1
|
Rossiaud L, Miagoux Q, Benabides M, Reiss O, Jauze L, Jarrige M, Polvèche H, Malfatti E, Laforêt P, Ronzitti G, Nissan X, Hoch L. Galectin-3: a novel biomarker of glycogen storage disease type III. Cell Death Discov 2025; 11:173. [PMID: 40229243 PMCID: PMC11997124 DOI: 10.1038/s41420-025-02452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/06/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
Glycogen storage disease type III (GSDIII) is a rare genetic disorder leading to abnormal glycogen storage in the liver and skeletal muscle. In this study, we conducted a comparative gene expression analysis of several in vitro and in vivo models and identified galectin-3 as a potential biomarker of the disease. Interestingly, we also observed a significant decrease in galectin-3 expression in mice treated with an AAV gene therapy. Finally, galectin-3 expression was studied in muscle biopsies of GSDIII patients, confirming its increase in patient tissue. Beyond the identification of this novel biomarker, our study offers a new perspective for future therapeutic developments.
Collapse
Affiliation(s)
- Lucille Rossiaud
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research Unit UMR_S951, Evry, France
| | - Quentin Miagoux
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Manon Benabides
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Océane Reiss
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Louisa Jauze
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research Unit UMR_S951, Evry, France
| | - Margot Jarrige
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Hélène Polvèche
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Edoardo Malfatti
- Reference Center for Neuromuscular Disorders, APHP Henri Mondor University Hospital, Créteil, France
- Université Paris Est Créteil, Inserm, U955, IMRB, Créteil, France
| | - Pascal Laforêt
- Neurology Department, Nord/Est/Île-de-France Neuromuscular Reference Center, FHU PHENIX, AP-HP, Raymond-Poincaré Hospital, Garches, France
| | - Giuseppe Ronzitti
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research Unit UMR_S951, Evry, France
| | - Xavier Nissan
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France
- IStem, CECS, Corbeil-Essonnes, France
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France
| | - Lucile Hoch
- Université Paris-Saclay, Université d'Evry, Inserm, IStem, UMR861, Corbeil-Essonnes, France.
- IStem, CECS, Corbeil-Essonnes, France.
- IStem, CECS, The Research and Innovation Team, Corbeil-Essonnes, France.
| |
Collapse
|
2
|
de Menezes YKT, Lee J, Cheng-Zhang JQ, Johnson MA, Ranatunga RN, Kemaladewi DU. Targeting Galectin-3 to modulate inflammation in LAMA2-deficient congenital muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642905. [PMID: 40161708 PMCID: PMC11952532 DOI: 10.1101/2025.03.12.642905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
LAMA2-deficient congenital muscular dystrophy (LAMA2-CMD) is a severe neuromuscular disorder characterized by muscle degeneration, chronic inflammation, and fibrosis. While inflammation is one the hallmarks of LAMA2-CMD, the immune cell composition in laminin-deficient muscles remains understudied. Consequently, targeted pharmacological intervention to reduce inflammation remains underexplored. Here, we characterized the immune landscape in the dyW mouse model of LAMA2-CMD using RNA sequencing and flow cytometry. Transcriptomic analysis of dyW quadriceps femoris muscle identified 2,143 differentially expressed genes, with most upregulated genes linked to immune-related pathways. Lgals3 (Galectin-3) was significantly upregulated and identified as a key upstream regulator of the immune-related pathways. Flow cytometry revealed elevated leukocyte (CD45⁺) infiltration, with macrophages as the predominant population. Pro-inflammatory (M1) macrophages were increased, whereas anti-inflammatory (M2) macrophages remained low, indicating persistent and unresolved inflammation. Notably, Galectin-3 + macrophages were significantly enriched, suggesting that Galectin-3 drives inflammation in LAMA2-CMD. Treatment of dyW mice with TD-139, a Galectin-3 inhibitor, reduced leukocyte infiltration, decreased Galectin-3 + macrophages, and shifted macrophage polarization toward an M2 anti-inflammatory profile. RNA sequencing of TD-139-treated dyW muscles showed upregulation of muscle contraction pathways and downregulation of fibrosis-related genes. These findings highlight Galectin-3 + macrophages as key contributors to LAMA2-CMD pathophysiology and support further exploration of TD-139 as a potential therapeutic strategy for LAMA2-CMD and other dystrophic conditions driven by chronic inflammation.
Collapse
|
3
|
Redhead C, Taye N, Hubmacher D. En route towards a personalized medicine approach: Innovative therapeutic modalities for connective tissue disorders. Matrix Biol 2023; 122:46-54. [PMID: 37657665 PMCID: PMC10529529 DOI: 10.1016/j.matbio.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/03/2023]
Abstract
Connective tissue disorders can be caused by pathogenic variants (mutations) in genes encoding extracellular matrix (ECM) proteins. Such disorders typically manifest during development or postnatal growth and result in significant morbidity and mortality. The development of curative treatments for connective tissue disorders is hampered in part by the inability of many mature connective tissues to efficiently regenerate. To be most effective, therapeutic strategies designed to preserve or restore tissue function will likely need to be initiated during phases of significant endogenous connective tissue remodeling and organ sculpting postnatally and directly target the underlying ECM protein mutations. With recent advances in whole exome sequencing, in-vitro and in-vivo disease modeling, and the development of mutation-specific molecular therapeutic modalities, it is now feasible to directly correct disease-causing mutations underlying connective tissue disorders and ameliorate their pathogenic consequences. These technological advances may lead to potentially curative personalized medicine approaches for connective tissue disorders that have previously been considered incurable. In this review, we highlight innovative therapeutic modalities including gene replacement, exon skipping, DNA/mRNA editing, and pharmacological approaches that were used to preserve or restore tissue function in the context of connective tissue disorders. Inherent to a successful application of these approaches is the need to deepen the understanding of mechanisms that regulate ECM formation and homeostasis, and to decipher how individual mutations in ECM proteins compromise ECM and connective tissue development and function.
Collapse
Affiliation(s)
- Charlene Redhead
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nandaraj Taye
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dirk Hubmacher
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Oliveira-Santos A, Dagda M, Wittmann J, Smalley R, Burkin DJ. Vemurafenib improves muscle histopathology in a mouse model of LAMA2-related congenital muscular dystrophy. Dis Model Mech 2023; 16:dmm049916. [PMID: 37021539 PMCID: PMC10184677 DOI: 10.1242/dmm.049916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a neuromuscular disease affecting around 1-9 in 1,000,000 children. LAMA2-CMD is caused by mutations in the LAMA2 gene resulting in the loss of laminin-211/221 heterotrimers in skeletal muscle. LAMA2-CMD patients exhibit severe hypotonia and progressive muscle weakness. Currently, there is no effective treatment for LAMA2-CMD and patients die prematurely. The loss of laminin-α2 results in muscle degeneration, defective muscle repair and dysregulation of multiple signaling pathways. Signaling pathways that regulate muscle metabolism, survival and fibrosis have been shown to be dysregulated in LAMA2-CMD. As vemurafenib is a US Food and Drug Administration (FDA)-approved serine/threonine kinase inhibitor, we investigated whether vemurafenib could restore some of the serine/threonine kinase-related signaling pathways and prevent disease progression in the dyW-/- mouse model of LAMA2-CMD. Our results show that vemurafenib reduced muscle fibrosis, increased myofiber size and reduced the percentage of fibers with centrally located nuclei in dyW-/- mouse hindlimbs. These studies show that treatment with vemurafenib restored the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways in skeletal muscle. Together, our results indicate that vemurafenib partially improves histopathology but does not improve muscle function in a mouse model of LAMA2-CMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Jennifer Wittmann
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Robert Smalley
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Dean J. Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| |
Collapse
|
5
|
Bugiardini E, Nunes AM, Oliveira‐Santos A, Dagda M, Fontelonga TM, Barraza‐Flores P, Pittman AM, Morrow JM, Parton M, Houlden H, Elliott PM, Syrris P, Maas RP, Akhtar MM, Küsters B, Raaphorst J, Schouten M, Kamsteeg E, van Engelen B, Hanna MG, Phadke R, Lopes LR, Matthews E, Burkin DJ. Integrin α7 Mutations Are Associated With Adult-Onset Cardiac Dysfunction in Humans and Mice. J Am Heart Assoc 2022; 11:e026494. [PMID: 36444867 PMCID: PMC9851448 DOI: 10.1161/jaha.122.026494] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022]
Abstract
Background Integrin α7β1 is a major laminin receptor in skeletal and cardiac muscle. In skeletal muscle, integrin α7β1 plays an important role during muscle development and has been described as an important modifier of skeletal muscle diseases. The integrin α7β1 is also highly expressed in the heart, but its precise role in cardiac function is unknown. Mutations in the integrin α7 gene (ITGA7) have been reported in children with congenital myopathy. Methods and Results In this study, we described skeletal and cardiac muscle pathology in Itga7-/- mice and 5 patients from 2 unrelated families with ITGA7 mutations. Proband in family 1 presented a homozygous c.806_818del [p.S269fs] variant, and proband in family 2 was identified with 2 intron variants in the ITGA7 gene. The complete absence of the integrin α7 protein in muscle supports the ITGA7 mutations are pathogenic. We performed electrocardiography, echocardiography, or cardiac magnetic resonance imaging, and histological biopsy analyses in patients with ITGA7 deficiency and Itga7-/- mice. The patients exhibited cardiac dysrhythmia and dysfunction from the third decade of life and late-onset respiratory insufficiency, but with relatively mild limb muscle involvement. Mice demonstrated corresponding abnormalities in cardiac conduction and contraction as well as diaphragm muscle fibrosis. Conclusions Our data suggest that loss of integrin α7 causes a novel form of adult-onset cardiac dysfunction indicating a critical role for the integrin α7β1 in normal cardiac function and highlights the need for long-term cardiac monitoring in patients with ITGA7-related congenital myopathy.
