1
|
Widrick JJ, Lambert MR, de Souza Leite F, Jung YL, Park J, Conner JR, Lee EA, Beggs AH, Kunkel LM. High resolution kinematic approach for quantifying impaired mobility of dystrophic zebrafish larvae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627004. [PMID: 39713379 PMCID: PMC11661059 DOI: 10.1101/2024.12.05.627004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Dystrophin-deficient zebrafish larvae are a small, genetically tractable vertebrate model of Duchenne muscular dystrophy well suited for early stage therapeutic development. However, current approaches for evaluating their impaired mobility, a physiologically relevant therapeutic target, are characterized by low resolution and high variability. To address this, we used high speed videography and deep learning-based markerless motion capture to develop linked-segment models of larval escape response (ER) swimming. Kinematic models provided repeatable, high precision estimates of larval ER performance. Effect sizes for ER peak instantaneous acceleration and speed, final displacement, and ER distance were 2 to 3.5 standard deviations less for dystrophin-deficient mutants vs. wild-types. Further analysis revealed that mutants swam slower because of a reduction in their tail stroke frequency with little change in tail stroke amplitude. Kinematic variables were highly predictive of the dystrophic phenotype with ≤ 3% of larvae misclassified by random forest and support vector machine models. Tail kinematics also performed as well as in vitro assessments of tail muscle contractility in classifying larvae as mutants or wild-type, suggesting that ER kinematics could serve as a non-lethal proxy for direct measurements of muscle function. In summary, ER kinematics can be used as precise, physiologically relevant, non-lethal biomarkers of the dystrophic phenotype. The open-source approach described here may have applications not only for studies of skeletal muscle disease but for other disciplines that use larval mobility as an experimental outcome.
Collapse
Affiliation(s)
- Jeffrey J. Widrick
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Matthias R. Lambert
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Felipe de Souza Leite
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Youngsook Lucy Jung
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| | - Junseok Park
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| | - James R. Conner
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| | - Alan H. Beggs
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| | - Louis M. Kunkel
- Division of Genetics and Genomics, Dept. of Pediatrics, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
2
|
Barrett P, Louie KW, Dupont JB, Mack DL, Maves L. Uncovering the Embryonic Origins of Duchenne Muscular Dystrophy. WIREs Mech Dis 2024; 16:e1653. [PMID: 39444092 PMCID: PMC11563919 DOI: 10.1002/wsbm.1653] [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: 07/05/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a severe degenerative muscle disease caused by mutations in the DMD gene, which encodes dystrophin. Despite its initial description in the late 19th century by French neurologist Guillaume Duchenne de Boulogne, and identification of causal DMD genetic mutations in the 1980s, therapeutics remain challenging. The current standard of care is corticosteroid treatment, which delays the progression of muscle dysfunction but is associated with significant adverse effects. Emerging therapeutic approaches, including AAV-mediated gene transfer, CRISPR gene editing, and small molecule interventions, are under development but face considerable obstacles. Although DMD is viewed as a progressive muscle disease, muscle damage and abnormal molecular signatures are already evident during fetal myogenesis. This early onset of pathology suggests that the limited success of current therapies may partly be due to their administration after aberrant embryonic myogenesis has occurred in the absence of dystrophin. Consequently, identifying optimal therapeutic strategies and intervention windows for DMD may depend on a better understanding of the earliest DMD disease mechanisms. As newer techniques are applied, the field is gaining increasingly detailed insights into the early muscle developmental abnormalities in DMD. A comprehensive understanding of the initial events in DMD pathogenesis and progression will facilitate the generation and testing of effective therapeutic interventions.
