1
|
Xin J, Liu S. Identifying hub genes and dysregulated pathways in Duchenne muscular dystrophy. Int J Neurosci 2025; 135:375-387. [PMID: 38179963 DOI: 10.1080/00207454.2024.2302551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
PURPOSE The aim of this study was to identify the hub genes and dysregulated pathways in the progression of duchenne muscular dystrophy (DMD) and to unveil detailedly the cellular and molecular mechanisms associated with DMD for developing efficacious treatments in the future. MATERIAL AND METHODS Three mRNA microarray datasets (GSE13608, GSE38417 and GSE109178) were downloaded from Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) between DMD and normal tissues were obtained via R package. Function enrichment analyses were implemented respectively using DAVID online database. The network analysis of protein-protein interaction network (PPI) was conducted using String. Cytoscape and String were used to analyse modules and screen hub genes. The expression of the identified hub genes was confirmed in mdx mice through using qRT-PCR. RESULTS In total, 519 DEGs were identified, consisting of 393 upregulated genes and 126 downregulated genes. The enriched functions and pathways of the DEGs mainly involve extracellular matrix organization, collagen fibril organization, interferon-gamma-mediated signaling pathway, muscle contraction, endoplasmic reticulum lumen, MHC class II receptor activity, phagosome, graft-versus-host disease, cardiomyocytes, calcium signaling pathway. Twelve hub genes were discovered and biological process analysis proved that these genes were mainly enriched cell cycle, cell division. The result of qRT-PCR suggested that increase in expression of CD44, ECT2, TYMS, MAGEL2, HLA-DMA, SERPINH1, TNNT2 was confirmed in mdx mice and the downregulation of ASB2 and LEPREL1 was also observed. CONCLUSION In conclusion, DEGs and hub genes identified in the current research help us probe the molecular mechanisms underlying the pathogenesis and progression of DMD, and provide candidate targets for diagnosis and treatment of DMD.
Collapse
Affiliation(s)
- Jianzeng Xin
- College of life sciences, Yantai University, Yantai, P. R. China
| | - Sheng Liu
- School of Pharmacy, Yantai University, Yantai, P. R. China
| |
Collapse
|
2
|
Boccanegra B, Lenti R, Mantuano P, Conte E, Tulimiero L, Piercy RJ, Cappellari O, Hildyard JCW, De Luca A. Determination of qPCR reference genes suitable for normalizing gene expression in a novel model of Duchenne muscular dystrophy, the D2-mdx mouse. PLoS One 2024; 19:e0310714. [PMID: 39535998 PMCID: PMC11560031 DOI: 10.1371/journal.pone.0310714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a X-linked neuromuscular disorder arising from mutations in the dystrophin gene, leading to a progressive muscle wasting and disability. Currently there is no universal therapy, and there is thus a strong interest in preclinical studies for finding novel treatments. The most widely used and characterized mouse model for DMD is the C57BL/10ScSn-Dmdmdx/J (BL10-mdx), but this model exhibits mild pathology and does not replicate key features of human disease. The D2.B10-Dmdmdx/J (D2-mdx) mouse is a more recent model which seems to better mimics the complex human DMD phenotype. However, the D2-mdx mouse remains less extensively characterised than its BL10-mdx counterpart. Quantitative PCR analysis of gene expression is an important tool to monitor disease progression and evaluate therapeutic efficacy, but measurements must be normalised to stably expressed reference genes, which should ideally be determined and validated empirically. We examined gene expression in the gastrocnemius (GC), diaphragm (DIA) and heart in the D2-mdx mouse, the BL10-mdx mouse, and appropriate strain-matched wild-type controls (D2-wt and BL10-wt), from 4 to 52 weeks of age, using a large panel of candidate references (ACTB, AP3D1, CSNK2A2, GAPDH, HPRT1, PAK1IP1, RPL13A, SDHA, and in the heart, also HTATSF1 and HMBS). Data was analyzed using GeNorm, Bestkeeper, deltaCt and Normfinder algorithms to identify stable references under multiple possible scenarios. We show that CSNK2A2, AP3D1 and ACTB represent strong universal reference genes in both GC and DIA, regardless of age, muscle type, strain and genotype, while HTATSF1 and SDHA are optimal for the heart. GAPDH, HPRT1 and RPL13A were conversely revealed to be poor references, showing tissue-, age- or disease-specific changes in expression. Our results illustrate the importance of determining appropriate reference genes for specific comparative scenarios, but also reconfirm that universal panels can nevertheless be identified for normalising gene expression studies in even complex pathological states.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Roberta Lenti
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Mantuano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Lisamaura Tulimiero
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - Richard J. Piercy
- Department of Clinical Sciences and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, United Kingdom
| | - Ornella Cappellari
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | - John C. W. Hildyard
- Department of Clinical Sciences and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, United Kingdom
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
3
|
Mehmood H, Kasher PR, Barrett-Jolley R, Walmsley GL. Aligning with the 3Rs: alternative models for research into muscle development and inherited myopathies. BMC Vet Res 2024; 20:477. [PMID: 39425123 PMCID: PMC11488271 DOI: 10.1186/s12917-024-04309-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Inherited and acquired muscle diseases are an important cause of morbidity and mortality in human medical and veterinary patients. Researchers use models to study skeletal muscle development and pathology, improve our understanding of disease pathogenesis and explore new treatment options. Experiments on laboratory animals, including murine and canine models, have led to huge advances in congenital myopathy and muscular dystrophy research that have translated into clinical treatment trials in human patients with these debilitating and often fatal conditions. Whilst animal experimentation has enabled many significant and impactful discoveries that otherwise may not have been possible, we have an ethical and moral, and in many countries also a legal, obligation to consider alternatives. This review discusses the models available as alternatives to mammals for muscle development, biology and disease research with a focus on inherited myopathies. Cell culture models can be used to replace animals for some applications: traditional monolayer cultures (for example, using the immortalised C2C12 cell line) are accessible, tractable and inexpensive but developmentally limited to immature myotube stages; more recently, developments in tissue engineering have led to three-dimensional cultures with improved differentiation capabilities. Advances in computer modelling and an improved understanding of pathogenetic mechanisms are likely to herald new models and opportunities for replacement. Where this is not possible, a 3Rs approach advocates partial replacement with the use of less sentient animals (including invertebrates (such as worms Caenorhabditis elegans and fruit flies Drosophila melanogaster) and embryonic stages of small vertebrates such as the zebrafish Danio rerio) alongside refinement of experimental design and improved research practices to reduce the numbers of animals used and the severity of their experience. An understanding of the advantages and disadvantages of potential models is essential for researchers to determine which can best facilitate answering a specific scientific question. Applying 3Rs principles to research not only improves animal welfare but generates high-quality, reproducible and reliable data with translational relevance to human and animal patients.
Collapse
Affiliation(s)
- Hashir Mehmood
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Allianceand the, University of Manchester , Manchester, M6 8HD, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Gemma L Walmsley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, South Wirral, Neston, CH64 7TE, UK.
| |
Collapse
|
4
|
Stirm M, Klymiuk N, Nagashima H, Kupatt C, Wolf E. Pig models for translational Duchenne muscular dystrophy research. Trends Mol Med 2024; 30:950-964. [PMID: 38749865 DOI: 10.1016/j.molmed.2024.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 10/12/2024]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the X-linked DMD gene, resulting in the absence of dystrophin, progressive muscle degeneration, and heart failure. Genetically tailored pig models resembling human DMD mutations recapitulate the biochemical, clinical, and pathological hallmarks of DMD with an accelerated disease progression compared to human patients. DMD pigs have been used to evaluate therapeutic concepts such as gene editing to reframe a disrupted DMD reading frame or the delivery of artificial chromosome vectors carrying the complete DMD gene. Moreover, DMD pigs have been instrumental in validating new diagnostic modalities such as multispectral optoacoustic tomography (MSOT) for non-invasive monitoring of disease progression. DMD pigs may thus help to bridge the gap between proof-of-concept studies in cellular or rodent models and clinical studies in patients.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Large Animal Models in Cardiovascular Research, Internal Medical Department I, Technical University of Munich (TU Munich), 81675 Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa 214-8571, Japan
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, 81675 Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
5
|
Delaney R, O'Halloran KD. Respiratory performance in Duchenne muscular dystrophy: Clinical manifestations and lessons from animal models. Exp Physiol 2024; 109:1426-1445. [PMID: 39023735 PMCID: PMC11363095 DOI: 10.1113/ep091967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal genetic neuromuscular disease. Lack of dystrophin in skeletal muscles leads to intrinsic weakness, injury, subsequent degeneration and fibrosis, decreasing contractile function. Dystropathology eventually presents in all inspiratory and expiratory muscles of breathing, severely curtailing their critical function. In people with DMD, premature death is caused by respiratory or cardiac failure. There is an urgent need to develop therapies that improve quality of life and extend life expectancy in DMD. Surprisingly, there is a dearth of information on respiratory control in animal models of DMD, and respiratory outcome measures are often limited or absent in clinical trials. Characterization of respiratory performance in murine and canine models has revealed extensive remodelling of the diaphragm, the major muscle of inspiration. However, significant compensation by extradiaphragmatic muscles of breathing is evident in early disease, contributing to preservation of peak respiratory system performance. Loss of compensation afforded by accessory muscles in advanced disease is ultimately associated with compromised respiratory performance. A new and potentially more translatable murine model of DMD, the D2.mdx mouse, has recently been developed. Respiratory performance in D2.mdx mice is yet to be characterized fully. However, based on histopathological features, D2.mdx mice might serve as useful preclinical models, facilitating the testing of new therapeutics that rescue respiratory function. This review summarizes the pathophysiological mechanisms associated with DMD both in humans and in animal models, with a focus on breathing. We consider the translational value of each model to human DMD and highlight the urgent need for comprehensive characterization of breathing in representative preclinical models to better inform human trials.
Collapse
|
6
|
Escobar-Huertas JF, Vaca-González JJ, Guevara JM, Ramirez-Martinez AM, Trabelsi O, Garzón-Alvarado DA. Duchenne and Becker muscular dystrophy: Cellular mechanisms, image analysis, and computational models: A review. Cytoskeleton (Hoboken) 2024; 81:269-286. [PMID: 38224155 DOI: 10.1002/cm.21826] [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: 05/24/2023] [Revised: 11/21/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
The muscle is the principal tissue that is capable to transform potential energy into kinetic energy. This process is due to the transformation of chemical energy into mechanical energy to enhance the movements and all the daily activities. However, muscular tissues can be affected by some pathologies associated with genetic alterations that affect the expression of proteins. As the muscle is a highly organized structure in which most of the signaling pathways and proteins are related to one another, pathologies may overlap. Duchenne muscular dystrophy (DMD) is one of the most severe muscle pathologies triggering degeneration and muscle necrosis. Several mathematical models have been developed to predict muscle response to different scenarios and pathologies. The aim of this review is to describe DMD and Becker muscular dystrophy in terms of cellular behavior and molecular disorders and to present an overview of the computational models implemented to understand muscle behavior with the aim of improving regenerative therapy.