Collapse
Affiliation(s)
- Enrico Bugiardini
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Andreia M. Nunes
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Ariany Oliveira‐Santos
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Marisela Dagda
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Tatiana M. Fontelonga
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Pamela Barraza‐Flores
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| | - Alan M. Pittman
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
- St George’sUniversity of LondonLondonUnited Kingdom
| | - Jasper M. Morrow
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Matthew Parton
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Henry Houlden
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Perry M. Elliott
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Petros Syrris
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Roderick P. Maas
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Mohammed M. Akhtar
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Benno Küsters
- Department of PathologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Joost Raaphorst
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Meyke Schouten
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Erik‐Jan Kamsteeg
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - Baziel van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and BehaviourRadboud University Medical CenterNijmegenThe Netherlands
| | - Michael G. Hanna
- Queen Square Centre for Neuromuscular DiseasesQueen Square Institute of Neurology, UCL and National Hospital for Neurology and NeurosurgeryLondonUnited Kingdom
| | - Rahul Phadke
- Division of NeuropathologyUCL Institute of NeurologyLondonUnited Kingdom
- Dubowitz Neuromuscular Centre, MRC Centre for Neuromuscular DiseasesUCL Great Ormond Street Institute of Child HealthLondonUnited Kingdom
| | - Luis R. Lopes
- Barts Heart Centre, Barts Health NHS TrustLondonUnited Kingdom
- Centre for Heart Muscle DiseaseInstitute of Cardiovascular Science, University College LondonLondonUnited Kingdom
| | - Emma Matthews
- The Atkinson Morley Neuromuscular Centre and Regional Neurosciences CentreSt George’s University Hospitals NHS Foundation TrustLondonUnited Kingdom
- Molecular and Clinical Sciences Research Institute, St George’s University of LondonLondonUnited Kingdom
| | - Dean J. Burkin
- Department of PharmacologyUniversity of Nevada Reno, School of Medicine, Center for Molecular MedicineRenoNV
| |
Collapse
|
6
|
Ba MA, Aiyuk A, Hernández K, Evasovic JM, Wuebbles RD, Burkin DJ, Singer CA. Transgenic overexpression of α7 integrin in smooth muscle attenuates allergen-induced airway inflammation in a murine model of asthma. FASEB Bioadv 2022; 4:724-740. [PMID: 36349295 PMCID: PMC9635010 DOI: 10.1096/fba.2022-00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 12/03/2022] Open
Abstract
Asthma is a chronic inflammatory disorder of the lower airways characterized by modulation of airway smooth muscle (ASM) function. Infiltration of smooth muscle by inflammatory mediators is partially regulated by transmembrane integrins and the major smooth muscle laminin receptor α7β1 integrin plays a critical role in the maintenance of ASM phenotype. The goal of the current study was to investigate the role of α7 integrin in asthma using smooth muscle-specific α7 integrin transgenic mice (TgSM-Itgα7) using both acute and chronic OVA sensitization and challenge protocols that mimic mild to severe asthmatic phenotypes. Transgenic over-expression of the α7 integrin in smooth muscle resulted in a significant decrease in airway resistance relative to controls, reduced the total number of inflammatory cells and substantially inhibited the production of crucial Th2 and Th17 cytokines in airways. This was accompanied by decreased secretion of various inflammatory chemokines such as eotaxin/CCL11, KC/CXCL3, MCP-1/CCL2, and MIP-1β/CCL4. Additionally, α7 integrin overexpression significantly decreased ERK1/2 phosphorylation in the lungs of TgSM-Itgα7 mice and affected proliferative, contractile, and inflammatory downstream effectors of ERK1/2 that drive smooth muscle phenotype in the lung. Taken together, these results support the hypothesis that enhanced expression of α7 integrin in vivo inhibits allergic inflammation and airway resistance. Moreover, we identify ERK1/2 as a potential target by which α7 integrin signals to regulate airway inflammation. We conclude that identification of therapeutics targeting an increase in smooth muscle α7 integrin expression could serve as a potential novel treatment for asthma.
Collapse
Affiliation(s)
- Mariam A. Ba
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Annemarie Aiyuk
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Karla Hernández
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Jon M. Evasovic
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Ryan D. Wuebbles
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Dean J. Burkin
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| | - Cherie A. Singer
- Department of PharmacologyUniversity of Nevada School of MedicineRenoNevadaUSA
| |
Collapse
|
7
|
An artificial LAMA2-GelMA hydrogel microenvironment for the development of pancreatic endocrine progenitors. Biomaterials 2022; 291:121882. [DOI: 10.1016/j.biomaterials.2022.121882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/15/2022] [Accepted: 10/23/2022] [Indexed: 11/21/2022]
|
8
|
Orkin JD, Montague MJ, Tejada-Martinez D, de Manuel M, Del Campo J, Cheves Hernandez S, Di Fiore A, Fontsere C, Hodgson JA, Janiak MC, Kuderna LFK, Lizano E, Martin MP, Niimura Y, Perry GH, Valverde CS, Tang J, Warren WC, de Magalhães JP, Kawamura S, Marquès-Bonet T, Krawetz R, Melin AD. The genomics of ecological flexibility, large brains, and long lives in capuchin monkeys revealed with fecalFACS. Proc Natl Acad Sci U S A 2021; 118:e2010632118. [PMID: 33574059 PMCID: PMC7896301 DOI: 10.1073/pnas.2010632118] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ecological flexibility, extended lifespans, and large brains have long intrigued evolutionary biologists, and comparative genomics offers an efficient and effective tool for generating new insights into the evolution of such traits. Studies of capuchin monkeys are particularly well situated to shed light on the selective pressures and genetic underpinnings of local adaptation to diverse habitats, longevity, and brain development. Distributed widely across Central and South America, they are inventive and extractive foragers, known for their sensorimotor intelligence. Capuchins have among the largest relative brain size of any monkey and a lifespan that exceeds 50 y, despite their small (3 to 5 kg) body size. We assemble and annotate a de novo reference genome for Cebus imitator Through high-depth sequencing of DNA derived from blood, various tissues, and feces via fluorescence-activated cell sorting (fecalFACS) to isolate monkey epithelial cells, we compared genomes of capuchin populations from tropical dry forests and lowland rainforests and identified population divergence in genes involved in water balance, kidney function, and metabolism. Through a comparative genomics approach spanning a wide diversity of mammals, we identified genes under positive selection associated with longevity and brain development. Additionally, we provide a technological advancement in the use of noninvasive genomics for studies of free-ranging mammals. Our intra- and interspecific comparative study of capuchin genomics provides insights into processes underlying local adaptation to diverse and physiologically challenging environments, as well as the molecular basis of brain evolution and longevity.
Collapse
Affiliation(s)
- Joseph D Orkin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
| | - Michael J Montague
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19146
| | - Daniela Tejada-Martinez
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
- Doctorado en Ciencias mención Ecología y Evolución, Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Marc de Manuel
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Javier Del Campo
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | | | - Anthony Di Fiore
- Department of Anthropology and Primate Molecular Ecology and Evolution Laboratory, University of Texas at Austin, Austin, TX 78712
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, 170901 Cumbayá, Ecuador
| | - Claudia Fontsere
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Jason A Hodgson
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, United Kingdom
| | - Mareike C Janiak
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
- School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, United Kingdom
| | - Lukas F K Kuderna
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Esther Lizano
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Maria Pia Martin
- Kids Saving the Rainforest Wildlife Rescue Center, 60601 Quepos, Costa Rica
| | - Yoshihito Niimura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - George H Perry
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | | | - Jia Tang
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Wesley C Warren
- Division of Animal Sciences, School of Medicine, University of Missouri, Columbia, MO 65211
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Shoji Kawamura
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562 Chiba, Japan
| | - Tomàs Marquès-Bonet
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, 08010 Barcelona, Spain
- Centro Nacional de Análisis Genómico-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T38 6A8, Canada
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T38 6A8, Canada
| |
Collapse
|
9
|
Packer D, Martin PT. Micro-laminin gene therapy can function as an inhibitor of muscle disease in the dy W mouse model of MDC1A. Mol Ther Methods Clin Dev 2021; 21:274-287. [PMID: 33869655 PMCID: PMC8026908 DOI: 10.1016/j.omtm.2021.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/03/2021] [Indexed: 02/04/2023]
Abstract
Gene replacement for laminin-α2-deficient congenital muscular dystrophy 1A (MDC1A) is currently not possible using a single adeno-associated virus (AAV) vector due to the large size of the LAMA2 gene. LAMA2 encodes laminin-α2, a subunit of the trimeric laminin-211 extracellular matrix (ECM) protein that is the predominant laminin expressed in skeletal muscle. LAMA2 expression stabilizes skeletal muscle, in part by binding membrane receptors via its five globular (G) domains. We created a small, AAV-deliverable, micro-laminin gene therapy that expresses these G1-5 domains, LAMA2(G1-5), to test their therapeutic efficacy in the dyW mouse model for MDC1A. We also fused the heparin-binding (HB) domain from HB epidermal growth factor-like growth factor (HB-EGF) to LAMA2(G1-5) to test whether this would increase muscle ECM expression. dyW mice treated intravenously with rAAV9.CMV.HB-LAMA2(G1-5) showed increased muscle ECM expression of transgenic protein relative to mice treated with rAAV9.CMV.LAMA2(G1-5) and showed improved weight-normalized forelimb grip strength relative to untreated dyW mice. Additionally, dyW muscle fibers expressing either micro-laminin protein showed some measures of reduced pathology, although levels of muscle cell apoptosis and inflammation were not decreased. Although systemic expression of rAAV9.CMV.HB-LAMA2(G1-5) did not inhibit all disease phenotypes, these studies demonstrate the feasibility of using a micro-laminin gene therapy strategy to deliver gene replacement for MDC1A.