Collapse
Affiliation(s)
- Philip Barrett
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| | - Ke'ale W Louie
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | | | - David L Mack
- Departments of Rehabilitation Medicine, Bioengineering and Neurobiology & Biophysics, Institute for Stem Cell and Regenerative Medicine, University of Washington Medicine, Seattle, Washington, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
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
|
Fefeu M, Blatzer M, Kneppers A, Briand D, Rocheteau P, Haroche A, Hardy D, Juchet-Martin M, Danckaert A, Coudoré F, Tutakhail A, Huchet C, Lafoux A, Mounier R, Mir O, Gaillard R, Chrétien F. Serotonin reuptake inhibitors improve muscle stem cell function and muscle regeneration in male mice. Nat Commun 2024; 15:6457. [PMID: 39085209 PMCID: PMC11291725 DOI: 10.1038/s41467-024-50220-4] [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: 06/16/2021] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Serotonin reuptake inhibitor antidepressants such as fluoxetine are widely used to treat mood disorders. The mechanisms of action include an increase in extracellular level of serotonin, neurogenesis, and growth of vessels in the brain. We investigated whether fluoxetine could have broader peripheral regenerative properties. Following prolonged administration of fluoxetine in male mice, we showed that fluoxetine increases the number of muscle stem cells and muscle angiogenesis, associated with positive changes in skeletal muscle function. Fluoxetine also improved skeletal muscle regeneration after single and multiples injuries with an increased muscle stem cells pool and vessel density associated with reduced fibrotic lesions and inflammation. Mice devoid of peripheral serotonin treated with fluoxetine did not exhibit beneficial effects during muscle regeneration. Specifically, pharmacological, and genetic inactivation of the 5-HT1B subtype serotonin receptor also abolished the enhanced regenerative process induced by fluoxetine. We highlight here a regenerative property of serotonin on skeletal muscle.
Collapse
Affiliation(s)
- Mylène Fefeu
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
- Université de Paris Cité, Paris, France
| | - Michael Blatzer
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - David Briand
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Pierre Rocheteau
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Alexandre Haroche
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France
| | - David Hardy
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | - Mélanie Juchet-Martin
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France
| | | | - François Coudoré
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Abdulkarim Tutakhail
- CESP, MOODS Team, Inserm, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Corinne Huchet
- TaRGeT, INSERM UMR 1089, Nantes Université, CHU Nantes, Nantes, France
| | - Aude Lafoux
- Therassay Platform, Capacités, Université de Nantes, IRS 2 Nantes Biotech, Nantes, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Olivier Mir
- Sarcoma Group, Gustave Roussy, Villejuif, France
| | - Raphaël Gaillard
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de psychiatrie, Paris, France.
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
| | - Fabrice Chrétien
- Institut Pasteur, Experimental Neuropathology Unit, Global Health Department, Paris, France.
- Université de Paris Cité, Paris, France.
- GHU Paris Psychiatrie & Neurosciences, site Sainte Anne, Service Hospitalo-Universitaire de neuropathologie, Paris, France.
| |
Collapse
|
5
|
Karuppasamy M, English KG, Henry CA, Manzini MC, Parant JM, Wright MA, Ruparelia AA, Currie PD, Gupta VA, Dowling JJ, Maves L, Alexander MS. Standardization of zebrafish drug testing parameters for muscle diseases. Dis Model Mech 2024; 17:dmm050339. [PMID: 38235578 PMCID: PMC10820820 DOI: 10.1242/dmm.050339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Skeletal muscular diseases predominantly affect skeletal and cardiac muscle, resulting in muscle weakness, impaired respiratory function and decreased lifespan. These harmful outcomes lead to poor health-related quality of life and carry a high healthcare economic burden. The absence of promising treatments and new therapies for muscular disorders requires new methods for candidate drug identification and advancement in animal models. Consequently, the rapid screening of drug compounds in an animal model that mimics features of human muscle disease is warranted. Zebrafish are a versatile model in preclinical studies that support developmental biology and drug discovery programs for novel chemical entities and repurposing of established drugs. Due to several advantages, there is an increasing number of applications of the zebrafish model for high-throughput drug screening for human disorders and developmental studies. Consequently, standardization of key drug screening parameters, such as animal husbandry protocols, drug compound administration and outcome measures, is paramount for the continued advancement of the model and field. Here, we seek to summarize and explore critical drug treatment and drug screening parameters in the zebrafish-based modeling of human muscle diseases. Through improved standardization and harmonization of drug screening parameters and protocols, we aim to promote more effective drug discovery programs.