Collapse
Affiliation(s)
- J F Escobar-Huertas
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Juan Jairo Vaca-González
- Escuela de pregrado, Dirección Académica, Vicerrectoría de Sede, Universidad Nacional de Colombia, Sede la Paz, Cesar, Colombia
| | - Johana María Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Olfa Trabelsi
- Université de technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - D A Garzón-Alvarado
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
7
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
8
|
Riddell DO, Hildyard JCW, Harron RCM, Taylor-Brown F, Kornegay JN, Wells DJ, Piercy RJ. Longitudinal assessment of skeletal muscle functional mechanics in the DE50-MD dog model of Duchenne muscular dystrophy. Dis Model Mech 2023; 16:dmm050395. [PMID: 38050706 PMCID: PMC10753191 DOI: 10.1242/dmm.050395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin (DMD) gene, is associated with fatal muscle degeneration and atrophy. Patients with DMD have progressive reductions in skeletal muscle strength and resistance to eccentric muscle stretch. Using the DE50-MD dog model of DMD, we assessed tibiotarsal joint (TTJ) flexor and extensor force dynamics, and the resistance of dystrophic muscle to eccentric stretch. Male DE50-MD and wild-type (WT) dogs were analysed every 3 months until 18 months of age. There was an age-associated decline in eccentric contraction resistance in DE50-MD TTJ flexors that discriminated, with high statistical power, WT from DE50-MD individuals. For isometric contraction, at the majority of timepoints, DE50-MD dogs had lower maximum absolute and relative TTJ flexor force, reduced TTJ muscle contraction times and prolonged relaxation compared to those in WT dogs. Cranial tibial muscles, the primary TTJ flexor, of 18-month-old DE50-MD dogs had significant numbers of regenerating fibres as expected, but also fewer type I fibres and more hybrid fibres than those in WT dogs. We conclude that these parameters, in particular, the eccentric contraction decrement, could be used as objective outcome measures for pre-clinical assessment in DE50-MD dogs.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW10TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW10TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW10TU, UK
| | - Frances Taylor-Brown
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW10TU, UK
| | - Joe N. Kornegay
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW10TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW10TU, UK
| |
Collapse
|
9
|
Jankelunas L, Murthy VD, Chen AV, Minor KM, Friedenberg SG, Cullen JN, Guo LT, Mickelson JR, Shelton GD. Novel COL6A3 frameshift variant in American Staffordshire Terrier dogs with Ullrich-like congenital muscular dystrophy. J Vet Intern Med 2023; 37:2504-2509. [PMID: 37706358 PMCID: PMC10658572 DOI: 10.1111/jvim.16862] [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: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Two (male and female) 10-month-old American Staffordshire Terrier littermates presented for progressive weakness, joint contracture, and distal limb joint hyperlaxity beginning around 6 months of age. Neurological examination, serum creatine kinase activity, infectious disease titers, cerebrospinal fluid analysis, and electrodiagnostic testing were performed. Muscle biopsies were collected for histopathology and immunofluorescence staining for localization of dystrophy associated proteins. Whole-genome sequencing (WGS) was performed on 1 affected dog. Variants were compared to a database of 671 unaffected dogs of multiple breeds. Histopathology confirmed a dystrophic phenotype and immunofluorescence staining of muscle cryosections revealed an absence of staining for collagen-6. WGS identified a homozygous 1 bp deletion in the COL6A3 gene, unique to the first affected dog. Sanger sequencing confirmed the homozygous presence of the frameshift variant in both affected dogs. This report describes the clinical features and most likely genetic basis of an Ullrich-like recessively inherited form of congenital muscular dystrophy in American Staffordshire Terriers.
Collapse
Affiliation(s)
- Leanne Jankelunas
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Vishal D. Murthy
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Annie V. Chen
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Katie M. Minor
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Jonah N. Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Ling T. Guo
- Department of Pathology, School of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - James R. Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - G. Diane Shelton
- Department of Pathology, School of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
10
|
Shelton GD, Minor KM, Friedenberg SG, Cullen JN, Guo LT, Mickelson JR. Current Classification of Canine Muscular Dystrophies and Identification of New Variants. Genes (Basel) 2023; 14:1557. [PMID: 37628610 PMCID: PMC10454810 DOI: 10.3390/genes14081557] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
The spectrum of canine muscular dystrophies has rapidly grown with the recent identification of several more affected breeds and associated mutations. Defects include those in genes and protein products associated with the sarcolemma (dystrophin deficient X-linked muscular dystrophy and sarcoglycan-deficient limb-girdle muscular dystrophy) and with the extracellular matrix (collagen 6, laminin α2, and α-dystroglycan-deficient congenital muscular dystrophies). With the increasing application of whole genome sequencing and whole exome sequencing, the clinical and pathological spectra associated with specific neuromuscular genetic defects are constantly evolving. In this report, we provide a brief overview of the current status of gene defects reported in canine muscular dystrophies. We also report the causative mutations for novel forms of X-linked muscular dystrophy in Brittany spaniels and in a French bulldog.
Collapse
Affiliation(s)
- G. Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA;
| | - Katie M. Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (K.M.M.); (J.R.M.)
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (S.G.F.); (J.N.C.)
| | - Jonah N. Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (S.G.F.); (J.N.C.)
| | - Ling T. Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA;
| | - James R. Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA; (K.M.M.); (J.R.M.)
| |
Collapse
|
11
|
de Sousa AA, Rigby Dames BA, Graff EC, Mohamedelhassan R, Vassilopoulos T, Charvet CJ. Going beyond established model systems of Alzheimer's disease: companion animals provide novel insights into the neurobiology of aging. Commun Biol 2023; 6:655. [PMID: 37344566 PMCID: PMC10284893 DOI: 10.1038/s42003-023-05034-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/12/2023] [Indexed: 06/23/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by brain plaques, tangles, and cognitive impairment. AD is one of the most common age-related dementias in humans. Progress in characterizing AD and other age-related disorders is hindered by a perceived dearth of animal models that naturally reproduce diseases observed in humans. Mice and nonhuman primates are model systems used to understand human diseases. Still, these model systems lack many of the biological characteristics of Alzheimer-like diseases (e.g., plaques, tangles) as they grow older. In contrast, companion animal models (cats and dogs) age in ways that resemble humans. Both companion animal models and humans show evidence of brain atrophy, plaques, and tangles, as well as cognitive decline with age. We embrace a One Health perspective, which recognizes that the health of humans is connected to those of animals, and we illustrate how such a perspective can work synergistically to enhance human and animal health. A comparative biology perspective is ideally suited to integrate insights across veterinary and human medical disciplines and solve long-standing problems in aging.
Collapse
Affiliation(s)
- Alexandra A de Sousa
- Centre for Health and Cognition, Bath Spa University, Bath, UK
- Department of Psychology, University of Bath, Bath, UK
| | - Brier A Rigby Dames
- Department of Psychology, University of Bath, Bath, UK
- Department of Computer Science, University of Bath, Bath, UK
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Emily C Graff
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Rania Mohamedelhassan
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Tatianna Vassilopoulos
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.
| |
Collapse
|
12
|
Birch SM, Lawlor MW, Conlon TJ, Guo LJ, Crudele JM, Hawkins EC, Nghiem PP, Ahn M, Meng H, Beatka MJ, Fickau BA, Prieto JC, Styner MA, Struharik MJ, Shanks C, Brown KJ, Golebiowski D, Bettis AK, Balog-Alvarez CJ, Clement N, Coleman KE, Corti M, Pan X, Hauschka SD, Gonzalez JP, Morris CA, Schneider JS, Duan D, Chamberlain JS, Byrne BJ, Kornegay JN. Assessment of systemic AAV-microdystrophin gene therapy in the GRMD model of Duchenne muscular dystrophy. Sci Transl Med 2023; 15:eabo1815. [PMID: 36599002 PMCID: PMC11107748 DOI: 10.1126/scitranslmed.abo1815] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by the absence of dystrophin, a membrane-stabilizing protein encoded by the DMD gene. Although mouse models of DMD provide insight into the potential of a corrective therapy, data from genetically homologous large animals, such as the dystrophin-deficient golden retriever muscular dystrophy (GRMD) model, may more readily translate to humans. To evaluate the clinical translatability of an adeno-associated virus serotype 9 vector (AAV9)-microdystrophin (μDys5) construct, we performed a blinded, placebo-controlled study in which 12 GRMD dogs were divided among four dose groups [control, 1 × 1013 vector genomes per kilogram (vg/kg), 1 × 1014 vg/kg, and 2 × 1014 vg/kg; n = 3 each], treated intravenously at 3 months of age with a canine codon-optimized microdystrophin construct, rAAV9-CK8e-c-μDys5, and followed for 90 days after dosing. All dogs received prednisone (1 milligram/kilogram) for a total of 5 weeks from day -7 through day 28. We observed dose-dependent increases in tissue vector genome copy numbers; μDys5 protein in multiple appendicular muscles, the diaphragm, and heart; limb and respiratory muscle functional improvement; and reduction of histopathologic lesions. As expected, given that a truncated dystrophin protein was generated, phenotypic test results and histopathologic lesions did not fully normalize. All administrations were well tolerated, and adverse events were not seen. These data suggest that systemically administered AAV-microdystrophin may be dosed safely and could provide therapeutic benefit for patients with DMD.