Collapse
Affiliation(s)
- Davin Packer
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
- Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Paul T. Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Corresponding author Paul T. Martin, Center for Gene Therapy, Abigail Wexner Research Institute, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43209, USA.
| |
Collapse
|
10
|
Nosacka RL, Delitto AE, Delitto D, Patel R, Judge SM, Trevino JG, Judge AR. Distinct cachexia profiles in response to human pancreatic tumours in mouse limb and respiratory muscle. J Cachexia Sarcopenia Muscle 2020; 11:820-837. [PMID: 32039571 PMCID: PMC7296265 DOI: 10.1002/jcsm.12550] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cancer cachexia is a life-threatening metabolic syndrome that causes significant loss of skeletal muscle mass and significantly increases mortality in cancer patients. Currently, there is an urgent need for better understanding of the molecular pathophysiology of this disease so that effective therapies can be developed. The majority of pre-clinical studies evaluating skeletal muscle's response to cancer have focused on one or two pre-clinical models, and almost all have focused specifically on limb muscles. In the current study, we reveal key differences in the histology and transcriptomic signatures of a limb muscle and a respiratory muscle in orthotopic pancreatic cancer patient-derived xenograft (PDX) mice. METHODS To create four cohorts of PDX mice evaluated in this study, tumours resected from four pancreatic ductal adenocarcinoma patients were portioned and attached to the pancreas of immunodeficient NSG mice. RESULTS Body weight, muscle mass, and fat mass were significantly decreased in each PDX line. Histological assessment of cryosections taken from the tibialis anterior (TA) and diaphragm (DIA) revealed differential effects of tumour burden on their morphology. Subsequent genome-wide microarray analysis on TA and DIA also revealed key differences between their transcriptomes in response to cancer. Genes up-regulated in the DIA were enriched for extracellular matrix protein-encoding genes and genes related to the inflammatory response, while down-regulated genes were enriched for mitochondria related protein-encoding genes. Conversely, the TA showed up-regulation of canonical atrophy-associated pathways such as ubiquitin-mediated protein degradation and apoptosis, and down-regulation of genes encoding extracellular matrix proteins. CONCLUSIONS These data suggest that distinct biological processes may account for wasting in different skeletal muscles in response to the same tumour burden. Further investigation into these differences will be critical for the future development of effective clinical strategies to counter cancer cachexia.
Collapse
Affiliation(s)
- Rachel L Nosacka
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Andrea E Delitto
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Dan Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, USA
| | - Rohan Patel
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, USA
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, USA
| |
Collapse
|
11
|
Gawlik KI, Durbeej M. A Family of Laminin α2 Chain-Deficient Mouse Mutants: Advancing the Research on LAMA2-CMD. Front Mol Neurosci 2020; 13:59. [PMID: 32457577 PMCID: PMC7188397 DOI: 10.3389/fnmol.2020.00059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
The research on laminin α2 chain-deficient congenital muscular dystrophy (LAMA2-CMD) advanced rapidly in the last few decades, largely due to availability of good mouse models for the disease and a strong interest in preclinical studies from scientists all over the world. These mouse models continue to provide a solid platform for understanding the LAMA2-CMD pathology. In addition, they enable researchers to test laborious, necessary routines, but also the most creative scientific approaches in order to design therapy for this devastating disorder. In this review we present animals belonging to the laminin α2 chain-deficient “dy/dy” mouse family (dy/dy, dy2J/dy2J, dy3K/dy3K, dyW/dyW, et al.) and a summary of the scientific progress they facilitated. We also raise a few questions that need to be addressed in order to maximize the usefulness of laminin α2 murine mutants and to further advance the LAMA2-CMD studies. We believe that research opportunities offered by the mouse models for LAMA2-CMD will continuously support our efforts to find a treatment for the disease.
Collapse
Affiliation(s)
- Kinga I Gawlik
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Madeleine Durbeej
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Barraza-Flores P, Bates CR, Oliveira-Santos A, Burkin DJ. Laminin and Integrin in LAMA2-Related Congenital Muscular Dystrophy: From Disease to Therapeutics. Front Mol Neurosci 2020; 13:1. [PMID: 32116540 PMCID: PMC7026472 DOI: 10.3389/fnmol.2020.00001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a devastating neuromuscular disease caused by mutations in the LAMA2 gene. These mutations result in the complete absence or truncated expression of the laminin-α2 chain. The α2-chain is a major component of the laminin-211 and laminin-221 isoforms, the predominant laminin isoforms in healthy adult skeletal muscle. Mutations in this chain result in progressive skeletal muscle degeneration as early as neonatally. Laminin-211/221 is a ligand for muscle cell receptors integrin-α7β1 and α-dystroglycan. LAMA2 mutations are correlated with integrin-α7β1 disruption in skeletal muscle. In this review, we will summarize laminin-211/221 interactions with integrin-α7β1 in LAMA2-CMD muscle. Additionally, we will summarize recent developments using upregulation of laminin-111 in the sarcolemma of laminin-α2-deficient muscle. We will discuss potential mechanisms of action by which laminin-111 is able to prevent myopathy. These published studies demonstrate that laminin-111 is a disease modifier of LAMA2-CMD through different methods of delivery. Together, these studies show the potential for laminin-111 therapy as a novel paradigm for the treatment of LAMA2-CMD.
Collapse
Affiliation(s)
- Pamela Barraza-Flores
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Christina R Bates
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Ariany Oliveira-Santos
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Dean J Burkin
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| |
Collapse
|
13
|
Besser RR, Bowles AC, Alassaf A, Carbonero D, Claure I, Jones E, Reda J, Wubker L, Batchelor W, Ziebarth N, Silvera R, Khan A, Maciel R, Saporta M, Agarwal A. Enzymatically crosslinked gelatin-laminin hydrogels for applications in neuromuscular tissue engineering. Biomater Sci 2020; 8:591-606. [PMID: 31859298 PMCID: PMC7141910 DOI: 10.1039/c9bm01430f] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We report a water-soluble and non-toxic method to incorporate additional extracellular matrix proteins into gelatin hydrogels, while obviating the use of chemical crosslinkers such as glutaraldehyde. Gelatin hydrogels were fabricated using a range of gelatin concentrations (4%-10%) that corresponded to elastic moduli of approximately 1 kPa-25 kPa, respectively, a substrate stiffness relevant for multiple cell types. Microbial transglutaminase was then used to enzymatically crosslink a layer of laminin on top of gelatin hydrogels, resulting in 2-component gelatin-laminin hydrogels. Human induced pluripotent stem cell derived spinal spheroids readily adhered and rapidly extended axons on GEL-LN hydrogels. Axons displayed a more mature morphology and superior electrophysiological properties on GEL-LN hydrogels compared to the controls. Schwann cells on GEL-LN hydrogels adhered and proliferated normally, displayed a healthy morphology, and maintained the expression of Schwann cell specific markers. Lastly, skeletal muscle cells on GEL-LN hydrogels achieved long-term culture for up to 28 days without delamination, while expressing higher levels of terminal genes including myosin heavy chain, MyoD, MuSK, and M-cadherin suggesting enhanced maturation potential and myotube formation compared to the controls. Future studies will employ the superior culture outcomes of this hybrid substrate for engineering functional neuromuscular junctions and related organ on a chip applications.
Collapse
Affiliation(s)
- Rachel R Besser
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Barraza-Flores P, Fontelonga TM, Wuebbles RD, Hermann HJ, Nunes AM, Kornegay JN, Burkin DJ. Laminin-111 protein therapy enhances muscle regeneration and repair in the GRMD dog model of Duchenne muscular dystrophy. Hum Mol Genet 2019; 28:2686-2695. [PMID: 31179490 PMCID: PMC6687953 DOI: 10.1093/hmg/ddz086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 03/22/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked disease affecting ~1 in 5000 males. DMD patients exhibit progressive muscle degeneration and weakness, leading to loss of ambulation and premature death from cardiopulmonary failure. We previously reported that mouse Laminin-111 (msLam-111) protein could reduce muscle pathology and improve muscle function in the mdx mouse model for DMD. In this study, we examined the ability of msLam-111 to prevent muscle disease progression in the golden retriever muscular dystrophy (GRMD) dog model of DMD. The msLam-111 protein was injected into the cranial tibial muscle compartment of GRMD dogs and muscle strength and pathology were assessed. The results showed that msLam-111 treatment increased muscle fiber regeneration and repair with improved muscle strength and reduced muscle fibrosis in the GRMD model. Together, these findings support the idea that Laminin-111 could serve as a novel protein therapy for the treatment of DMD.
