain less understood. In this report, we demonstrate that the inhibition of MMP-9 augments the proliferation of satellite cells in dystrophic muscle. MMP-9 inhibition also causes significant reduction in percentage of M1 macrophages with concomitant increase in the proportion of promyogenic M2 macrophages in mdx mice. Moreover, inhibition of MMP-9 increases the expression of Notch ligands and receptors, and Notch target genes in skeletal muscle of mdx mice. Furthermore, our results show that while MMP-9 inhibition augments the expression of components of canonical Wnt signaling, it reduces the expression of genes whose products are involved in activation of non-canonical Wnt signaling in mdx mice. Finally, the inhibition of MMP-9 was found to dramatically improve the engraftment of transplanted myoblasts in skeletal muscle of mdx mice. Collectively, our study suggests that the inhibition of MMP-9 is a promising approach to stimulate myofiber regeneration and improving engraftment of muscle progenitor cells in dystrophic muscle.
Collapse
|
45
|
Bozzi M, Di Stasio E, Scaglione GL, Desiderio C, Martelli C, Giardina B, Sciandra F, Brancaccio A. Probing the stability of the "naked" mucin-like domain of human α-dystroglycan. BMC BIOCHEMISTRY 2013; 14:15. [PMID: 23815856 PMCID: PMC3704865 DOI: 10.1186/1471-2091-14-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 06/20/2013] [Indexed: 11/10/2022]
Abstract
Background α-Dystroglycan (α-DG) is heavily glycosylated within its central mucin-like domain. The glycosylation shell of α-dystroglycan is known to largely influence its functional properties toward extracellular ligands. The structural features of this α-dystroglycan domain have been poorly studied so far. For the first time, we have attempted a recombinant expression approach in E. coli cells, in order to analyze by biochemical and biophysical techniques this important domain of the α-dystroglycan core protein. Results We expressed the recombinant mucin-like domain of human α-dystroglycan in E. coli cells, and purified it as a soluble peptide of 174 aa. A cleavage event, that progressively emerges under repeated cycles of freeze/thaw, occurs at the carboxy side of Arg461, liberating a 151 aa fragment as revealed by mass spectrometry analysis. The mucin-like peptide lacks any particular fold, as confirmed by its hydrodynamic properties and its fluorescence behavior under guanidine hydrochloride denaturation. Dynamic light scattering has been used to demonstrate that this mucin-like peptide is arranged in a conformation that is prone to aggregation at room temperature, with a melting temperature of ~40°C, which indicates a pronounced instability. Such a conclusion has been corroborated by trypsin limited proteolysis, upon which the protein has been fully degraded in less than 60 min. Conclusions Our analysis indirectly confirms the idea that the mucin-like domain of α-dystroglycan needs to be extensively glycosylated in order to reach a stable conformation. The absence/reduction of glycosylation by itself may greatly reduce the stability of the dystroglycan complex. Although an altered pattern of α-dystroglycan O-mannosylation, that is not significantly changing its overall glycosylation fraction, represents the primary molecular clue behind currently known dystroglycanopathies, it cannot be ruled out that still unidentified forms of αDG-related dystrophy might originate by a more substantial reduction of α-dystroglycan glycosylation and by its consequent destabilization.