Collapse
Affiliation(s)
- Sharla M. Birch
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | | | - Thomas J. Conlon
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Lee-Jae Guo
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | | | - Eleanor C. Hawkins
- North Carolina State University, College of Veterinary Medicine, Raleigh, NC; 27606
| | - Peter P. Nghiem
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Mihye Ahn
- University of Nevada-Reno, Reno, NV; 89557
| | - Hui Meng
- Medical College of Wisconsin, Milwaukee, WI; 53226
| | | | | | | | | | | | | | | | | | - Amanda K. Bettis
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Cynthia J. Balog-Alvarez
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Nathalie Clement
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Kirsten E. Coleman
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Manuela Corti
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Xiufang Pan
- University of Missouri, School of Medicine, Columbia, MO 65212
| | | | | | | | | | - Dongsheng Duan
- University of Missouri, School of Medicine, Columbia, MO 65212
| | | | - Barry J. Byrne
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Joe. N. Kornegay
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| |
Collapse
|
13
|
Chey YCJ, Arudkumar J, Aartsma-Rus A, Adikusuma F, Thomas PQ. CRISPR applications for Duchenne muscular dystrophy: From animal models to potential therapies. WIREs Mech Dis 2023; 15:e1580. [PMID: 35909075 PMCID: PMC10078488 DOI: 10.1002/wsbm.1580] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
CRISPR gene-editing technology creates precise and permanent modifications to DNA. It has significantly advanced our ability to generate animal disease models for use in biomedical research and also has potential to revolutionize the treatment of genetic disorders. Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disease that could potentially benefit from the development of CRISPR therapy. It is commonly associated with mutations that disrupt the reading frame of the DMD gene that encodes dystrophin, an essential scaffolding protein that stabilizes striated muscles and protects them from contractile-induced damage. CRISPR enables the rapid generation of various animal models harboring mutations that closely simulates the wide variety of mutations observed in DMD patients. These models provide a platform for the testing of sequence-specific interventions like CRISPR therapy that aim to reframe or skip DMD mutations to restore functional dystrophin expression. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,CSIRO Synthetic Biology Future Science Platform, Canberra, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Stevens R, Kanazono S, Petesch S, Guo LT, Shelton GD. Dystrophin-Deficient Muscular Dystrophy in Two Male Juvenile Brittanys. J Am Anim Hosp Assoc 2022; 58:292-296. [PMID: 36315862 DOI: 10.5326/jaaha-ms-7255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 06/16/2023]
Abstract
A 6 mo old and a 7 mo old male intact Brittany were presented for progressive exercise intolerance, failure to grow, and dysphagia. Creatine kinase activity was markedly and persistently elevated in both dogs. Based on the neurological examination, clinical signs localized to the neuromuscular system. Electromyography revealed complex repetitive discharges in multiple muscle groups. Immunofluorescence of biopsies confirmed dystrophin-deficient muscular dystrophy. This is the first report describing dystrophin-deficient muscular dystrophy in the Brittany breed. Currently, no specific therapies are available for this form of myopathy. The presence of dystrophin deficiency in the two dogs suggests an inherited myopathy rather than a spontaneous mutation. The location of the dogs in the United States and Japan suggests a wide distribution of this dystrophy and should alert clinicians to the existence of this myopathy in the Brittany breed. A mutation in the DMD gene has not yet been identified.
Collapse
Affiliation(s)
- Rebecca Stevens
- From Burlington Emergency and Veterinary Specialists, Williston, Vermont (R.S.)
| | | | - Scott Petesch
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Matthew J. Ryan Veterinary Hospital, Philadelphia, Pennsylvania (S.P.)
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California (L.T.G., G.D.S.)
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California (L.T.G., G.D.S.)
| |
Collapse
|
15
|
A Nonsense Variant in the DMD Gene Causes X-Linked Muscular Dystrophy in the Maine Coon Cat. Animals (Basel) 2022; 12:ani12212928. [PMID: 36359052 PMCID: PMC9653713 DOI: 10.3390/ani12212928] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Feline dystrophin-deficient muscular dystrophy (ddMD) is a fatal disease characterized by progressive weakness and degeneration of skeletal muscles and is caused by variants in the DMD gene. To date, only two feline causal variants have been identified. This study reports two cases of male Maine coon siblings that presented with muscular hypertrophy, growth retardation, weight loss, and vomiting. (2) Both cats were clinically examined and histopathology and immunofluorescent staining of the affected muscle was performed. DMD mRNA was sequenced to identify putative causal variants. (3) Both cats showed a significant increase in serum creatine kinase activity. Electromyography and histopathological examination of the muscle samples revealed abnormalities consistent with a dystrophic phenotype. Immunohistochemical testing revealed the absence of dystrophin, confirming the diagnosis of dystrophin-deficient muscular dystrophy. mRNA sequencing revealed a nonsense variant in exon 11 of the feline DMD gene, NC_058386.1 (XM_045050794.1): c.1180C > T (p.(Arg394*)), which results in the loss of the majority of the dystrophin protein. Perfect X-linked segregation of the variant was established in the pedigree. (4) ddMD was described for the first time in the Maine coon and the c.1180C>T variant was confirmed as the causal variant.
Collapse
|
16
|
Shelton GD, Minor KM, Vieira NM, Kunkel LM, Friedenberg SG, Cullen JN, Guo LT, Zatz M, Mickelson JR. Tandem duplication within the DMD gene in Labrador retrievers with a mild clinical phenotype. Neuromuscul Disord 2022; 32:836-841. [PMID: 36041985 PMCID: PMC10040250 DOI: 10.1016/j.nmd.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/30/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022]
Abstract
A form of dystrophinopathy with mild or subclinical neuromuscular signs has been previously reported in a family of Labrador retrievers. Markedly and persistently elevated creatine kinase activity was first noted at 6 months of age. Skeletal muscle biopsies revealed a dystrophic phenotype, with dystrophin non-detectable on western blotting and immunohistochemical staining, and with increased utrophin expression. In this report we demonstrate with western blotting that α-dystroglycan is present at essentially normal levels. Whole genome sequencing has also now revealed an approximately 400kb tandem genomic DNA duplication including exons 2-7 of the DMD gene that was inserted into intron 7 of the wild type gene. Skeletal muscle cDNA from 2 cases contained DMD transcripts as expected from an in-frame properly-spliced exon 2-7 tandem insertion. A similar 5' duplication involving DMD exons 2-7 has been reported in a human family with dilated cardiomyopathy but without skeletal myopathy. This is the 3rd confirmed mutation in the DMD gene in Labrador retrievers.
Collapse
Affiliation(s)
- G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, LaJolla, CA, USA.
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Natassia M Vieira
- The Division of Genetics and Genomics, Boston Children's Hospital, Department of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
| | - Louis M Kunkel
- The Division of Genetics and Genomics, Boston Children's Hospital, Department of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Jonah N Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, LaJolla, CA, USA
| | - Mayana Zatz
- Human Genome and Stem Cell Center, Biosciences Institute, University of Sao Paulo, Brazil
| | - James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| |
Collapse
|
17
|
Hart CC, Lee YI, Hammers DW, Sweeney HL. Evaluation of the DBA/2J mouse as a potential background strain for genetic models of cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 1:100012. [PMID: 37206988 PMCID: PMC10195103 DOI: 10.1016/j.jmccpl.2022.100012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The potential use of the D2.mdx mouse (the mdx mutation on the DBA/2J genetic background) as a preclinical model of the cardiac aspects of Duchenne muscular dystrophy (DMD) has been criticized based on speculation that the DBA/2J genetic background displays an inherent hypertrophic cardiomyopathy (HCM) phenotype. Accordingly, the goal of the current study was to further examine the cardiac status of this mouse strain over a 12-month period to determine if observable signs of HCM develop, including histopathology and pathological enlargement of the myocardium. Previous reports have documented heightened TGFβ signaling in the DBA2/J striated muscles, as compared to the C57 background, which, as expected, is manifested as increased cardiomyocyte size, wall thickness, and heart mass as compared to the C57 background. While normalized heart mass is larger in the DBA/2J mice, compared to age-matched C57/BL10 mice, both strains similarly increase in size from 4 to 12 months of age. We also report that DBA/2J mice contain equivalent amounts of left ventricular collagen as healthy canine and human samples. In a longitudinal echocardiography study, neither sedentary nor exercised DBA/2J mice demonstrated left ventricular wall thickening or cardiac functional deficits. In summary, we find no evidence of HCM, nor any other cardiac pathology, and thus propose that it is an appropriate background strain for genetic modeling of cardiac diseases, including the cardiomyopathy associated with DMD.
Collapse
Affiliation(s)
| | | | | | - H. Lee Sweeney
- Corresponding author at: 1200 Newell, Dr. ARB R5-216, Gainesville, FL 32610-0267, United States of America. (H.L. Sweeney)
| |
Collapse
|
18
|
Ullal TV, Marks SL, Belafsky PC, Conklin JL, Pandolfino JE. A Comparative Assessment of the Diagnosis of Swallowing Impairment and Gastroesophageal Reflux in Canines and Humans. Front Vet Sci 2022; 9:889331. [PMID: 35754550 PMCID: PMC9228035 DOI: 10.3389/fvets.2022.889331] [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: 03/04/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
Swallowing impairment is a highly prevalent and clinically significant problem affecting people and dogs. There are myriad causes of swallowing impairment of which gastroesophageal reflux is the most common in both species. Similarities in anatomy and physiology between humans and canines results in analogous swallowing disorders including cricopharyngeus muscle achalasia, esophageal achalasia, hiatal herniation, and gastroesophageal reflux with secondary esophagitis and esophageal dysmotility. Accordingly, the diagnostic approach to human and canine patients with swallowing impairment is similar. Diagnostic procedures such as swallowing fluoroscopy, high-resolution manometry, pH/impedance monitoring, and endolumenal functional luminal imaging probe can be performed in both species; however, nasofacial conformation, increased esophageal length, and the difficulty of completing several of these procedures in awake dogs are inherent challenges that need to be considered. Human patients can convey their symptoms and respond to verbal cues, whereas veterinarians must rely on clinical histories narrated by pet owners followed by comprehensive physical examination and observation of the animal eating different food consistencies and drinking water. Dogs may also be unwilling to drink or eat in the hospital setting and may be resistant to physical restraint during diagnostic procedures. Despite the species differences and diagnostic challenges, dogs are a natural animal model for many oropharyngeal and esophageal disorders affecting people, which presents a tremendous opportunity for shared learnings. This manuscript reviews the comparative aspects of esophageal anatomy and physiology between humans and canines, summarizes the diagnostic assessment of swallowing impairment in both species, and discusses future considerations for collaborative medicine and translational research.