Collapse
Affiliation(s)
- Pamela Barraza-Flores
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Tatiana M Fontelonga
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Ryan D Wuebbles
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Hailey J Hermann
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Andreia M Nunes
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
15
|
Nguyen Q, Lim KRQ, Yokota T. Current understanding and treatment of cardiac and skeletal muscle pathology in laminin-α2 chain-deficient congenital muscular dystrophy. APPLICATION OF CLINICAL GENETICS 2019; 12:113-130. [PMID: 31308722 PMCID: PMC6618038 DOI: 10.2147/tacg.s187481] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/08/2019] [Indexed: 01/04/2023]
Abstract
Congenital muscular dystrophy (CMD) is a class of severe early-onset muscular dystrophies affecting skeletal/cardiac muscles as well as the central nervous system (CNS). Laminin-α2 chain-deficient congenital muscular dystrophy (LAMA2 MD), also known as merosin-deficient congenital muscular dystrophy type 1A (MDC1A), is an autosomal recessive CMD characterized by severe muscle weakness and degeneration apparent at birth or in the first 6 months of life. LAMA2 MD is the most common congenital muscular dystrophy, affecting approximately 4 in 500,000 children. The most common cause of death in early-onset LAMA2 MD is respiratory tract infection, with 30% of them dying within the first decade of life. LAMA2 MD is caused by loss-of-function mutations in the LAMA2 gene encoding for the laminin-α2 chain, one of the subunits of laminin-211. Laminin-211 is an extracellular matrix protein that functions to stabilize the basement membrane and muscle fibers during contraction. Since laminin-α2 is expressed in many tissue types including skeletal muscle, cardiac muscle, Schwann cells, and trophoblasts, patients with LAMA2 MD experience a multi-systemic clinical presentation depending on the extent of laminin-α2 chain deficiency. Cardiac manifestations are typically associated with a complete absence of laminin-α2; however, recent case reports highlight cardiac involvement in partial laminin-α2 chain deficiency. Laminin-211 is also expressed in the brain, and many patients have abnormalities on brain imaging; however, mental retardation and/or seizures are rarely seen. Currently, there is no cure for LAMA2 MD, but various therapies are being investigated in an effort to lessen the severity of LAMA2 MD. For example, antisense oligonucleotide-mediated exon skipping and CRISPR-Cas9 genome editing have efficiently restored the laminin-α2 chain in mouse models in vivo. This review consolidates information on the clinical presentation, genetic basis, pathology, and current treatment approaches for LAMA2 MD.
Collapse
Affiliation(s)
- Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Kenji Rowel Q Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB, Canada
| |
Collapse
|
16
|
Willmann R, Gordish-Dressman H, Meinen S, Rüegg MA, Yu Q, Nagaraju K, Kumar A, Girgenrath M, Coffey CBM, Cruz V, Van Ry PM, Bogdanik L, Lutz C, Rutkowski A, Burkin DJ. Improving Reproducibility of Phenotypic Assessments in the DyW Mouse Model of Laminin-α2 Related Congenital Muscular Dystrophy. J Neuromuscul Dis 2019; 4:115-126. [PMID: 28550268 PMCID: PMC5467719 DOI: 10.3233/jnd-170217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Laminin-α2 related Congenital Muscular Dystrophy (LAMA2-CMD) is a progressive muscle disease caused by partial or complete deficiency of laminin-211, a skeletal muscle extracellular matrix protein. In the last decade, basic science research has queried underlying disease mechanisms in existing LAMA2-CMD murine models and identified possible clinical targets and pharmacological interventions. Experimental rigor in preclinical studies is critical to efficiently and accurately quantify both negative and positive results, degree of efficiency of potential therapeutics and determine whether to move a compound forward for additional preclinical testing. In this review, we compare published available data measured to assess three common parameters in the widely used mouse model DyW, that mimics LAMA2-CMD, we quantify variability and analyse its possible sources. Finally, on the basis of this analysis, we suggest standard set of assessments and the use of available standardized protocols, to reduce variability of outcomes in the future and to improve the value of preclinical research.
Collapse
Affiliation(s)
- Raffaella Willmann
- Swiss Foundation for Research on Muscle Diseases, Cortaillod, Switzerland.,Biozentrum, University of Basel, Basel, Switzerland
| | | | | | | | - Qing Yu
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - Ayar Kumar
- Department of Health Sciences, Boston University, Boston, MA, USA
| | | | - Caroline B M Coffey
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| | - Vivian Cruz
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| | - Pam M Van Ry
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| | | | | | - Anne Rutkowski
- Cure Congenital Muscular Dystrophy and Kaiser SCPMG, Los Angeles, CA, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
17
|
Schaaf GJ, Canibano-Fraile R, van Gestel TJM, van der Ploeg AT, Pijnappel WWMP. Restoring the regenerative balance in neuromuscular disorders: satellite cell activation as therapeutic target in Pompe disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:280. [PMID: 31392192 DOI: 10.21037/atm.2019.04.48] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Skeletal muscle is capable of efficiently regenerating after damage in a process mediated by tissue-resident stem cells called satellite cells. This regenerative potential is often compromised under muscle-degenerative conditions. Consequently, the damage produced during degeneration is not efficiently repaired and the balance between repair and damage is lost. Here we review recent progress on the role of satellite cell-mediated repair in neuromuscular disorders with a focus on Pompe disease, an inherited metabolic myopathy caused by deficiency of the lysosomal enzyme acid alpha glucosidase (GAA). Studies performed in patient biopsies as well as in Pompe disease mouse models demonstrate that muscle regeneration activity is compromised despite progressing muscle damage. We describe disease-specific mechanisms of satellite cell dysfunction to highlight the differences between Pompe disease and muscle dystrophies. The mechanisms involved provide possible targets for therapy, such as modulation of autophagy, muscle exercise, and pharmacological modulation of satellite cell activation. Most of these approaches are still experimental, although promising in animal models, still warrant caution with respect to their safety and efficiency profile.
Collapse
Affiliation(s)
- Gerben J Schaaf
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rodrigo Canibano-Fraile
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tom J M van Gestel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Reinhard JR, Lin S, McKee KK, Meinen S, Crosson SC, Sury M, Hobbs S, Maier G, Yurchenco PD, Rüegg MA. Linker proteins restore basement membrane and correct LAMA2-related muscular dystrophy in mice. Sci Transl Med 2018; 9:9/396/eaal4649. [PMID: 28659438 DOI: 10.1126/scitranslmed.aal4649] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Abstract
LAMA2-related muscular dystrophy (LAMA2 MD or MDC1A) is the most frequent form of early-onset, fatal congenital muscular dystrophies. It is caused by mutations in LAMA2, the gene encoding laminin-α2, the long arm of the heterotrimeric (α2, β1, and γ1) basement membrane protein laminin-211 (Lm-211). We establish that despite compensatory expression of laminin-α4, giving rise to Lm-411 (α4, β1, and γ1), muscle basement membrane is labile in LAMA2 MD biopsies. Consistent with this deficit, recombinant Lm-411 polymerized and bound to cultured myotubes only weakly. Polymerization and cell binding of Lm-411 were enhanced by addition of two specifically designed linker proteins. One, called αLNNd, consists of the N-terminal part of laminin-α1 and the laminin-binding site of nidogen-1. The second, called mini-agrin (mag), contains binding sites for laminins and α-dystroglycan. Transgenic expression of mag and αLNNd in a mouse model for LAMA2 MD fully restored basement membrane stability, recovered muscle force and size, increased overall body weight, and extended life span more than five times to a maximum survival beyond 2 years. These findings provide a mechanistic understanding of LAMA2 MD and establish a strong basis for a potential treatment.
Collapse
Affiliation(s)
| | - Shuo Lin
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Karen K McKee
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Sarina Meinen
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Stephanie C Crosson
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Maurizio Sury
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Samantha Hobbs
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Peter D Yurchenco
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Markus A Rüegg
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
19
|
Gawlik KI, Harandi VM, Cheong RY, Petersén Å, Durbeej M. Laminin α1 reduces muscular dystrophy in dy 2J mice. Matrix Biol 2018; 70:36-49. [PMID: 29544677 DOI: 10.1016/j.matbio.2018.02.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 10/17/2022]
Abstract
Muscular dystrophies, including laminin α2 chain-deficient muscular dystrophy (LAMA2-CMD), are associated with immense personal, social and economic burdens. Thus, effective treatments are urgently needed. LAMA2-CMD is either a severe, early-onset condition with complete laminin α2 chain-deficiency or a milder, late-onset form with partial laminin α2 chain-deficiency. Mouse models dy3K/dy3K and dy2J/dy2J, respectively, recapitulate these two forms of LAMA2-CMD very well. We have previously demonstrated that laminin α1 chain significantly reduces muscular dystrophy in laminin α2 chain-deficient dy3K/dy3K mice. Among all the different pre-clinical approaches that have been evaluated in mice, laminin α1 chain-mediated therapy has been shown to be one of the most effective lines of attack. However, it has remained unclear if laminin α1 chain-mediated treatment is also applicable for partial laminin α2 chain-deficiency. Hence, we have generated dy2J/dy2J mice (that express a substantial amount of an N-terminal truncated laminin α2 chain) overexpressing laminin α1 chain in the neuromuscular system. The laminin α1 chain transgene ameliorated the dystrophic phenotype, restored muscle strength and reduced peripheral neuropathy. Thus, these findings provide additional support for the development of laminin α1 chain-based therapy for LAMA2-CMD.