Collapse
|
46
|
Shin J, Tajrishi MM, Ogura Y, Kumar A. Wasting mechanisms in muscular dystrophy. Int J Biochem Cell Biol 2013; 45:2266-79. [PMID: 23669245 DOI: 10.1016/j.biocel.2013.05.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022]
Abstract
Muscular dystrophy is a group of more than 30 different clinical genetic disorders that are characterized by progressive skeletal muscle wasting and degeneration. Primary deficiency of specific extracellular matrix, sarcoplasmic, cytoskeletal, or nuclear membrane protein results in several secondary changes such as sarcolemmal instability, calcium influx, fiber necrosis, oxidative stress, inflammatory response, breakdown of extracellular matrix, and eventually fibrosis which leads to loss of ambulance and cardiac and respiratory failure. A number of molecular processes have now been identified which hasten disease progression in human patients and animal models of muscular dystrophy. Accumulating evidence further suggests that aberrant activation of several signaling pathways aggravate pathological cascades in dystrophic muscle. Although replacement of defective gene with wild-type is paramount to cure, management of secondary pathological changes has enormous potential to improving the quality of life and extending lifespan of muscular dystrophy patients. In this article, we have reviewed major cellular and molecular mechanisms leading to muscle wasting in muscular dystrophy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Jonghyun Shin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
47
|
Nedergaard A, Karsdal MA, Sun S, Henriksen K. Serological muscle loss biomarkers: an overview of current concepts and future possibilities. J Cachexia Sarcopenia Muscle 2013; 4:1-17. [PMID: 22996343 PMCID: PMC3581612 DOI: 10.1007/s13539-012-0086-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/21/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The skeletal muscle mass is the largest organ in the healthy body, comprising 30-40 % of the body weight of an adult man. It confers protection from trauma, locomotion, ventilation, and it represents a "sink" in glucose metabolism and a reservoir of amino acids to other tissues such as the brain and blood cells. Naturally, loss of muscle has dire consequences for health as well as functionality. Muscle loss is a natural consequence of especially aging, inactivity, and their associated metabolic dysfunction, but it is strongly accelerated in critical illness such as organ failure, sepsis, or cancer. Whether this muscle loss is considered a primary or secondary condition, it is known that muscle loss is a symptom that predicts morbidity and mortality and one that is known to impact quality of life and independence. Therefore, monitoring of muscle mass is relevant in a number of pathologies as well as in clinical trials as measures of efficacy as well as safety. METHODS AND RESULTS Existing biomarkers of muscle mass or muscle loss have shown to be either too unreliable or too impractical in relation to the perceived clinical benefit to reach regular clinical research or use. We suggest serological neoepitope biomarkers as a possible technology to address some of these problems. Blood biomarkers of this kind have previously been shown to respond with high sensitivity and shorter time to minimum significant change than available biomarkers of muscle mass. We provide brief reviews of existing muscle mass or function biomarker technologies, muscle protein biology, and existing neoepitope biomarkers and proceed to present tentative recommendations on how to select and detect neoepitope biomarkers. CONCLUSION We suggest that serological peptide biomarkers whose tissue and pathology specificity are derived from post-translational modification of proteins in tissues of interest, presenting so-called neoepitopes, represents an exciting candidate technology to fill out an empty niche in biomarker technology.
Collapse
|
48
|
Alameddine HS. Matrix metalloproteinases in skeletal muscles: Friends or foes? Neurobiol Dis 2012; 48:508-18. [DOI: 10.1016/j.nbd.2012.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/28/2012] [Accepted: 07/25/2012] [Indexed: 12/13/2022] Open
|
49
|
Taniguti APT, Matsumura CÍY, Rodrigues-Simioni LÉ, Neto HS, Marques MJ. Suramin affects metalloproteinase-9 activity and increases beta-dystroglycan levels in the diaphragm of the dystrophin-deficientmdxMOUSE. Muscle Nerve 2012; 46:810-3. [DOI: 10.1002/mus.23468] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
50
|
Reciprocal interaction between TRAF6 and notch signaling regulates adult myofiber regeneration upon injury. Mol Cell Biol 2012; 32:4833-45. [PMID: 23028045 DOI: 10.1128/mcb.00717-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle is a postmitotic tissue that repairs and regenerates through activation of a population of stem-cell-like satellite cells. However, signaling mechanisms governing adult skeletal muscle regeneration remain less understood. In the present study, we have investigated the role of tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), an adaptor protein involved in receptor-mediated activation of multiple signaling pathways in regeneration of adult myofibers. Skeletal muscle-specific depletion of TRAF6 in mice (TRAF6(mko)) improved regeneration of myofibers upon injury with a concomitant increase in the number of satellite cells and activation of the Notch signaling pathway. Ex vivo cultures of TRAF6(mko) myofiber explants demonstrated an increase in the proliferative capacity of myofiber-associated satellite cells accompanied by an upregulation of Notch ligands. Deletion of TRAF6 also inhibited the activity of transcription factor NF-κB and the expression of inflammatory cytokines and augmented the M2c macrophage phenotype in injured muscle tissues. Collectively, our study demonstrates that specific inhibition of TRAF6 improves satellite cell activation and skeletal muscle regeneration through upregulation of Notch signaling and reducing the inflammatory repertoire.
Collapse
|