Collapse
Affiliation(s)
- Tarini V Ullal
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Stanley L Marks
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Peter C Belafsky
- Department of Otolaryngology, Center for Voice and Swallowing, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jeffrey L Conklin
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, UCLA Robert G. Kardashian Center for Esophageal Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - John E Pandolfino
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
19
|
Nance RL, Sajib AM, Smith BF. Canine models of human cancer: Bridging the gap to improve precision medicine. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:67-99. [PMID: 35595353 DOI: 10.1016/bs.pmbts.2021.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dogs are remarkable, adaptable, and dependable creatures that have evolved alongside humans while contributing tremendously to our survival. Our canine companions share many similarities to human disease, particularly cancer. With the advancement of next-generation sequencing technology, we are beginning to unravel the complexity of cancer and the vast intra- and intertumoral heterogeneity that makes treatment difficult. Consequently, precision medicine has emerged as a therapeutic approach to improve patient survival by evaluating and classifying an individual tumor's molecular profile. Many canine and human cancers share striking similarities in terms of genotypic, phenotypic, clinical, and histological presentations. Dogs are superior to rodent models of cancer because they are a naturally heterogeneous population in which tumors occur spontaneously, are exposed to similar environmental conditions, and show more similarities in key modulators of tumorigenesis and clinical response, including the immune system, drug metabolism, and gut microbiome. In this chapter, we will explore various canine models of human cancers and emphasize the dog's critical role in advancing precision medicine and improving the survival of both man and man's best friend.
Collapse
Affiliation(s)
- Rebecca L Nance
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Abdul Mohin Sajib
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Bruce F Smith
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States.
| |
Collapse
|
20
|
Tao YX. Mutations in melanocortin-4 receptor: From fish to men. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:215-257. [PMID: 35595350 DOI: 10.1016/bs.pmbts.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Melanocortin-4 receptor (MC4R), expressed abundantly in the hypothalamus, is a critical regulator of energy homeostasis, including both food intake and energy expenditure. Shortly after the publication in 1997 of the Mc4r knockout phenotypes in mice, including increased food intake and severe obesity, the first mutations in MC4R were reported in humans in 1998. Studies in the subsequent two decades have established MC4R mutation as the most common monogenic form of obesity, especially in early-onset severe obesity. Studies in animals, from fish to mammals, have established the conserved physiological roles of MC4R in all vertebrates in regulating energy balance. Drug targeting MC4R has been recently approved for treating morbid genetic obesity. How the MC4R can be exploited for animal production is highly worthy of active investigation.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.
| |
Collapse
|
21
|
Stirm M, Fonteyne LM, Shashikadze B, Stöckl JB, Kurome M, Keßler B, Zakhartchenko V, Kemter E, Blum H, Arnold GJ, Matiasek K, Wanke R, Wurst W, Nagashima H, Knieling F, Walter MC, Kupatt C, Fröhlich T, Klymiuk N, Blutke A, Wolf E. Pig models for Duchenne muscular dystrophy – from disease mechanisms to validation of new diagnostic and therapeutic concepts. Neuromuscul Disord 2022; 32:543-556. [DOI: 10.1016/j.nmd.2022.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
|
22
|
Schneider SM, Sansom GT, Guo LJ, Furuya S, Weeks BR, Kornegay JN. Natural History of Histopathologic Changes in Cardiomyopathy of Golden Retriever Muscular Dystrophy. Front Vet Sci 2022; 8:759585. [PMID: 35252412 PMCID: PMC8892215 DOI: 10.3389/fvets.2021.759585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked inherited myopathy that causes progressive skeletal and cardiac muscle disease. Heart lesions were described in the earliest DMD reports, and cardiomyopathy is now the leading cause of death. However, diagnostics and treatment for cardiomyopathy have lagged behind those for appendicular and respiratory skeletal muscle disease. Most animal model studies have been done in the mdx mouse, which has a relatively mild form of cardiomyopathy. Dogs with the genetically homologous condition, Golden Retriever muscular dystrophy (GRMD), develop progressive cardiomyopathy analogous to that seen in DMD. Previous descriptive studies of GRMD cardiomyopathy have mostly been limited to selective sampling of the hearts from young dogs. Methods and Results We systematically assessed cardiac lesions in 31 GRMD and carrier dogs aged 3 to 76 months and a separate cohort of 2–10-year-old normal hounds. Both semi-quantitative lesion scoring and quantitation of the cross-sectional area of fibrosis distinguished dogs with GRMD disease from normal dogs. The carriers generally had intermediate involvement but had even greater fibrosis than GRMD dogs. Fatty infiltration was the most prominent feature in some older GRMD dogs. Vascular hypertrophy was increased in GRMD dogs and correlated positively with lesion severity. Purkinje fiber vacuolation was also increased but did not correlate with lesion severity. Histopathologic changes correlated with late gadolinium enhancement on cardiac MRI. Conclusion These features are generally compatible with those of DMD and further validate GRMD as a useful model to study cardiomyopathy pathogenesis and treatment. Additionally, the nature of some degenerative lesions suggests that functional hypoxia or non-thrombotic ischemia may contribute to disease progression.
Collapse
Affiliation(s)
- Sarah M. Schneider
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- *Correspondence: Sarah M. Schneider
| | - Garett T. Sansom
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, United States
| | - Lee-Jae Guo
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Shinji Furuya
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Brad R. Weeks
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Joe N. Kornegay
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
23
|
Rutledge AM, Guo LJ, Lord LE, Leal AR, Deramus J, López SM, Russell A, Nghiem PP. Comprehensive assessment of physical activity correlated with muscle function in canine Duchenne muscular dystrophy. Ann Phys Rehabil Med 2021; 65:101611. [PMID: 34844013 DOI: 10.1016/j.rehab.2021.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Alexis M Rutledge
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - Lee-Jae Guo
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - Laney E Lord
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - Amanda R Leal
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - John Deramus
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - Sara Mata López
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458
| | - Alan Russell
- Edgewise Therapeutics, 3415 Colorado Ave, Boulder, CO 80303
| | - Peter P Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458.
| |
Collapse
|
24
|
Shelton GD, Minor KM, Guo LT, Friedenberg SG, Cullen JN, Hord JM, Venzke D, Anderson ME, Devereaux M, Prouty SJ, Handelman C, Campbell KP, Mickelson JR. Muscular dystrophy-dystroglycanopathy in a family of Labrador retrievers with a LARGE1 mutation. Neuromuscul Disord 2021; 31:1169-1178. [PMID: 34654610 PMCID: PMC8963908 DOI: 10.1016/j.nmd.2021.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
Alpha-dystroglycan (αDG) is a highly glycosylated cell surface protein with a significant role in cell-to-extracellular matrix interactions in muscle. αDG interaction with extracellular ligands relies on the activity of the LARGE1 glycosyltransferase that synthesizes and extends the heteropolysaccharide matriglycan. Abnormalities in αDG glycosylation and formation of matriglycan are the pathogenic mechanisms for the dystroglycanopathies, a group of congenital muscular dystrophies. Muscle biopsies were evaluated from related 6-week-old Labrador retriever puppies with poor suckling, small stature compared to normal litter mates, bow-legged stance and markedly elevated creatine kinase activities. A dystrophic phenotype with marked degeneration and regeneration, multifocal mononuclear cell infiltration and endomysial fibrosis was identified on muscle cryosections. Single nucleotide polymorphism (SNP) array genotyping data on the family members identified three regions of homozygosity in 4 cases relative to 8 controls. Analysis of whole genome sequence data from one of the cases identified a stop codon mutation in the LARGE1 gene that truncates 40% of the protein. Immunofluorescent staining and western blotting demonstrated the absence of matriglycan in skeletal muscle and heart from affected dogs. Compared to control, LARGE enzyme activity was not detected. This is the first report of a dystroglycanopathy in dogs.
Collapse
Affiliation(s)
- G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709 United States.
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709 United States
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Jonah N Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Jeffrey M Hord
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - David Venzke
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Mary E Anderson
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Megan Devereaux
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Sally J Prouty
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Caryl Handelman
- Veterinary Housecalls of Long Island, Commack, NY 11725 United States
| | - Kevin P Campbell
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| |
Collapse
|
25
|
Zhang Y, Nishiyama T, Olson EN, Bassel-Duby R. CRISPR/Cas correction of muscular dystrophies. Exp Cell Res 2021; 408:112844. [PMID: 34571006 PMCID: PMC8530959 DOI: 10.1016/j.yexcr.2021.112844] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/10/2021] [Accepted: 09/23/2021] [Indexed: 12/28/2022]
Abstract
Muscular dystrophies are a heterogeneous group of monogenic neuromuscular disorders which lead to progressive muscle loss and degeneration of the musculoskeletal system. The genetic causes of muscular dystrophies are well characterized, but no effective treatments have been developed so far. The discovery and application of the CRISPR/Cas system for genome editing offers a new path for disease treatment with the potential to permanently correct genetic mutations. The post-mitotic and multinucleated features of skeletal muscle provide an ideal target for CRISPR/Cas therapeutic genome editing because correction of a subpopulation of nuclei can provide benefit to the whole myofiber. In this review, we provide an overview of the CRISPR/Cas system and its derivatives in genome editing, proposing potential CRISPR/Cas-based therapies to correct diverse muscular dystrophies, and we discuss challenges for translating CRISPR/Cas genome editing to a viable therapy for permanent correction of muscular dystrophies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takahiko Nishiyama
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
26
|
Swiderski K, Lynch GS. Murine models of Duchenne muscular dystrophy: is there a best model? Am J Physiol Cell Physiol 2021; 321:C409-C412. [PMID: 34260298 DOI: 10.1152/ajpcell.00212.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Kristy Swiderski
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Florczyk-Soluch U, Polak K, Dulak J. The multifaceted view of heart problem in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:5447-5468. [PMID: 34091693 PMCID: PMC8257522 DOI: 10.1007/s00018-021-03862-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
Dystrophin is a large protein serving as local scaffolding repetitively bridging cytoskeleton and the outside of striated muscle cell. As such dystrophin is a critical brick primarily in dystrophin-associated protein complex (DAGC) and in a larger submembranous unit, costamere. Accordingly, the lack of functional dystrophin laying at the root of Duchenne muscular dystrophy (DMD) drives sarcolemma instability. From this point on, the cascade inevitably leading to the death of myocyte begins. In cardiomyocytes, intracellular calcium overload and related mitochondrial-mediated cell death mainly contribute to myocardial dysfunction and dilation while other protein dysregulation and/or mislocalization may affect electrical conduction system and favor arrhythmogenesis. Although clinically DMD manifests as progressive muscle weakness and skeletal muscle symptoms define characteristic of DMD, it is the heart problem the biggest challenge that most often develop in the form of dilated cardiomyopathy (DCM). Current standards of treatment and recent progress in respiratory care, introduced in most settings in the 1990s, have improved quality of life and median life expectancy to 4th decade of patient's age. At the same time, cardiac causes of death related to DMD increases. Despite preventive and palliative cardiac treatments available, the prognoses remain poor. Direct therapeutic targeting of dystrophin deficiency is critical, however, hindered by the large size of the dystrophin cDNA and/or stochastic, often extensive genetic changes in DMD gene. The correlation between cardiac involvement and mutations affecting specific dystrophin isoforms, may provide a mutation-specific cardiac management and novel therapeutic approaches for patients with CM. Nonetheless, the successful cardiac treatment poses a big challenge and may require combined therapy to combat dystrophin deficiency and its after-effects (critical in DMD pathogenesis). This review locates the multifaceted heart problem in the course of DMD, balancing the insights into basic science, translational efforts and clinical manifestation of dystrophic heart disease.