Collapse
Affiliation(s)
- Kinga I Gawlik
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Vahid M Harandi
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Madeleine Durbeej
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Hightower RM, Alexander MS. Genetic modifiers of Duchenne and facioscapulohumeral muscular dystrophies. Muscle Nerve 2018; 57:6-15. [PMID: 28877560 PMCID: PMC5759757 DOI: 10.1002/mus.25953] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2017] [Indexed: 01/05/2023]
Abstract
Muscular dystrophy is defined as the progressive wasting of skeletal muscles that is caused by inherited or spontaneous genetic mutations. Next-generation sequencing has greatly improved the accuracy and speed of diagnosis for different types of muscular dystrophy. Advancements in depth of coverage, convenience, and overall reduced cost have led to the identification of genetic modifiers that are responsible for phenotypic variability in affected patients. These genetic modifiers have been postulated to explain key differences in disease phenotypes, including age of loss of ambulation, steroid responsiveness, and the presence or absence of cardiac defects in patients with the same form of muscular dystrophy. This review highlights recent findings on genetic modifiers of Duchenne and facioscapulohumeral muscular dystrophies based on animal and clinical studies. These genetic modifiers hold great promise to be developed into novel therapeutic targets for the treatment of muscular dystrophies. Muscle Nerve 57: 6-15, 2018.
Collapse
Affiliation(s)
- Rylie M. Hightower
- University of Alabama at Birmingham Graduate School of Biomedical Sciences, Birmingham, AL 35294
| | - Matthew S. Alexander
- Department of Pediatrics, Division of Neurology at Children’s of Alabama and the University of Alabama at Birmingham, Birmingham, AL, 35294
- Department of Genetics, the University of Alabama at Birmingham, Birmingham, AL, 35294
| |
Collapse
|
21
|
Nunes AM, Wuebbles RD, Sarathy A, Fontelonga TM, Deries M, Burkin DJ, Thorsteinsdóttir S. Impaired fetal muscle development and JAK-STAT activation mark disease onset and progression in a mouse model for merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2017; 26:2018-2033. [PMID: 28334989 DOI: 10.1093/hmg/ddx083] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/02/2017] [Indexed: 12/13/2022] Open
Abstract
Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a dramatic neuromuscular disease in which crippling muscle weakness is evident from birth. Here, we use the dyW mouse model for human MDC1A to trace the onset of the disease during development in utero. We find that myotomal and primary myogenesis proceed normally in homozygous dyW-/- embryos. Fetal dyW-/- muscles display the same number of myofibers as wildtype (WT) muscles, but by E18.5 dyW-/- muscles are significantly smaller and muscle size is not recovered post-natally. These results suggest that fetal dyW-/- myofibers fail to grow at the same rate as WT myofibers. Consistent with this hypothesis between E17.5 and E18.5 dyW-/- muscles display a dramatic drop in the number of Pax7- and myogenin-positive cells relative to WT muscles, suggesting that dyW-/- muscles fail to generate enough muscle cells to sustain fetal myofiber growth. Gene expression analysis of dyW-/- E17.5 muscles identified a significant increase in the expression of the JAK-STAT target gene Pim1 and muscles from 2-day and 3-week old dyW-/- mice demonstrate a dramatic increase in pSTAT3 relative to WT muscles. Interestingly, myotubes lacking integrin α7β1, a laminin-receptor, also show a significant increase in pSTAT3 levels compared with WT myotubes, indicating that α7β1 can act as a negative regulator of STAT3 activity. Our data reveal for the first time that dyW-/- mice exhibit a myogenesis defect already in utero. We propose that overactivation of JAK-STAT signaling is part of the mechanism underlying disease onset and progression in dyW-/- mice.
Collapse
Affiliation(s)
- Andreia M Nunes
- Departamento de Biologia Animal, Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal.,Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Ryan D Wuebbles
- Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Apurva Sarathy
- Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Tatiana M Fontelonga
- Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Marianne Deries
- Departamento de Biologia Animal, Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Dean J Burkin
- Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Sólveig Thorsteinsdóttir
- Departamento de Biologia Animal, Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal.,Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| |
Collapse
|
22
|
Fibrosis development in early-onset muscular dystrophies: Mechanisms and translational implications. Semin Cell Dev Biol 2017; 64:181-190. [DOI: 10.1016/j.semcdb.2016.09.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
|
23
|
Potent pro-inflammatory and pro-fibrotic molecules, osteopontin and galectin-3, are not major disease modulators of laminin α2 chain-deficient muscular dystrophy. Sci Rep 2017; 7:44059. [PMID: 28281577 PMCID: PMC5345027 DOI: 10.1038/srep44059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/01/2017] [Indexed: 01/21/2023] Open
Abstract
A large number of human diseases are caused by chronic tissue injury with fibrosis potentially leading to organ failure. There is a need for more effective anti-fibrotic therapies. Congenital muscular dystrophy type 1A (MDC1A) is a devastating form of muscular dystrophy caused by laminin α2 chain-deficiency. It is characterized with early inflammation and build-up of fibrotic lesions, both in patients and MDC1A mouse models (e.g. dy3K/dy3K). Despite the enormous impact of inflammation on tissue remodelling in disease, the inflammatory response in MDC1A has been poorly described. Consequently, a comprehensive understanding of secondary mechanisms (impaired regeneration, enhanced fibrosis) leading to deterioration of muscle phenotype in MDC1A is missing. We have monitored inflammatory processes in dy3K/dy3K muscle and created mice deficient in laminin α2 chain and osteopontin or galectin-3, two pro-inflammatory and pro-fibrotic molecules drastically increased in dystrophic muscle. Surprisingly, deletion of osteopontin worsened the phenotype of dy3K/dy3K mice and loss of galectin-3 did not reduce muscle pathology. Our results indicate that osteopontin could even be a beneficial immunomodulator in MDC1A. This knowledge is essential for the design of future therapeutic interventions for muscular dystrophies that aim at targeting inflammation, especially that osteopontin inhibition has been suggested for Duchenne muscular dystrophy therapy.
Collapse
|
24
|
Abstract
Skeletal muscle performs an essential function in human physiology with defects in genes encoding a variety of cellular components resulting in various types of inherited muscle disorders. Muscular dystrophies (MDs) are a severe and heterogeneous type of human muscle disease, manifested by progressive muscle wasting and degeneration. The disease pathogenesis and therapeutic options for MDs have been investigated for decades using rodent models, and considerable knowledge has been accumulated on the cause and pathogenetic mechanisms of this group of human disorders. However, due to some differences between disease severity and progression, what is learned in mammalian models does not always transfer to humans, prompting the desire for additional and alternative models. More recently, zebrafish have emerged as a novel and robust animal model for the study of human muscle disease. Zebrafish MD models possess a number of distinct advantages for modeling human muscle disorders, including the availability and ease of generating mutations in homologous disease-causing genes, the ability to image living muscle tissue in an intact animal, and the suitability of zebrafish larvae for large-scale chemical screens. In this chapter, we review the current understanding of molecular and cellular mechanisms involved in MDs, the process of myogenesis in zebrafish, and the structural and functional characteristics of zebrafish larval muscles. We further discuss the insights gained from the key zebrafish MD models that have been so far generated, and we summarize the attempts that have been made to screen for small molecules inhibitors of the dystrophic phenotypes using these models. Overall, these studies demonstrate that zebrafish is a useful in vivo system for modeling aspects of human skeletal muscle disorders. Studies using these models have contributed both to the understanding of the pathogenesis of muscle wasting disorders and demonstrated their utility as highly relevant models to implement therapeutic screening regimens.
Collapse
Affiliation(s)
- M Li
- Monash University, Clayton, VIC, Australia
| | - K J Hromowyk
- The Ohio State University, Columbus, OH, United States
| | - S L Amacher
- The Ohio State University, Columbus, OH, United States
| | - P D Currie
- Monash University, Clayton, VIC, Australia
| |
Collapse
|
25
|
Yao Y, Norris EH, Mason CE, Strickland S. Laminin regulates PDGFRβ(+) cell stemness and muscle development. Nat Commun 2016; 7:11415. [PMID: 27138650 PMCID: PMC4857399 DOI: 10.1038/ncomms11415] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/22/2016] [Indexed: 12/15/2022] Open
Abstract
Muscle-resident PDGFRβ+ cells, which include pericytes and PW1+ interstitial cells (PICs), play a dual role in muscular dystrophy. They can either undergo myogenesis to promote muscle regeneration or differentiate into adipocytes and other cells to compromise regeneration. How the differentiation and fate determination of PDGFRβ+ cells are regulated, however, remains unclear. Here, by utilizing a conditional knockout mouse line, we report that PDGFRβ+ cell-derived laminin inhibits their proliferation and adipogenesis, but is indispensable for their myogenesis. In addition, we show that laminin alone is able to partially reverse the muscle dystrophic phenotype in these mice at the molecular, structural and functional levels. Further RNAseq analysis reveals that laminin regulates PDGFRβ+ cell differentiation/fate determination via gpihbp1. These data support a critical role of laminin in the regulation of PDGFRβ+ cell stemness, identify an innovative target for future drug development and may provide an effective treatment for muscular dystrophy. Muscle PDGFRβ+ cells are interstitial stem/progenitor cells with myogenic potential. Here, Yao et al. show that PDGFRβ+ cell-derived laminin actively regulates their proliferation, differentiation and fate determination.