Collapse
Affiliation(s)
- Muthukumar Karuppasamy
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Katherine G. English
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Clarissa A. Henry
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA
| | - M. Chiara Manzini
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Melissa A. Wright
- Department of Pediatrics, Section of Child Neurology, University of Colorado at Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Avnika A. Ruparelia
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter D. Currie
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
- EMBL Australia, Victorian Node, Monash University, Clayton, Victoria 3800, Australia
| | - Vandana A. Gupta
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James J. Dowling
- Division of Neurology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
- Program for Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- UAB Center for Neurodegeneration and Experimental Therapeutics (CNET), Birmingham, AL 35294, USA
| |
Collapse
|
6
|
Tesoriero C, Greco F, Cannone E, Ghirotto F, Facchinello N, Schiavone M, Vettori A. Modeling Human Muscular Dystrophies in Zebrafish: Mutant Lines, Transgenic Fluorescent Biosensors, and Phenotyping Assays. Int J Mol Sci 2023; 24:8314. [PMID: 37176020 PMCID: PMC10179009 DOI: 10.3390/ijms24098314] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of myopathies characterized by progressive muscle weakness leading to death from heart or respiratory failure. MDs are caused by mutations in genes involved in both the development and organization of muscle fibers. Several animal models harboring mutations in MD-associated genes have been developed so far. Together with rodents, the zebrafish is one of the most popular animal models used to reproduce MDs because of the high level of sequence homology with the human genome and its genetic manipulability. This review describes the most important zebrafish mutant models of MD and the most advanced tools used to generate and characterize all these valuable transgenic lines. Zebrafish models of MDs have been generated by introducing mutations to muscle-specific genes with different genetic techniques, such as (i) N-ethyl-N-nitrosourea (ENU) treatment, (ii) the injection of specific morpholino, (iii) tol2-based transgenesis, (iv) TALEN, (v) and CRISPR/Cas9 technology. All these models are extensively used either to study muscle development and function or understand the pathogenetic mechanisms of MDs. Several tools have also been developed to characterize these zebrafish models by checking (i) motor behavior, (ii) muscle fiber structure, (iii) oxidative stress, and (iv) mitochondrial function and dynamics. Further, living biosensor models, based on the expression of fluorescent reporter proteins under the control of muscle-specific promoters or responsive elements, have been revealed to be powerful tools to follow molecular dynamics at the level of a single muscle fiber. Thus, zebrafish models of MDs can also be a powerful tool to search for new drugs or gene therapies able to block or slow down disease progression.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Francesca Greco
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Elena Cannone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Francesco Ghirotto
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| |
Collapse
|
7
|
Singh J, Patten SA. Modeling neuromuscular diseases in zebrafish. Front Mol Neurosci 2022; 15:1054573. [PMID: 36583079 PMCID: PMC9794147 DOI: 10.3389/fnmol.2022.1054573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Neuromuscular diseases are a diverse group of conditions that affect the motor system and present some overlapping as well as distinct clinical manifestations. Although individually rare, the combined prevalence of NMDs is similar to Parkinson's. Over the past decade, new genetic mutations have been discovered through whole exome/genome sequencing, but the pathogenesis of most NMDs remains largely unexplored. Little information on the molecular mechanism governing the progression and development of NMDs accounts for the continual failure of therapies in clinical trials. Different aspects of the diseases are typically investigated using different models from cells to animals. Zebrafish emerges as an excellent model for studying genetics and pathogenesis and for developing therapeutic interventions for most NMDs. In this review, we describe the generation of different zebrafish genetic models mimicking NMDs and how they are used for drug discovery and therapy development.