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
28
|
Musculoskeletal magnetic resonance imaging in the DE50-MD dog model of Duchenne muscular dystrophy. Neuromuscul Disord 2021; 31:736-751. [PMID: 34384671 PMCID: PMC8449064 DOI: 10.1016/j.nmd.2021.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022]
Abstract
Normalized muscle volumes distinguish between wild type and DE50-MD dogs. Global muscle T2 signal intensities discriminate between wild type and DE50-MD dogs. Musculoskeletal changes detected by MRI reflect those seen in human DMD patients. Musculoskeletal MRI in this model will be useful to assess treatment efficacy.
The DE50-MD canine model of Duchenne muscular dystrophy (DMD) has a dystrophin gene splice site mutation causing deletion of exon 50, an out-of-frame transcript and absence of dystrophin expression in striated muscles. We hypothesized that the musculoskeletal phenotype of DE50-MD dogs could be detected using Magnetic Resonance Imaging (MRI), that it would progress with age and that it would reflect those in other canine models and DMD patients. 15 DE50-MD and 10 age-matched littermate wild type (WT) male dogs underwent MRI every 3 months from 3 to 18 months of age. Normalized muscle volumes, global muscle T2 and ratio of post- to pre-gadolinium T1-weighted SI were evaluated in 7 pelvic limb and 4 lumbar muscles bilaterally. DE50-MD dogs, compared to WT, had smaller volumes in all muscles, except the cranial sartorius; global muscle T2 was significantly higher in DE50-MD dogs compared to WT. Muscle volumes plateaued and global muscle T2 decreased with age. Normalized muscle volumes and global muscle T2 revealed significant differences between groups longitudinally and should be useful to determine efficacy of therapeutics in this model with suitable power and low sample sizes. Musculoskeletal changes reflect those of DMD patients and other dog models.
Collapse
|
29
|
Gaina G, Popa (Gruianu) A. Muscular dystrophy: Experimental animal models and therapeutic approaches (Review). Exp Ther Med 2021; 21:610. [PMID: 33936267 PMCID: PMC8082581 DOI: 10.3892/etm.2021.10042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
The muscular dystrophies are a heterogeneous group of genetically inherited diseases characterized by muscle weakness and progressive wasting, which can cause premature death in severe forms. Although >30 years have passed since the identification of the first protein involved in a type of muscular dystrophy, there is no effective treatment for these disabling disorders. In the last decade, several novel therapeutic approaches have been developed and investigated as promising therapeutic approaches aimed to ameliorate the dystrophic phenotype either by restoring dystrophin expression or by compensating for dystrophin deficiency. Concurrently, with the development of therapeutic approaches, in addition to naturally occurring animal models, a wide range of genetically engineered animal models has been generated. The use of animals as models of muscular dystrophies has greatly improved the understanding of the pathogenicity of these diseases and has proven useful in gene therapy studies. In this review, we summarize these latest innovative therapeutic approaches to muscular dystrophies and the usefulness of the various most common experimental animal models.
Collapse
Affiliation(s)
- Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Alexandra Popa (Gruianu)
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Animal Production and Public Health, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania
| |
Collapse
|
30
|
Martin PT, Zygmunt DA, Ashbrook A, Hamilton S, Packer D, Birch SM, Bettis AK, Balog-Alvarez CJ, Guo LJ, Nghiem PP, Kornegay JN. Short-term treatment of golden retriever muscular dystrophy (GRMD) dogs with rAAVrh74.MHCK7.GALGT2 induces muscle glycosylation and utrophin expression but has no significant effect on muscle strength. PLoS One 2021; 16:e0248721. [PMID: 33770101 PMCID: PMC7997012 DOI: 10.1371/journal.pone.0248721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/03/2021] [Indexed: 11/30/2022] Open
Abstract
We have examined the effects of intravenous (IV) delivery of rAAVrh74.MHCK7.GALGT2 in the golden retriever muscular dystrophy (GRMD) model of Duchenne Muscular Dystrophy (DMD). After baseline testing, GRMD dogs were treated at 3 months of age and reassessed at 6 months. This 3–6 month age range is a period of rapid disease progression, thus offering a relatively short window to establish treatment efficacy. Measures analyzed included muscle AAV transduction, GALGT2 transgene expression, GALGT2-induced glycosylation, muscle pathology, and muscle function. A total of five dogs were treated, 4 at 2x1014vg/kg and one at 6x1014vgkg. The 2x1014vg/kg dose led to transduction of regions of the heart with 1–3 vector genomes (vg) per nucleus, while most skeletal muscles were transduced with 0.25–0.5vg/nucleus. GALGT2-induced glycosylation paralleled levels of myofiber vg transduction, with about 90% of cardiomyocytes having increased glycosylation versus 20–35% of all myofibers across the skeletal muscles tested. Conclusions from phenotypic testing were limited by the small number of dogs. Treated dogs had less pronounced fibrosis and overall lesion severity when compared to control groups, but surprisingly no significant changes in limb muscle function measures. GALGT2-treated skeletal muscle and heart had elevated levels of utrophin protein expression and GALGT2-induced expression of glycosylated α dystroglycan, providing further evidence of a treatment effect. Serum chemistry, hematology, and cardiac function measures were largely unchanged by treatment. Cumulatively, these data show that short-term intravenous treatment of GRMD dogs with rAAVrh74.MHCK7.GALGT2 at high doses can induce muscle glycosylation and utrophin expression and may be safe over a short 3-month interval, but that such treatments had only modest effects on muscle pathology and did not significantly improve muscle strength.
Collapse
Affiliation(s)
- Paul T. Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- * E-mail:
| | - Deborah A. Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Sonia Hamilton
- Neuroscience Undergraduate Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Davin Packer
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Sharla M. Birch
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Amanda K. Bettis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Cynthia J. Balog-Alvarez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Lee-Jae Guo
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Peter P. Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Joe N. Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| |
Collapse
|
31
|
Fortin JS, Hakim CH, Korte S, Yang NN, Fitzgerald SD, Johnson GC, Smith BF, Duan D. Widespread severe myodegeneration in a compound heterozygote female dog with dystrophin deficiency. Vet Med Sci 2021; 7:654-659. [PMID: 33502125 PMCID: PMC8136971 DOI: 10.1002/vms3.433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/02/2020] [Accepted: 01/09/2021] [Indexed: 12/02/2022] Open
Abstract
The University of Missouri (MU) has established a colony of dystrophin‐deficient dogs with a mixed breed background to mirror the variable pathologic effects of dystrophinopathies between persons of a given kindred to further the understanding of the genetic and molecular basis of the variable phenotype; thus to facilitate discovery of an effective therapeutic strategy. Herein we report the phenotype and genotype of a normal‐appearing 10‐month‐old colony female that died suddenly. At necropsy examination, there were reduced skeletal and laryngeal muscle volume and mild dilatation of the oesophagus. Microscopic findings consisted of extensive degeneration and regeneration of the axial skeletal, tongue, oesophageal, and laryngeal muscles that were characterized by considerable central nucleation, individual fibre mineralization and interstitial fibrosis. The myocardial findings were limited to infiltration of adipose cells in the interstitium. The female dog was a compound heterozygote with one X chromosome carrying a point mutation in intron 6 of the dystrophin gene and the other X chromosome carrying a repetitive element insertion in intron 13 of the dystrophin gene. Although the direct cause of death was uncertain, it might likely be due to sudden cardiac death as has been seen in Duchenne muscular dystrophy patients. This case demonstrated dystrophinopathy in female dogs that have no ameliorating normal X chromosome.
Collapse
Affiliation(s)
- Jessica S Fortin
- Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, MO, USA
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.,National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Scott Korte
- Office of Animal Resources, University of Missouri, Columbia, MO, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Scott D Fitzgerald
- Veterinary Diagnostic Laboratory, Michigan State University, Lansing, MI, USA
| | - Gayle C Johnson
- Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, MO, USA
| | - Bruce F Smith
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA
| |
Collapse
|
32
|
Mickelson JR, Minor KM, Guo LT, Friedenberg SG, Cullen JN, Ciavarella A, Hambrook LE, Brenner KM, Helmond SE, Marks SL, Shelton GD. Sarcoglycan A mutation in miniature dachshund dogs causes limb-girdle muscular dystrophy 2D. Skelet Muscle 2021; 11:2. [PMID: 33407862 PMCID: PMC7789357 DOI: 10.1186/s13395-020-00257-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND A cohort of related miniature dachshund dogs with exercise intolerance, stiff gait, dysphagia, myoglobinuria, and markedly elevated serum creatine kinase activities were identified. METHODS Muscle biopsy histopathology, immunofluorescence microscopy, and western blotting were combined to identify the specific pathologic phenotype of the myopathy, and whole genome SNP array genotype data and whole genome sequencing were combined to determine its genetic basis. RESULTS Muscle biopsies were dystrophic. Sarcoglycanopathy, a form of limb-girdle muscular dystrophy, was suspected based on immunostaining and western blotting, where α, β, and γ-sarcoglycan were all absent or reduced. Genetic mapping and whole genome sequencing identified a premature stop codon mutation in the sarcoglycan A subunit gene (SGCA). Affected dachshunds were confirmed on several continents. CONCLUSIONS This first SGCA mutation found in dogs adds to the literature of genetic bases of canine muscular dystrophies and their usefulness as comparative models of human disease.
Collapse
Affiliation(s)
- James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55113, USA.