Collapse
Affiliation(s)
- Yao Yao
- Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA.,College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, Minnesota 55812, USA
| | - Erin H Norris
- Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10065, USA.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, New York 10065, USA.,Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York 10065, USA.,The Feil Family Brain and Mind Research Institute, New York, New York 10065, USA
| | - Sidney Strickland
- Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA
| |
Collapse
|
26
|
Tjondrokoesoemo A, Schips T, Kanisicak O, Sargent MA, Molkentin JD. Genetic overexpression of Serpina3n attenuates muscular dystrophy in mice. Hum Mol Genet 2016; 25:1192-202. [PMID: 26744329 DOI: 10.1093/hmg/ddw005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/05/2016] [Indexed: 01/06/2023] Open
Abstract
Muscular dystrophy (MD) is associated with mutations in genes that stabilize the myofiber plasma membrane, such as through the dystrophin-glycoprotein complex (DGC). Instability of this complex or defects in membrane repair/integrity leads to calcium influx and myofiber necrosis leading to progressive dystrophic disease. MD pathogenesis is also associated with increased skeletal muscle protease levels and activity that could augment weakening of the sarcolemma through greater degradation of cellular attachment complexes. Here, we observed a compensatory increase in the serine protease inhibitor Serpina3n in mouse models of MD and after acute muscle tissue injury. Serpina3n muscle-specific transgenic mice were generated to model this increase in expression, which reduced the activity of select proteases in dystrophic skeletal muscle and protected muscle from both acute injury with cardiotoxin and from chronic muscle disease in the mdx or Sgcd(-/-) MD genetic backgrounds. The Serpina3n transgene mitigated muscle degeneration and fibrosis, reduced creatine kinase serum levels, restored running capacity on a treadmill and reduced muscle membrane leakiness in vivo that is characteristic of mdx and Sgcd(-/-) mice. Mechanistically, we show that increased Serpina3n promotes greater sarcolemma membrane integrity and stability in dystrophic mouse models in association with increased membrane residence of the integrins, the DGC/utrophin-glycoprotein complex of proteins and annexin A1. Hence, Serpina3n blocks endogenous increases in the activity of select skeletal muscle resident proteases during injury or dystrophic disease, which stabilizes the sarcolemma leading to less myofiber degeneration and increased regeneration. These results suggest the use of select protease inhibitors as a strategy for treating MD.
Collapse
Affiliation(s)
| | - Tobias Schips
- Department of Pediatrics, University of Cincinnati and
| | | | | | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati and Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC7020, Cincinnati, OH 45229, USA
| |
Collapse
|
27
|
Riederer I, Bonomo AC, Mouly V, Savino W. Laminin therapy for the promotion of muscle regeneration. FEBS Lett 2015; 589:3449-53. [DOI: 10.1016/j.febslet.2015.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/25/2015] [Accepted: 10/06/2015] [Indexed: 12/18/2022]
|
28
|
Gawlik KI, Durbeej M. Deletion of integrin α7 subunit does not aggravate the phenotype of laminin α2 chain-deficient mice. Sci Rep 2015; 5:13916. [PMID: 26355035 PMCID: PMC4564817 DOI: 10.1038/srep13916] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/10/2015] [Indexed: 11/09/2022] Open
Abstract
Laminin-211 is a major constituent of the skeletal muscle basement membrane, exerting its biological functions by binding to cell surface receptors integrin α7β1 and dystroglycan (the latter is part of the dystrophin-glycoprotein complex). The importance of these molecules for normal muscle function is underscored by the fact that their respective deficiency leads to different forms of muscular dystrophy with different severity in humans and animal models. We recently demonstrated that laminin α2 chain and members of the dystrophin-glycoprotein complex have overlapping but non-redundant roles despite being part of the same adhesion complex. To analyse whether laminin-211 and integrin α7 subunit have non-redundant functions we generated mice deficient in laminin α2 chain and integrin α7 subunit (dy3K/itga7). We show that lack of both molecules did not exacerbate the severe phenotype of laminin α2-chain deficient animals. They displayed the same weight, survival and dystrophic pattern of muscle biopsy, with similar degree of inflammation and fibrosis. These data suggest that laminin-211 and integrin α7β1 have intersecting roles in skeletal muscle.
Collapse
Affiliation(s)
- Kinga I Gawlik
- Department of Experimental Medical Science, Muscle Biology Unit, Lund University, Sweden
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Muscle Biology Unit, Lund University, Sweden
| |
Collapse
|
29
|
Burkin DJ, Fontelonga TM. Mesothelioma cells breaking bad: loss of integrin α7 promotes cell motility and poor clinical outcomes in patients. J Pathol 2015; 237:282-4. [PMID: 26174987 DOI: 10.1002/path.4587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 07/06/2015] [Accepted: 07/10/2015] [Indexed: 01/04/2023]
Abstract
Mesothelioma is a disease of pleural cells lining the lungs which is often caused by exposure to asbestos. The molecular mechanism(s) that regulate the transformation of pleural mesothelioma cells to a migratory and malignant phenotype are unclear. In recent work published in this journal, Laszlo et al performed a set of elegant experiments to identify a key molecular mechanism that may explain the aggressive nature of this disease. Using patient-derived mesothelioma cells with high versus low migratory activity, the authors conducted a genome-wide expression analysis. They identified a significant reduction in ITGA7 expression only in highly migratory malignant pleural mesothelioma cells and showed that loss of ITGA7 expression was associated with methylation of the promoter. Forced expression of integrin α7 reversed the migratory phenotype of these cells. Finally, the authors identified a strong correlation between ITGA7 expression and patient survival. Together, these results identify expression of integrin α7 as a molecular mechanism for the aggressive migratory transformation of mesothelioma and identify a potentially novel diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Dean J Burkin
- Department of Pharmacology, Center for Molecular Medicine, University of Nevada School of Medicine, 1664 N Virginia Avenue, Reno, NV, 89557, USA
| | - Tatiana M Fontelonga
- Department of Pharmacology, Center for Molecular Medicine, University of Nevada School of Medicine, 1664 N Virginia Avenue, Reno, NV, 89557, USA
| |
Collapse
|
30
|
Durbeej M. Laminin-α2 Chain-Deficient Congenital Muscular Dystrophy: Pathophysiology and Development of Treatment. CURRENT TOPICS IN MEMBRANES 2015; 76:31-60. [PMID: 26610911 DOI: 10.1016/bs.ctm.2015.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Laminin-211 is a major constituent of the skeletal muscle basement membrane. It stabilizes skeletal muscle and influences signal transduction events from the myomatrix to the muscle cell. Mutations in the gene encoding the α2 chain of laminin-211 lead to congenital muscular dystrophy type 1A (MDC1A), a life-threatening disease characterized by severe hypotonia, progressive muscle weakness, and joint contractures. Common complications include severely impaired motor ability, respiratory failure, and feeding difficulties. Several adequate animal models for laminin-α2 chain deficiency exist and analyses of different MDC1A mouse models have led to a significant improvement in our understanding of MDC1A pathogenesis. Importantly, the animal models have been indispensable tools for the preclinical development of new therapeutic approaches for laminin-α2 chain deficiency, highlighting a number of important disease driving mechanisms that can be targeted by pharmacological approaches. In this chapter, I will describe laminin-211 and discuss the cellular and molecular pathophysiology of MDC1A as well as progression toward development of treatment.
Collapse
Affiliation(s)
- Madeleine Durbeej
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
31
|
de Oliveira BM, Matsumura CY, Fontes-Oliveira CC, Gawlik KI, Acosta H, Wernhoff P, Durbeej M. Quantitative proteomic analysis reveals metabolic alterations, calcium dysregulation, and increased expression of extracellular matrix proteins in laminin α2 chain-deficient muscle. Mol Cell Proteomics 2014; 13:3001-13. [PMID: 24994560 DOI: 10.1074/mcp.m113.032276] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Congenital muscular dystrophy with laminin α2 chain deficiency (MDC1A) is one of the most severe forms of muscular disease and is characterized by severe muscle weakness and delayed motor milestones. The genetic basis of MDC1A is well known, yet the secondary mechanisms ultimately leading to muscle degeneration and subsequent connective tissue infiltration are not fully understood. In order to obtain new insights into the molecular mechanisms underlying MDC1A, we performed a comparative proteomic analysis of affected muscles (diaphragm and gastrocnemius) from laminin α2 chain-deficient dy(3K)/dy(3K) mice, using multidimensional protein identification technology combined with tandem mass tags. Out of the approximately 700 identified proteins, 113 and 101 proteins, respectively, were differentially expressed in the diseased gastrocnemius and diaphragm muscles compared with normal muscles. A large portion of these proteins are involved in different metabolic processes, bind calcium, or are expressed in the extracellular matrix. Our findings suggest that metabolic alterations and calcium dysregulation could be novel mechanisms that underlie MDC1A and might be targets that should be explored for therapy. Also, detailed knowledge of the composition of fibrotic tissue, rich in extracellular matrix proteins, in laminin α2 chain-deficient muscle might help in the design of future anti-fibrotic treatments. All MS data have been deposited in the ProteomeXchange with identifier PXD000978 (http://proteomecentral.proteomexchange.org/dataset/PXD000978).