Collapse
Affiliation(s)
- Jaskaran Singh
- INRS – Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Shunmoogum A. Patten
- INRS – Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada,Departement de Neurosciences, Université de Montréal, Montréal, QC, Canada,Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC, Canada,*Correspondence: Shunmoogum A. Patten,
| |
Collapse
|
8
|
Gosselin MRF, Mournetas V, Borczyk M, Verma S, Occhipinti A, Róg J, Bozycki L, Korostynski M, Robson SC, Angione C, Pinset C, Gorecki DC. Loss of full-length dystrophin expression results in major cell-autonomous abnormalities in proliferating myoblasts. eLife 2022; 11:e75521. [PMID: 36164827 PMCID: PMC9514850 DOI: 10.7554/elife.75521] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects myofibers and muscle stem cells, causing progressive muscle degeneration and repair defects. It was unknown whether dystrophic myoblasts-the effector cells of muscle growth and regeneration-are affected. Using transcriptomic, genome-scale metabolic modelling and functional analyses, we demonstrate, for the first time, convergent abnormalities in primary mouse and human dystrophic myoblasts. In Dmdmdx myoblasts lacking full-length dystrophin, the expression of 170 genes was significantly altered. Myod1 and key genes controlled by MyoD (Myog, Mymk, Mymx, epigenetic regulators, ECM interactors, calcium signalling and fibrosis genes) were significantly downregulated. Gene ontology analysis indicated enrichment in genes involved in muscle development and function. Functionally, we found increased myoblast proliferation, reduced chemotaxis and accelerated differentiation, which are all essential for myoregeneration. The defects were caused by the loss of expression of full-length dystrophin, as similar and not exacerbated alterations were observed in dystrophin-null Dmdmdx-βgeo myoblasts. Corresponding abnormalities were identified in human DMD primary myoblasts and a dystrophic mouse muscle cell line, confirming the cross-species and cell-autonomous nature of these defects. The genome-scale metabolic analysis in human DMD myoblasts showed alterations in the rate of glycolysis/gluconeogenesis, leukotriene metabolism, and mitochondrial beta-oxidation of various fatty acids. These results reveal the disease continuum: DMD defects in satellite cells, the myoblast dysfunction affecting muscle regeneration, which is insufficient to counteract muscle loss due to myofiber instability. Contrary to the established belief, our data demonstrate that DMD abnormalities occur in myoblasts, making these cells a novel therapeutic target for the treatment of this lethal disease.
Collapse
Affiliation(s)
- Maxime RF Gosselin
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| | | | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Justyna Róg
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Lukasz Bozycki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Samuel C Robson
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Centre for Enzyme Innovation, University of PortsmouthPortsmouthUnited Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | | | - Dariusz C Gorecki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
9
|
Kawahara G. [Therapeutic drug screening with zebrafish models]. Nihon Yakurigaku Zasshi 2021; 156:355-358. [PMID: 34719569 DOI: 10.1254/fpj.21060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The zebrafish mutant strains to mimic human diseases have been developed to study human diseases and to discover novel therapeutic drugs. The characteristics of zebrafish, small size, large clutch size, ex utero development, body transparency, and genetic tractability are very useful to research approaches like therapeutic drug screening. Here we introduce and discuss zebrafish models of human diseases and methods of drugs screening using them. Specifically, we show examples of therapeutic drug screens using zebrafish models of a muscular dystrophy to identify new candidates to improve phenotypes in skeletal muscle. The drug screening also reveals new drug actions and unexpected directions for future therapeutic ways. Phenotypic drug screen using zebrafish is an important for rapidly developing and validating therapeutics for human diseases.