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55113, USA
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093-0709, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55113, USA
| | - Jonah N Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55113, USA
| | | | | | - Karen M Brenner
- Centre for Animal Referral and Emergency, Collingwood, Victoria, Australia
| | - Sarah E Helmond
- Animal Referral Hospital, Homebush, New South Wales, Australia
| | - Stanley L Marks
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093-0709, USA
| |
Collapse
|
33
|
Terrill JR, Al-Mshhdani BA, Duong MN, Wingate CD, Abbas Z, Baustista AP, Bettis AK, Balog-Alvarez CJ, Kornegay JN, Nghiem PP, Grounds MD, Arthur PG. Oxidative damage to urinary proteins from the GRMD dog and mdx mouse as biomarkers of dystropathology in Duchenne muscular dystrophy. PLoS One 2020; 15:e0240317. [PMID: 33031394 PMCID: PMC7544076 DOI: 10.1371/journal.pone.0240317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-chromosome linked muscle-wasting disease affecting about 1 in 3500–6000 boys worldwide. Myofibre necrosis and subsequent loss of muscle mass are due to several molecular sequelae, such as inflammation and oxidative stress. We have recently shown increased neutrophils, highly reactive oxidant hypochlorous acid (HOCl) generation by myeloperoxidase (MPO), and associated oxidative stress in muscle from the GRMD dog and mdx mouse models for DMD. These findings have led us to hypothesise that generation of HOCl by myeloperoxidase released from neutrophils has a significant role in dystropathology. Since access to muscle from DMD patients is limited, the aim of this study was to develop methods to study this pathway in urine. Using immunoblotting to measure markers of protein oxidation, we show increased labelling of proteins with antibodies to dinitrophenylhydrazine (DNP, oxidative damage) and DiBrY (halogenation by reactive oxidants from myeloperoxidase) in GRMD and mdx urine. A strong positive correlation was observed between DiBrY labelling in dog urine and muscle. A strong positive correlation was also observed when comparing DNP and DiBrY labelling (in muscle and urine) to markers of dystropathology (plasma creatine kinase) and neutrophil presence (muscle MPO). Our results indicate the presence of neutrophil mediated oxidative stress in both models, and suggest that urine is a suitable bio-fluid for the measurement of such biomarkers. These methods could be employed in future studies into the role of neutrophil mediated oxidative stress in DMD and other inflammatory pathologies.
Collapse
Affiliation(s)
- Jessica R. Terrill
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
- * E-mail:
| | - Basma A. Al-Mshhdani
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Marisa N. Duong
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Catherine D. Wingate
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Zahra Abbas
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Angelo P. Baustista
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Amanda K. Bettis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Cynthia J. Balog-Alvarez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Joe N. Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Peter P. Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Miranda D. Grounds
- School of Human Sciences, The University of Western Australia, Perth, Australia
| | - Peter G. Arthur
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
34
|
Brunetti B, Muscatello LV, Letko A, Papa V, Cenacchi G, Grillini M, Murgiano L, Jagannathan V, Drögemüller C. X-Linked Duchenne-Type Muscular Dystrophy in Jack Russell Terrier Associated with a Partial Deletion of the Canine DMD Gene. Genes (Basel) 2020; 11:genes11101175. [PMID: 33049940 PMCID: PMC7600251 DOI: 10.3390/genes11101175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 11/29/2022] Open
Abstract
A 9-month old male Jack Russell Terrier started showing paraparesis of the hindlimbs after a walk. Hospitalized, the dog went into cardiac arrest, and later died. Necroscopic examination revealed a severe thickness of the diaphragm, esophagus, and base of the tongue, leading to the diagnosis of muscular dystrophy. The histology confirmed the marked size variation, regeneration, and fibrosis replacement of the skeletal muscle fibers. Immunohistochemistry demonstrated the absence of dystrophin confirming the diagnosis. Transmission electron microscopy showed disarrangement of skeletal muscle fibers. Finally, whole-genome sequencing identified a ~368kb deletion spanning 19 exons of the canine dystrophin (DMD) gene. This pathogenic loss-of-function variant most likely explains the observed disease phenotype. The X-chromosomal variant was absent in seven controls of the same breed. Most likely, this partial deletion of the DMD gene was either transmitted on the maternal path within the family of the affected dog or arose de novo. This study revealed a spontaneous partial deletion in DMD gene in a Jack Russell Terrier showing a Duchenne-type muscular dystrophy due to non-functional dystrophin.
Collapse
Affiliation(s)
- Barbara Brunetti
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy;
- Correspondence: ; Tel.: +39-051-2097960
| | - Luisa V. Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy;
| | - Anna Letko
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (A.L.); (L.M.); (V.J.); (C.D.)
| | - Valentina Papa
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (V.P.); (G.C.)
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (V.P.); (G.C.)
| | - Marco Grillini
- Pathology Unit, S Orsola Malpighi Hospital, University of Bologna, 40138 Bologna, Italy;
| | - Leonardo Murgiano
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (A.L.); (L.M.); (V.J.); (C.D.)
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (A.L.); (L.M.); (V.J.); (C.D.)
| | - Cord Drögemüller
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (A.L.); (L.M.); (V.J.); (C.D.)
| |
Collapse
|
35
|
Barthélémy I, Calmels N, Weiss RB, Tiret L, Vulin A, Wein N, Peccate C, Drougard C, Beroud C, Deburgrave N, Thibaud JL, Escriou C, Punzón I, Garcia L, Kaplan JC, Flanigan KM, Leturcq F, Blot S. X-linked muscular dystrophy in a Labrador Retriever strain: phenotypic and molecular characterisation. Skelet Muscle 2020; 10:23. [PMID: 32767978 PMCID: PMC7412789 DOI: 10.1186/s13395-020-00239-0] [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: 02/27/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background Canine models of Duchenne muscular dystrophy (DMD) are a valuable tool to evaluate potential therapies because they faithfully reproduce the human disease. Several cases of dystrophinopathies have been described in canines, but the Golden Retriever muscular dystrophy (GRMD) model remains the most used in preclinical studies. Here, we report a new spontaneous dystrophinopathy in a Labrador Retriever strain, named Labrador Retriever muscular dystrophy (LRMD). Methods A colony of LRMD dogs was established from spontaneous cases. Fourteen LRMD dogs were followed-up and compared to the GRMD standard using several functional tests. The disease causing mutation was studied by several molecular techniques and identified using RNA-sequencing. Results The main clinical features of the GRMD disease were found in LRMD dogs; the functional tests provided data roughly overlapping with those measured in GRMD dogs, with similar inter-individual heterogeneity. The LRMD causal mutation was shown to be a 2.2-Mb inversion disrupting the DMD gene within intron 20 and involving the TMEM47 gene. In skeletal muscle, the Dp71 isoform was ectopically expressed, probably as a consequence of the mutation. We found no evidence of polymorphism in either of the two described modifier genes LTBP4 and Jagged1. No differences were found in Pitpna mRNA expression levels that would explain the inter-individual variability. Conclusions This study provides a full comparative description of a new spontaneous canine model of dystrophinopathy, found to be phenotypically equivalent to the GRMD model. We report a novel large DNA mutation within the DMD gene and provide evidence that LRMD is a relevant model to pinpoint additional DMD modifier genes.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Nadège Calmels
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Laboratoire de Diagnostic Génétique-Institut de Génétique Médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, 1 Place de L'Hôpital, 67091, Strasbourg, France
| | - Robert B Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Adeline Vulin
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France
| | - Nicolas Wein
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Cécile Peccate
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Carole Drougard
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Christophe Beroud
- Aix Marseille Université, INSERM, MMG, Bioinformatics & Genetics, Marseille, France.,APHM, Hôpital Timone Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | - Nathalie Deburgrave
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Jean-Laurent Thibaud
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Catherine Escriou
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Isabel Punzón
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Luis Garcia
- Université de Versailles Saint-Quentin-en-Yvelines, U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France
| | - Jean-Claude Kaplan
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Kevin M Flanigan
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - France Leturcq
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Stéphane Blot
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France.
| |
Collapse
|
36
|
Sanson M, Vu Hong A, Massourides E, Bourg N, Suel L, Amor F, Corre G, Bénit P, Barthélémy I, Blot S, Bigot A, Pinset C, Rustin P, Servais L, Voit T, Richard I, Israeli D. miR-379 links glucocorticoid treatment with mitochondrial response in Duchenne muscular dystrophy. Sci Rep 2020; 10:9139. [PMID: 32499563 PMCID: PMC7272451 DOI: 10.1038/s41598-020-66016-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal muscle disorder, caused by mutations in the DMD gene and affects approximately 1:5000-6000 male births. In this report, we identified dysregulation of members of the Dlk1-Dio3 miRNA cluster in muscle biopsies of the GRMD dog model. Of these, we selected miR-379 for a detailed investigation because its expression is high in the muscle, and is known to be responsive to glucocorticoid, a class of anti-inflammatory drugs commonly used in DMD patients. Bioinformatics analysis predicts that miR-379 targets EIF4G2, a translational factor, which is involved in the control of mitochondrial metabolic maturation. We confirmed in myoblasts that EIF4G2 is a direct target of miR-379, and identified the DAPIT mitochondrial protein as a translational target of EIF4G2. Knocking down DAPIT in skeletal myotubes resulted in reduced ATP synthesis and myogenic differentiation. We also demonstrated that this pathway is GC-responsive since treating mice with dexamethasone resulted in reduced muscle expression of miR-379 and increased expression of EIF4G2 and DAPIT. Furthermore, miR-379 seric level, which is also elevated in the plasma of DMD patients in comparison with age-matched controls, is reduced by GC treatment. Thus, this newly identified pathway may link GC treatment to a mitochondrial response in DMD.
Collapse
Affiliation(s)
- Mathilde Sanson
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Ai Vu Hong
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | | | - Nathalie Bourg
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Laurence Suel
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Fatima Amor
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Guillaume Corre
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Paule Bénit
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Inès Barthélémy
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Stephane Blot
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Anne Bigot
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute, Paris, France
| | | | - Pierre Rustin
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Isabelle Richard
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - David Israeli
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France.
| |
Collapse
|
37
|
Switonski M. Impact of gene therapy for canine monogenic diseases on the progress of preclinical studies. J Appl Genet 2020; 61:179-186. [PMID: 32189222 PMCID: PMC7148265 DOI: 10.1007/s13353-020-00554-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Rapid progress in knowledge of the organization of the dog genome has facilitated the identification of the mutations responsible for numerous monogenic diseases, which usually present a breed-specific distribution. The majority of these diseases have clinical and molecular counterparts in humans. The affected dogs have thus become valuable models for preclinical studies of gene therapy for problems such as eye diseases, immunodeficiency, lysosomal storage diseases, hemophilia, and muscular dystrophy. Successful gene therapies in dogs have significantly contributed to decisions to run clinical trials for several human diseases, such as Leber's congenital amaurosis 2-LCA2 (caused by a mutation of RPE65), X-linked retinitis pigmentosa-XLRP (caused by mutation RPGR), and achromatopsia (caused by mutation of CNGB3). Promising results were also obtained for canine as follows: hemophilia (A and B), mucopolysaccharidoses (MPS I, MPS IIIB, MPS VII), leukocyte adhesion deficiency (CLAD), and muscular dystrophy (a counterpart of human Duchenne dystrophy). Present knowledge on molecular background of canine monogenic diseases and their successful gene therapies prove that dogs have an important contribution to preclinical studies.