Collapse
Affiliation(s)
- Bruno Menezes de Oliveira
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Cintia Y Matsumura
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden; ¶Departament of Functional and Structural Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Cibely C Fontes-Oliveira
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Kinga I Gawlik
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Helena Acosta
- ‖Stem Cell Center, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Patrik Wernhoff
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Madeleine Durbeej
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden;
| |
Collapse
|
32
|
Körner Z, Fontes-Oliveira CC, Holmberg J, Carmignac V, Durbeej M. Bortezomib partially improves laminin α2 chain-deficient muscular dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1518-28. [PMID: 24631023 DOI: 10.1016/j.ajpath.2014.01.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
Congenital muscular dystrophy, caused by mutations in LAMA2 (the gene encoding laminin α2 chain), is a severe and incapacitating disease for which no therapy is yet available. We have recently demonstrated that proteasome activity is increased in laminin α2 chain-deficient muscle and that treatment with the nonpharmaceutical proteasome inhibitor MG-132 reduces muscle pathology in laminin α2 chain-deficient dy(3K)/dy(3K) mice. Here, we explore the use of the selective and therapeutic proteasome inhibitor bortezomib (currently used for treatment of relapsed multiple myeloma and mantle cell lymphoma) in dy(3K)/dy(3K) mice and in congenital muscular dystrophy type 1A muscle cells. Outcome measures included quantitative muscle morphology, gene and miRNA expression analyses, proteasome activity, motor activity, and survival. Bortezomib improved several histological hallmarks of disease, partially normalized miRNA expression (miR-1 and miR-133a), and enhanced body weight, locomotion, and survival of dy(3K)/dy(3K) mice. In addition, bortezomib reduced proteasome activity in congenital muscular dystrophy type 1A myoblasts and myotubes. These findings provide evidence that the proteasome inhibitor bortezomib partially reduces laminin α2 chain-deficient muscular dystrophy. Investigation of the clinical efficacy of bortezomib administration in congenital muscular dystrophy type 1A clinical trials may be warranted.
Collapse
Affiliation(s)
- Zandra Körner
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Johan Holmberg
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Virginie Carmignac
- Genetics of Developmental Abnormalities Team, EA4271, University of Burgundy, Dijon, France
| | - Madeleine Durbeej
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
33
|
Van Ry PM, Minogue P, Hodges BL, Burkin DJ. Laminin-111 improves muscle repair in a mouse model of merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2013; 23:383-96. [PMID: 24009313 DOI: 10.1093/hmg/ddt428] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a severe and fatal muscle-wasting disease with no cure. MDC1A patients and the dy(W-/-) mouse model exhibit severe muscle weakness, demyelinating neuropathy, failed muscle regeneration and premature death. We have recently shown that laminin-111, a form of laminin found in embryonic skeletal muscle, can substitute for the loss of laminin-211/221 and prevent muscle disease progression in the dy(W-/-) mouse model. What is unclear from these studies is whether laminin-111 can restore failed regeneration to laminin-α2-deficient muscle. To investigate the potential of laminin-111 protein therapy to improve muscle regeneration, laminin-111 or phosphate-buffered saline-treated laminin-α2-deficient muscle was damaged with cardiotoxin and muscle regeneration quantified. Our results show laminin-111 treatment promoted an increase in myofiber size and number, and an increased expression of α7β1 integrin, Pax7, myogenin and embryonic myosin heavy chain, indicating a restoration of the muscle regenerative program. Together, our results show laminin-111 restores muscle regeneration to laminin-α2-deficient muscle and further supports laminin-111 protein as a therapy for the treatment of MDC1A.
Collapse
Affiliation(s)
- Pam M Van Ry
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA and
| | | | | | | |
Collapse
|
34
|
Wuebbles RD, Sarathy A, Kornegay JN, Burkin DJ. Levels of α7 integrin and laminin-α2 are increased following prednisone treatment in the mdx mouse and GRMD dog models of Duchenne muscular dystrophy. Dis Model Mech 2013; 6:1175-84. [PMID: 23846963 PMCID: PMC3759337 DOI: 10.1242/dmm.012211] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disease for which there is no cure and limited treatment options. Prednisone is currently the first line treatment option for DMD and studies have demonstrated that it improves muscle strength. Although prednisone has been used for the treatment of DMD for decades, the mechanism of action of this drug remains unclear. Recent studies have shown that the α7β1 integrin is a major modifier of disease progression in mouse models of DMD and is therefore a target for drug-based therapies. In this study we examined whether prednisone increased α7β1 integrin levels in mdx mouse and GRMD dog models and myogenic cells from humans with DMD. Our results show that prednisone promotes an increase in α7 integrin protein in cultured myogenic cells and in the muscle of mdx and GRMD animal models of DMD. The prednisone-mediated increase in α7 integrin was associated with increased laminin-α2 in prednisone-treated dystrophin-deficient muscle. Together, our results suggest that prednisone acts in part through increased merosin in the muscle basal lamina and through sarcolemmal stabilization of α7β1 integrin in dystrophin-deficient muscle. These results indicate that therapies that target an increase in muscle α7β1 integrin, its signaling pathways and/or laminin could be therapeutic in DMD.
Collapse
Affiliation(s)
- Ryan D Wuebbles
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | |
Collapse
|
35
|
Han J, Gerstenhaber JA, Lazarovici P, Lelkes PI. Tissue Factor Activity and ECM-Related Gene Expression in Human Aortic Endothelial Cells Grown on Electrospun Biohybrid Scaffolds. Biomacromolecules 2013; 14:1338-48. [DOI: 10.1021/bm400450m] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jingjia Han
- School
of Biomedical
Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104,
United States
| | - Jonathan A. Gerstenhaber
- School
of Biomedical
Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104,
United States
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania 19122,
United States
| | - Philip Lazarovici
- School
of Biomedical
Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104,
United States
- School of Pharmacy
Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120,
Israel
| | - Peter I. Lelkes
- School
of Biomedical
Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104,
United States
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania 19122,
United States
| |
Collapse
|
36
|
AAV-mediated overexpression of human α7 integrin leads to histological and functional improvement in dystrophic mice. Mol Ther 2013; 21:520-5. [PMID: 23319059 DOI: 10.1038/mt.2012.281] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by mutations in the DMD gene, with loss of its gene product, dystrophin. Dystrophin helps link integral membrane proteins to the actin cytoskeleton and stabilizes the sarcolemma during muscle activity. We investigated an alternative therapeutic approach to dystrophin replacement by overexpressing human α7 integrin (ITGA7) using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal and cardiac muscle that links the extracellular matrix (ECM) to the actin skeleton. It is modestly upregulated in DMD muscle and has been proposed to be an important modifier of dystrophic symptoms. We delivered rAAV8.MCK.ITGA7 to the lower limb of mdx mice through isolated limb perfusion (ILP) of the femoral artery. We demonstrated ~50% of fibers in the tibialis anterior (TA) and extensor digitorum longus (EDL) overexpressing α7 integrin at the sarcolemma following AAV gene transfer. The increase in ITGA7 in skeletal muscle significantly protected against loss of force following eccentric contraction-induced injury compared with untreated (contralateral) muscles while specific force following tetanic contraction was unchanged. Reversal of additional dystrophic features included reduced Evans blue dye (EBD) uptake and increased muscle fiber diameter. Taken together, this data shows that rAAV8.MCK.ITGA7 gene transfer stabilizes the sarcolemma potentially preserving mdx muscle from further damage. This therapeutic approach demonstrates promise as a viable treatment for DMD with further implications for other forms of muscular dystrophy.