Collapse
|
10
|
Zabłocka B, Górecki DC, Zabłocki K. Disrupted Calcium Homeostasis in Duchenne Muscular Dystrophy: A Common Mechanism behind Diverse Consequences. Int J Mol Sci 2021; 22:11040. [PMID: 34681707 PMCID: PMC8537421 DOI: 10.3390/ijms222011040] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) leads to disability and death in young men. This disease is caused by mutations in the DMD gene encoding diverse isoforms of dystrophin. Loss of full-length dystrophins is both necessary and sufficient for causing degeneration and wasting of striated muscles, neuropsychological impairment, and bone deformities. Among this spectrum of defects, abnormalities of calcium homeostasis are the common dystrophic feature. Given the fundamental role of Ca2+ in all cells, this biochemical alteration might be underlying all the DMD abnormalities. However, its mechanism is not completely understood. While abnormally elevated resting cytosolic Ca2+ concentration is found in all dystrophic cells, the aberrant mechanisms leading to that outcome have cell-specific components. We probe the diverse aspects of calcium response in various affected tissues. In skeletal muscles, cardiomyocytes, and neurons, dystrophin appears to serve as a scaffold for proteins engaged in calcium homeostasis, while its interactions with actin cytoskeleton influence endoplasmic reticulum organisation and motility. However, in myoblasts, lymphocytes, endotheliocytes, and mesenchymal and myogenic cells, calcium abnormalities cannot be clearly attributed to the loss of interaction between dystrophin and the calcium toolbox proteins. Nevertheless, DMD gene mutations in these cells lead to significant defects and the calcium anomalies are a symptom of the early developmental phase of this pathology. As the impaired calcium homeostasis appears to underpin multiple DMD abnormalities, understanding this alteration may lead to the development of new therapies. In fact, it appears possible to mitigate the impact of the abnormal calcium homeostasis and the dystrophic phenotype in the total absence of dystrophin. This opens new treatment avenues for this incurable disease.
Collapse
Affiliation(s)
- Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Dariusz C. Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael’s Building, White Swan Road, Portsmouth PO1 2DT, UK
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
11
|
Animal models for researching approaches to therapy of Duchenne muscular dystrophy. Transgenic Res 2021; 30:709-725. [PMID: 34409525 DOI: 10.1007/s11248-021-00278-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/11/2021] [Indexed: 01/17/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a relatively widespread genetic disease which develops as a result of a mutation in the gene DMD encoding dystrophin. In this review, animal models of DMD are described. These models are used in preclinical studies to elucidate the pathogenesis of the disease or to develop effective treatments; each animal model has its own advantages and disadvantages. For instance, Caenorhabditis elegans, Drosophila melanogaster, and zebrafish (sapje) are suitable for large-scale chemical screening of large numbers of small molecules, but their disease phenotype differs from that of mammals. The use of larger animals is important for understanding of the potential efficacy of various treatments for DMD. While mdx mice have their advantages, they exhibit a milder disease phenotype compared to humans or dogs, making it difficult to evaluate the efficacy of new treatment for DMD. The disease in dogs and pigs is more severe and progresses faster than in mice, but it is more difficult to breed and obtain sufficient numbers of specimens in order to achieve statistically significant results. Moreover, working with large animals is also more labor-intensive. Therefore, when choosing the optimal animal model for research, it is worth considering all the goals and objectives.
Collapse
|
12
|
Dubińska-Magiera M, Migocka-Patrzałek M, Lewandowski D, Daczewska M, Jagla K. Zebrafish as a Model for the Study of Lipid-Lowering Drug-Induced Myopathies. Int J Mol Sci 2021; 22:5654. [PMID: 34073503 PMCID: PMC8198905 DOI: 10.3390/ijms22115654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/06/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced myopathies are classified as acquired myopathies caused by exogenous factors. These pathological conditions develop in patients without muscle disease and are triggered by a variety of medicaments, including lipid-lowering drugs (LLDs) such as statins, fibrates, and ezetimibe. Here we summarise the current knowledge gained via studies conducted using various models, such as cell lines and mammalian models, and compare them with the results obtained in zebrafish (Danio rerio) studies. Zebrafish have proven to be an excellent research tool for studying dyslipidaemias as a model of these pathological conditions. This system enables in-vivo characterization of drug and gene candidates to further the understanding of disease aetiology and develop new therapeutic strategies. Our review also considers important environmental issues arising from the indiscriminate use of LLDs worldwide. The widespread use and importance of drugs such as statins and fibrates justify the need for the meticulous study of their mechanism of action and the side effects they cause.
Collapse
Affiliation(s)
- Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (M.D.-M.); (M.M.-P.); (D.L.)
| | - Marta Migocka-Patrzałek
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (M.D.-M.); (M.M.-P.); (D.L.)
| | - Damian Lewandowski
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (M.D.-M.); (M.M.-P.); (D.L.)