Collapse
Affiliation(s)
- Marek Switonski
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, Poznan, Poland.
| |
Collapse
|
38
|
Barthélémy I, Hitte C, Tiret L. The Dog Model in the Spotlight: Legacy of a Trustful Cooperation. J Neuromuscul Dis 2020; 6:421-451. [PMID: 31450509 PMCID: PMC6918919 DOI: 10.3233/jnd-190394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dogs have long been used as a biomedical model system and in particular as a preclinical proof of concept for innovative therapies before translation to humans. A recent example of the utility of this animal model is the promising myotubularin gene delivery in boys affected by X-linked centronuclear myopathy after successful systemic, long-term efficient gene therapy in Labrador retrievers. Mostly, this is due to unique features that make dogs an optimal system. The continuous emergence of spontaneous inherited disorders enables the identification of reliable complementary molecular models for human neuromuscular disorders (NMDs). Dogs’ characteristics including size, lifespan and unprecedented medical care level allow a comprehensive longitudinal description of diseases. Moreover, the highly similar pathogenic mechanisms with human patients yield to translational robustness. Finally, interindividual phenotypic heterogeneity between dogs helps identifying modifiers and anticipates precision medicine issues. This review article summarizes the present list of molecularly characterized dog models for NMDs and provides an exhaustive list of the clinical and paraclinical assays that have been developed. This toolbox offers scientists a sensitive and reliable system to thoroughly evaluate neuromuscular function, as well as efficiency and safety of innovative therapies targeting these NMDs. This review also contextualizes the model by highlighting its unique genetic value, shaped by the long-term coevolution of humans and domesticated dogs. Because the dog is one of the most protected research animal models, there is considerable opposition to include it in preclinical projects, posing a threat to the use of this model. We thus discuss ethical issues, emphasizing that unlike many other models, the dog also benefits from its contribution to comparative biomedical research with a drastic reduction in the prevalence of morbid alleles in the breeding stock and an improvement in medical care.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Christophe Hitte
- CNRS, University of Rennes 1, UMR 6290, IGDR, Faculty of Medicine, SFR Biosit, Rennes, France
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
39
|
Lim KRQ, Nguyen Q, Dzierlega K, Huang Y, Yokota T. CRISPR-Generated Animal Models of Duchenne Muscular Dystrophy. Genes (Basel) 2020; 11:genes11030342. [PMID: 32213923 PMCID: PMC7141101 DOI: 10.3390/genes11030342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive neuromuscular disorder most commonly caused by mutations disrupting the reading frame of the dystrophin (DMD) gene. DMD codes for dystrophin, which is critical for maintaining the integrity of muscle cell membranes. Without dystrophin, muscle cells receive heightened mechanical stress, becoming more susceptible to damage. An active body of research continues to explore therapeutic treatments for DMD as well as to further our understanding of the disease. These efforts rely on having reliable animal models that accurately recapitulate disease presentation in humans. While current animal models of DMD have served this purpose well to some extent, each has its own limitations. To help overcome this, clustered regularly interspaced short palindromic repeat (CRISPR)-based technology has been extremely useful in creating novel animal models for DMD. This review focuses on animal models developed for DMD that have been created using CRISPR, their advantages and disadvantages as well as their applications in the DMD field.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Kasia Dzierlega
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Yiqing Huang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G 2H7, Canada
- Correspondence: ; Tel.: +1-780-492-1102
| |
Collapse
|
40
|
Challenges associated with homologous directed repair using CRISPR-Cas9 and TALEN to edit the DMD genetic mutation in canine Duchenne muscular dystrophy. PLoS One 2020; 15:e0228072. [PMID: 31961902 PMCID: PMC6974172 DOI: 10.1371/journal.pone.0228072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/07/2020] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene that abolish the expression of dystrophin protein. Dogs with the genetic homologue, golden retriever muscular dystrophy dog (GRMD), have a splice site mutation that leads to skipping of exon 7 and a stop codon in the DMD transcript. Gene editing via homology-directed repair (HDR) has been used in the mdx mouse model of DMD but not in GRMD. In this study, we used clustered regularly interspaced short palindromic repeats (CRISPR) and transcription activator-like effector nucleases (TALEN) to restore dystrophin expression via HDR in myoblasts/myotubes and later via intramuscular injection of GRMD dogs. In vitro, DNA and RNA were successfully corrected but dystrophin protein was not translated. With intramuscular injection of two different guide arms, sgRNA A and B, there was mRNA expression and Sanger sequencing confirmed inclusion of exon 7 for all treatments. On Western blot analysis, protein expression of up to 6% of normal levels was seen in two dogs injected with sgRNA B and up to 16% of normal in one dog treated with sgRNA A. TALEN did not restore any dystrophin expression. While there were no adverse effects, clear benefits were not seen on histopathologic analysis, immunofluorescence microscopy, and force measurements. Based on these results, methods must be modified to increase the efficiency of HDR-mediated gene repair and protein expression.
Collapse
|
41
|
Toledo GND, Moraes JRED. Utrophin, MHC and M1/M2 macrophages in GRMD dogs. CIÊNCIA ANIMAL BRASILEIRA 2020. [DOI: 10.1590/1809-6891v21e-52383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Abstract Muscular dystrophies are hereditary diseases that lead to progressive degeneration of the skeletal musculature. Golden Retriever dogs are used as animal models because they show a hereditary muscle disease similar to muscular dystrophy in humans. Aims: To evaluate the immunostaining of M1 (CD68) and M2 (CD163) macrophages, MHC I, MHC II and, utrophin in muscles of Golden Retriever dogs affected by muscular dystrophy (GRMD). Methods: Samples from 17 male dogs affected by GRMD were divided into GI - dystrophic dogs up to one year of age; and GII - dystrophic dogs over one-year-old. Results: Immunostaining of CD163 was higher than CD68 in both GI and GII. CD68 showed no variation between groups of dystrophic animals. MHC class I immunostaining was most evident in the biceps femoris and triceps brachialis. MHC class II was expressed mildly in four dystrophic muscle types in GI and GII. Utrophin immunostaining was higher in GII. Conclusion: M2 macrophages were one of the main mononuclear inflammatory cells found in dystrophic muscles. The number of M2 in muscles of dogs with GRMD increases with age, linking this cell subtype to permanent muscle damage.
Collapse
|
42
|
Regan D, Garcia K, Thamm D. Clinical, Pathological, and Ethical Considerations for the Conduct of Clinical Trials in Dogs with Naturally Occurring Cancer: A Comparative Approach to Accelerate Translational Drug Development. ILAR J 2019; 59:99-110. [PMID: 30668709 DOI: 10.1093/ilar/ily019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/26/2018] [Indexed: 01/21/2023] Open
Abstract
The role of comparative oncology in translational research is receiving increasing attention from drug developers and the greater biomedical research community. Pet dogs with spontaneous cancer are important and underutilized translational models, owing to dogs' large size and relative outbreeding, combined with their high incidence of certain tumor histotypes with significant biological, genetic, and histological similarities to their human tumor counterparts. Dogs with spontaneous tumors naturally develop therapy resistance and spontaneous metastasis, all in the context of an intact immune system. These fundamental features of cancer biology are often lacking in induced or genetically engineered preclinical tumor models and likely contribute to their poor predictive value and the associated overall high failure rate in oncology drug development. Thus, the conduct of clinical trials in pet dogs with naturally occurring cancer represents a viable surrogate and valuable intermediary step that should be increasingly incorporated into the cancer drug discovery and development pipeline. The development of molecular-targeted therapies has resulted in an expanded role of the pathologist in human oncology trials, and similarly the expertise of veterinary pathologists will be increasingly valuable to all phases of comparative oncology trial design and conduct. In this review, we provide a framework of clinical, ethical, and pathology-focused considerations for the increasing integration of translational research investigations in dogs with spontaneous cancer as a means to accelerate clinical cancer discovery and drug development.
Collapse
Affiliation(s)
- Daniel Regan
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Kelly Garcia
- Biologic Resources Laboratory, University of Illinois, Chicago, Illinois
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
43
|
Piktel JS, Wilson LD. Translational Models of Arrhythmia Mechanisms and Susceptibility: Success and Challenges of Modeling Human Disease. Front Cardiovasc Med 2019; 6:135. [PMID: 31552276 PMCID: PMC6748164 DOI: 10.3389/fcvm.2019.00135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/27/2019] [Indexed: 01/10/2023] Open
Abstract
We discuss large animal translational models of arrhythmia susceptibility and sudden cardiac death, focusing on important considerations when interpreting the data derived before applying them to human trials. The utility of large animal models of arrhythmia and the pros and cons of specific translational large animals used will be discussed, including the necessary tradeoffs between models designed to derive mechanisms vs. those to test therapies. Recent technical advancements which can be applied to large animal models of arrhythmias to better elucidate mechanistic insights will be introduced. Finally, some specific examples of past successes and challenges in translating the results of large animal models of arrhythmias to clinical trials and practice will be examined, and common themes regarding the success and failure of translating studies to therapy in man will be discussed.
Collapse
Affiliation(s)
| | - Lance D. Wilson
- Department of Emergency Medicine, Emergency Care Research Institute and Heart and Vascular Research Center, MetroHealth Campus of Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
44
|
Eresen A, Hafsa NE, Alic L, Birch SM, Griffin JF, Kornegay JN, Ji JX. Muscle percentage index as a marker of disease severity in golden retriever muscular dystrophy. Muscle Nerve 2019; 60:621-628. [PMID: 31397906 DOI: 10.1002/mus.26657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Golden retriever muscular dystrophy (GRMD) is a spontaneous X-linked canine model of Duchenne muscular dystrophy that resembles the human condition. Muscle percentage index (MPI) is proposed as an imaging biomarker of disease severity in GRMD. METHODS To assess MPI, we used MRI data acquired from nine GRMD samples using a 4.7 T small-bore scanner. A machine learning approach was used with eight raw quantitative mapping of MRI data images (T1m, T2m, two Dixon maps, and four diffusion tensor imaging maps), three types of texture descriptors (local binary pattern, gray-level co-occurrence matrix, gray-level run-length matrix), and a gradient descriptor (histogram of oriented gradients). RESULTS The confusion matrix, averaged over all samples, showed 93.5% of muscle pixels classified correctly. The classification, optimized in a leave-one-out cross-validation, provided an average accuracy of 80% with a discrepancy in overestimation for young (8%) and old (20%) dogs. DISCUSSION MPI could be useful for quantifying GRMD severity, but careful interpretation is needed for severe cases.