Collapse
|
37
|
NAD+ biosynthesis ameliorates a zebrafish model of muscular dystrophy. PLoS Biol 2012; 10:e1001409. [PMID: 23109907 PMCID: PMC3479101 DOI: 10.1371/journal.pbio.1001409] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 09/06/2012] [Indexed: 01/27/2023] Open
Abstract
NAD+ improves muscle tissue structure and function in dystrophic zebrafish by increasing basement membrane organization. Muscular dystrophies are common, currently incurable diseases. A subset of dystrophies result from genetic disruptions in complexes that attach muscle fibers to their surrounding extracellular matrix microenvironment. Cell-matrix adhesions are exquisite sensors of physiological conditions and mediate responses that allow cells to adapt to changing conditions. Thus, one approach towards finding targets for future therapeutic applications is to identify cell adhesion pathways that mediate these dynamic, adaptive responses in vivo. We find that nicotinamide riboside kinase 2b-mediated NAD+ biosynthesis, which functions as a small molecule agonist of muscle fiber-extracellular matrix adhesion, corrects dystrophic phenotypes in zebrafish lacking either a primary component of the dystrophin-glycoprotein complex or integrin alpha7. Exogenous NAD+ or a vitamin precursor to NAD+ reduces muscle fiber degeneration and results in significantly faster escape responses in dystrophic embryos. Overexpression of paxillin, a cell adhesion protein downstream of NAD+ in this novel cell adhesion pathway, reduces muscle degeneration in zebrafish with intact integrin receptors but does not improve motility. Activation of this pathway significantly increases organization of laminin, a major component of the extracellular matrix basement membrane. Our results indicate that the primary protective effects of NAD+ result from changes to the basement membrane, as a wild-type basement membrane is sufficient to increase resilience of dystrophic muscle fibers to damage. The surprising result that NAD+ supplementation ameliorates dystrophy in dystrophin-glycoprotein complex– or integrin alpha7–deficient zebrafish suggests the existence of an additional laminin receptor complex that anchors muscle fibers to the basement membrane. We find that integrin alpha6 participates in this pathway, but either integrin alpha7 or the dystrophin-glycoprotein complex is required in conjunction with integrin alpha6 to reduce muscle degeneration. Taken together, these results define a novel cell adhesion pathway that may have future therapeutic relevance for a broad spectrum of muscular dystrophies. A variety of diseases, both inherited and acquired, affect muscle tissues in humans. Critical to muscle homeostasis is the anchoring of muscle fibers to their surrounding microenvironment through cell adhesion complexes that help to resist the repeated stress experienced during muscle contraction. Genetic mutations in these complexes weaken this mechanical attachment, making fibers more susceptible to damage and death. The resulting increased fiber degeneration can eventually lead to progressive muscle-wasting diseases, known collectively as muscular dystrophies. Although clinical trials are ongoing, there is presently no way to cure the loss of muscle structure and function associated with these diseases. We identified a novel cell adhesion pathway involving integrin alpha6 that promotes adhesion of muscle cells to their microenvironment. Here, we show that activation of this pathway not only significantly reduces muscle degeneration but also improves the swimming ability of dystrophic zebrafish. We explore the likely benefits and limitations of this pathway in treating symptoms of congenital muscular dystrophies. Our findings suggest that activation of this pathway (for example, by boosting levels of NAD+) has the potential to ameliorate loss of muscle structure and function in multiple muscular dystrophies.
Collapse
|
38
|
Gupta VA, Kawahara G, Myers JA, Chen AT, Hall TE, Manzini MC, Currie PD, Zhou Y, Zon LI, Kunkel LM, Beggs AH. A splice site mutation in laminin-α2 results in a severe muscular dystrophy and growth abnormalities in zebrafish. PLoS One 2012; 7:e43794. [PMID: 22952766 PMCID: PMC3428294 DOI: 10.1371/journal.pone.0043794] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/24/2012] [Indexed: 11/18/2022] Open
Abstract
Congenital muscular dystrophy (CMD) is a clinically and genetically heterogeneous group of inherited muscle disorders. In patients, muscle weakness is usually present at or shortly after birth and is progressive in nature. Merosin deficient congenital muscular dystrophy (MDC1A) is a form of CMD caused by a defect in the laminin-α2 gene (LAMA2). Laminin-α2 is an extracellular matrix protein that interacts with the dystrophin-dystroglycan (DGC) complex in membranes providing stability to muscle fibers. In an N-ethyl-N-nitrosourea mutagenesis screen to develop zebrafish models of neuromuscular diseases, we identified a mutant fish that exhibits severe muscular dystrophy early in development. Genetic mapping identified a splice site mutation in the lama2 gene. This splice site is highly conserved in humans and this mutation results in mis-splicing of RNA and a loss of protein function. Homozygous lama2 mutant zebrafish, designated lama2cl501/cl501, exhibited reduced motor function and progressive degeneration of skeletal muscles and died at 8–15 days post fertilization. The skeletal muscles exhibited damaged myosepta and detachment of myofibers in the affected fish. Laminin-α2 deficiency also resulted in growth defects in the brain and eye of the mutant fish. This laminin-α2 deficient mutant fish represents a novel disease model to develop therapies for modulating splicing defects in congenital muscular dystrophies and to restore the muscle function in human patients with CMD.
Collapse
Affiliation(s)
- Vandana A. Gupta
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Genri Kawahara
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Jennifer A. Myers
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Aye T. Chen
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thomas E. Hall
- Australian Regenerative Medicine Institute, Monash University, Clayton Campus, Victoria, Australia
| | - M. Chiara Manzini
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton Campus, Victoria, Australia
| | - Yi Zhou
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leonard I. Zon
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, San Francisco, California, United States of America
| | - Louis M. Kunkel
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
| | - Alan H. Beggs
- Genomics Program and Division of Genetics, Boston Children’s Hospital, Harvard Medical School, The Manton Center for Orphan Disease Research, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
39
|
Sztal TE, Sonntag C, Hall TE, Currie PD. Epistatic dissection of laminin-receptor interactions in dystrophic zebrafish muscle. Hum Mol Genet 2012; 21:4718-31. [PMID: 22859503 DOI: 10.1093/hmg/dds312] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Laminins form essential components of the basement membrane and are integral to forming and maintaining muscle integrity. Mutations in the human Laminin-alpha2 (LAMA2) gene result in the most common form of congenital muscular dystrophy, MDC1A. We have previously identified a zebrafish model of MDC1A called candyfloss (caf), carrying a loss-of-function mutation in the zebrafish lama2 gene. In the skeletal muscle, laminins connect the muscle cell to the extracellular matrix (ECM) by binding either dystroglycan or integrins at the cell membrane. Through epistasis experiments, we have established that both adhesion systems individually contribute to the maintenance of fibre adhesions and exhibit muscle detachment phenotypes. However, larval zebrafish in which both adhesion systems are simultaneously genetically inactivated possess a catastrophic failure of muscle attachment that is far greater than a simple addition of individual phenotypes would predict. We provide evidence that this is due to other crucial laminins present in addition to Lama2, which aid muscle cell attachments and integrity. We have found that lama1 is important for maintaining attachments, whereas lama4 is localized and up-regulated in damaged fibres, which appears to contribute to fibre survival. Importantly, our results show that endogenous secretion of laminins from the surrounding tissues has the potential to reinforce fibre attachments and strengthen laminin-ECM attachments. Collectively these findings provide a better understanding of the cellular pathology of MDC1A and help in designing effective therapies.
Collapse
Affiliation(s)
- Tamar E Sztal
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | | | | | | |
Collapse
|
40
|
Rooney JE, Knapp JR, Hodges BL, Wuebbles RD, Burkin DJ. Laminin-111 protein therapy reduces muscle pathology and improves viability of a mouse model of merosin-deficient congenital muscular dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1593-602. [PMID: 22322301 DOI: 10.1016/j.ajpath.2011.12.019] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 10/11/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a lethal muscle-wasting disease that is caused by mutations in the LAMA2 gene, resulting in the loss of laminin-α2 protein. MDC1A patients exhibit severe muscle weakness from birth, are confined to a wheelchair, require ventilator assistance, and have reduced life expectancy. There are currently no effective treatments or cures for MDC1A. Laminin-α2 is required for the formation of heterotrimeric laminin-211 (ie, α2, β1, and γ1) and laminin-221 (ie, α2, β2, and γ1), which are major constituents of skeletal muscle basal lamina. Laminin-111 (ie, α1, β1, and γ1) is the predominant laminin isoform in embryonic skeletal muscle and supports normal skeletal muscle development in laminin-α2-deficient muscle but is absent from adult skeletal muscle. In this study, we determined whether treatment with Engelbreth-Holm-Swarm-derived mouse laminin-111 protein could rescue MDC1A in the dy(W-/-) mouse model. We demonstrate that laminin-111 protein systemically delivered to the muscles of laminin-α2-deficient mice prevents muscle pathology, improves muscle strength, and dramatically increases life expectancy. Laminin-111 also prevented apoptosis in laminin-α2-deficient mouse muscle and primary human MDC1A myogenic cells, which indicates a conserved mechanism of action and cross-reactivity between species. Our results demonstrate that laminin-111 can serve as an effective protein substitution therapy for the treatment of muscular dystrophy in the dy(W-/-) mouse model and establish the potential for its use in the treatment of MDC1A.
Collapse
Affiliation(s)
- Jachinta E Rooney
- Department of Pharmacology, Center for Molecular Medicine, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
The extracellular matrix (ECM) provides a solid scaffold and signals to cells through ECM receptors. The cell-matrix interactions are crucial for normal biological processes and when disrupted they may lead to pathological processes. In particular, the biological importance of ECM-cell membrane-cytoskeleton interactions in skeletal muscle is accentuated by the number of inherited muscle diseases caused by mutations in proteins conferring these interactions. In this review we introduce laminins, collagens, dystroglycan, integrins, dystrophin and sarcoglycans. Mutations in corresponding genes cause various forms of muscular dystrophy. The muscle disorders are presented as well as advances toward the development of treatment.
Collapse
Affiliation(s)
- Virginie Carmignac
- Muscle Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|