Collapse
Affiliation(s)
- Aydin Eresen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas
| | - Noor E Hafsa
- Department of Electrical and Computer Engineering, Texas A&M University, Doha, Qatar
| | - Lejla Alic
- Department of Electrical and Computer Engineering, Texas A&M University, Doha, Qatar.,Magnetic Detection & Imaging Group, Faculty of Science & Technology, University of Twente, Enschede, The Netherlands
| | - Sharla M Birch
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - John F Griffin
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Joe N Kornegay
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Jim X Ji
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas.,Department of Electrical and Computer Engineering, Texas A&M University, Doha, Qatar
| |
Collapse
|
45
|
Guo LJ, Soslow JH, Bettis AK, Nghiem PP, Cummings KJ, Lenox MW, Miller MW, Kornegay JN, Spurney CF. Natural History of Cardiomyopathy in Adult Dogs With Golden Retriever Muscular Dystrophy. J Am Heart Assoc 2019; 8:e012443. [PMID: 31411085 PMCID: PMC6759898 DOI: 10.1161/jaha.119.012443] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Duchenne muscular dystrophy (DMD) is an X‐linked disease that causes progressive muscle weakness. Affected boys typically die from respiratory or cardiac failure. Golden retriever muscular dystrophy (GRMD) is genetically homologous with DMD and causes analogous skeletal and cardiac muscle disease. Previous studies have detailed features of GRMD cardiomyopathy in mostly young dogs. Cardiac disease is not well characterized in adult GRMD dogs, and cardiac magnetic resonance (CMR) imaging studies have not been completed. Methods and Results We evaluated echocardiography and CMR in 24 adult GRMD dogs at different ages. Left ventricular systolic and diastolic functions, wall thickness, and myocardial strain were assessed with echocardiography. Features evaluated with CMR included left ventricular function, chamber size, myocardial mass, and late gadolinium enhancement. Our results largely paralleled those of DMD cardiomyopathy. Ejection fraction and fractional shortening correlated well with age, with systolic dysfunction occurring at ≈30 to 45 months. Circumferential strain was more sensitive than ejection fraction in early disease detection. Evidence of left ventricular chamber dilatation provided proof of dilated cardiomyopathy. Late gadolinium enhancement imaging showed DMD‐like left ventricular lateral wall lesions and earlier involvement of the anterior septum. Multiple functional indexes were graded objectively and added, with and without late gadolinium enhancement, to give cardiac and cardiomyopathy scores of disease severity. Consistent with DMD, there was parallel skeletal muscle involvement, as tibiotarsal joint flexion torque declined in tandem with cardiac function. Conclusions This study established parallels of progressive cardiomyopathy between dystrophic dogs and boys, further validating GRMD as a model of DMD cardiac disease.
Collapse
Affiliation(s)
- Lee-Jae Guo
- Department of Veterinary Integrative Biosciences College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX.,Texas A&M Institute for Preclinical Studies College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX
| | - Jonathan H Soslow
- Division of Pediatric Cardiology Department of Pediatrics Vanderbilt University Medical Center Nashville TN
| | - Amanda K Bettis
- Department of Veterinary Integrative Biosciences College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX
| | - Peter P Nghiem
- Department of Veterinary Integrative Biosciences College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX
| | - Kevin J Cummings
- Department of Population Medicine and Diagnostic Sciences College of Veterinary Medicine Cornell University Ithaca NY
| | - Mark W Lenox
- Department of Biomedical Engineering College of Engineering Texas A&M University College Station TX
| | - Matthew W Miller
- Department of Small Animal Clinical Sciences College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences College of Veterinary Medicine and Biomedical Sciences Texas A&M University College Station TX
| | - Christopher F Spurney
- Division of Cardiology and Center for Genetic Medicine Research Children's National Health System Washington DC
| |
Collapse
|
46
|
Eresen A, Birch SM, Alic L, Griffin JF, Kornegay JN, Ji JX. New Similarity Metric for Registration of MRI to Histology: Golden Retriever Muscular Dystrophy Imaging. IEEE Trans Biomed Eng 2019; 66:1222-1230. [DOI: 10.1109/tbme.2018.2870711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
47
|
Dynamic changes to lipid mediators support transitions among macrophage subtypes during muscle regeneration. Nat Immunol 2019; 20:626-636. [PMID: 30936495 PMCID: PMC6537107 DOI: 10.1038/s41590-019-0356-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
Muscle damage elicits a sterile immune response that facilitates complete regeneration. Here, we utilized mass spectrometry-based lipidomics to map the mediator lipidome during the transition from inflammation to resolution and regeneration in skeletal muscle injury. We observed the temporal regulation of glycerophospholipids and the production of pro-inflammatory (e.g., leukotrienes, prostaglandins) and specialized pro-resolving (e.g., resolvins, lipoxins) lipid mediators, which were modulated by ibuprofen. These time-dependent profiles were recapitulated in sorted neutrophils and Ly6Chi and Ly6Clo muscle-infiltrating macrophages, with a distinct pro-resolving signature observed in Ly6Clo macrophages. RNA-seq of macrophages stimulated with resolvin D2 (RvD2) showed similarities to transcriptional changes found during the temporal Ly6Chi to Ly6Clo macrophage transition. In vivo, RvD2 increased Ly6Clo macrophages and functional improvement of the regenerating muscle. These results reveal dynamic lipid mediator signatures of innate immune cells and provide a proof-of-concept for their exploitable effector roles in muscle regeneration.
Collapse
|
48
|
Nghiem PP, Kornegay JN. Gene therapies in canine models for Duchenne muscular dystrophy. Hum Genet 2019; 138:483-489. [PMID: 30734120 DOI: 10.1007/s00439-019-01976-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
Abstract
Therapies for Duchenne muscular dystrophy (DMD) must first be tested in animal models to determine proof-of-concept, efficacy, and importantly, safety. The murine and canine models for DMD are genetically homologous and most commonly used in pre-clinical testing. Although the mouse is a strong, proof-of-concept model, affected dogs show more analogous clinical and immunological disease progression compared to boys with DMD. As such, evaluating genetic therapies in the canine models may better predict response at the genetic, phenotypic, and immunological levels. We review the use of canine models for DMD and their benefits as it pertains to genetic therapy studies, including gene replacement, exon skipping, and gene editing.
Collapse
Affiliation(s)
- Peter P Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, 4458 TAMU, College Station, TX, 77843-4458, USA.
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, 4458 TAMU, College Station, TX, 77843-4458, USA
| |
Collapse
|
49
|
Magrath P, Maforo N, Renella P, Nelson SF, Halnon N, Ennis DB. Cardiac MRI biomarkers for Duchenne muscular dystrophy. Biomark Med 2018; 12:1271-1289. [PMID: 30499689 PMCID: PMC6462870 DOI: 10.2217/bmm-2018-0125] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal inherited genetic disorder that results in progressive muscle weakness and ultimately loss of ambulation, respiratory failure and heart failure. Cardiac MRI (MRI) plays an increasingly important role in the diagnosis and clinical care of boys with DMD and associated cardiomyopathies. Conventional cardiac MRI biomarkers permit measurements of global cardiac function and presence of fibrosis, but changes in these measures are late manifestations. Emerging MRI biomarkers of myocardial function and structure include the estimation of rotational mechanics and regional strain using MRI tagging; T1-mapping; and T2-mapping, a marker of inflammation, edema and fat. These emerging biomarkers provide earlier insights into cardiac involvement in DMD, improving patient care and aiding the evaluation of emerging therapies.
Collapse
Affiliation(s)
- Patrick Magrath
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Nyasha Maforo
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Physics & Biology in Medicine IDP, University of California, Los Angeles, CA 90095, USA
| | - Pierangelo Renella
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Medicine, Division of Pediatric Cardiology, CHOC Children's Hospital, Orange, CA 92868, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Nancy Halnon
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Medicine, Division of Pediatric Cardiology, University of California, Los Angeles, CA 90024, USA
| | - Daniel B Ennis
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Bioengineering, University of California, Los Angeles, CA 90095, USA.,Physics & Biology in Medicine IDP, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Haddix SG, Lee YI, Kornegay JN, Thompson WJ. Cycles of myofiber degeneration and regeneration lead to remodeling of the neuromuscular junction in two mammalian models of Duchenne muscular dystrophy. PLoS One 2018; 13:e0205926. [PMID: 30379896 PMCID: PMC6209224 DOI: 10.1371/journal.pone.0205926] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/03/2018] [Indexed: 12/02/2022] Open
Abstract
Mice lacking the sarcolemmal protein dystrophin, designated mdx, have been widely used as a model of Duchenne muscular dystrophy. Dystrophic mdx mice as they mature develop notable morphological abnormalities to their neuromuscular junctions, the peripheral cholinergic synapses responsible for activating muscle fibers. Most obviously the acetylcholine receptor aggregates are fragmented into small non-continuous, islands. This contrasts with wild type mice whose acetylcholine receptor aggregates are continuous and pretzel-shaped in appearance. We show here that these abnormalities in mdx mice are also present in a canine model of Duchenne muscular dystrophy and provide additional evidence to support the hypothesis that NMJ remodeling occurs due to myofiber degeneration and regeneration. Using a method to investigate synaptic AChR replacement, we show that neuromuscular junction remodeling in mdx animals is caused by muscle fiber degeneration and regeneration at the synaptic site and is mimicked by deliberate myofiber injury in wild type mice. Importantly, the innervating motor axon plays a crucial role in directing the remodeling of the neuromuscular junction in dystrophy, as has been recorded in aging and deliberate muscle fiber injury in wild type mice. The remodeling occurs repetitively through the life of the animal and the changes in junctions become greater with age.
Collapse
Affiliation(s)
- Seth G. Haddix
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| | - Young il Lee
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, United States of America
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Joe N. Kornegay
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, United States of America
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, United States of America
| | - Wesley J. Thompson
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, United States of America
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|