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (M.D.-M.); (M.M.-P.); (D.L.)
| | - Krzysztof Jagla
- Genetics Reproduction and Development Institute (iGReD), INSERM 1103, CNRS 6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63001 Clermont-Ferrand, France
| |
Collapse
|
13
|
Ellwood RA, Piasecki M, Szewczyk NJ. Caenorhabditis elegans as a Model System for Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22094891. [PMID: 34063069 PMCID: PMC8125261 DOI: 10.3390/ijms22094891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
The nematode worm Caenorhabditis elegans has been used extensively to enhance our understanding of the human neuromuscular disorder Duchenne Muscular Dystrophy (DMD). With new arising clinically relevant models, technologies and treatments, there is a need to reconcile the literature and collate the key findings associated with this model.
Collapse
Affiliation(s)
- Rebecca A. Ellwood
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Mathew Piasecki
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence:
| |
Collapse
|
14
|
Nutraceutical Screening in a Zebrafish Model of Muscular Dystrophy: Gingerol as a Possible Food Aid. Nutrients 2021; 13:nu13030998. [PMID: 33808773 PMCID: PMC8003371 DOI: 10.3390/nu13030998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an inherited neuromuscular disorder that causes loss of muscle mass and motor skills. In the era of genomic medicine, there is still no known cure for DMD. In clinical practice, there is a growing awareness of the possible importance of nutrition in neuromuscular diseases. This is mostly the result of patients’ or caregivers’ empirical reports of how active substances derived from food have led to improved muscle strength and, thus, better quality of life. In this report, we investigate several nutraceutical principles in the sapje strain of zebrafish, a validated model of DMD, in order to identify possible natural products that, if supplemented in the diet, might improve the quality of life of DMD patients. Gingerol, a constituent of fresh ginger, statistically increased the locomotion of mutant larvae and upregulated the expression of heme oxygenase 1, a target gene for therapy aimed at improving dystrophic symptoms. Although three other compounds showed a partial positive effect on locomotor and muscle structure phenotypes, our nutraceutical screening study lent preliminary support to the efficacy and safety only of gingerol. Gingerol could easily be proposed as a dietary supplement in DMD.
Collapse
|
15
|
Lambert MR, Spinazzola JM, Widrick JJ, Pakula A, Conner JR, Chin JE, Owens JM, Kunkel LM. PDE10A Inhibition Reduces the Manifestation of Pathology in DMD Zebrafish and Represses the Genetic Modifier PITPNA. Mol Ther 2020; 29:1086-1101. [PMID: 33221436 PMCID: PMC7934586 DOI: 10.1016/j.ymthe.2020.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder caused by mutations in the DMD gene. Absence of dystrophin protein leads to progressive degradation of skeletal and cardiac function and leads to premature death. Over the years, zebrafish have been increasingly used for studying DMD and are a powerful tool for drug discovery and therapeutic development. In our study, a birefringence screening assay led to identification of phosphodiesterase 10A (PDE10A) inhibitors that reduced the manifestation of dystrophic muscle phenotype in dystrophin-deficient sapje-like zebrafish larvae. PDE10A has been validated as a therapeutic target by pde10a morpholino-mediated reduction in muscle pathology and improvement in locomotion, muscle, and vascular function as well as long-term survival in sapje-like larvae. PDE10A inhibition in zebrafish and DMD patient-derived myoblasts were also associated with reduction of PITPNA expression that has been previously identified as a protective genetic modifier in two exceptional dystrophin-deficient golden retriever muscular dystrophy (GRMD) dogs that escaped the dystrophic phenotype. The combination of a phenotypic assay and relevant functional assessments in the sapje-like zebrafish enhances the potential for the prospective discovery of DMD therapeutics. Indeed, our results suggest a new application for a PDE10A inhibitor as a potential DMD therapeutic to be investigated in a mouse model of DMD.
Collapse
Affiliation(s)
- Matthias R Lambert
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Janelle M Spinazzola
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey J Widrick
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Pakula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - James R Conner
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Janice E Chin
- Rare Disease Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Jane M Owens
- Rare Disease Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; The Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Farr GH, Morris M, Gomez A, Pham T, Kilroy E, Parker EU, Said S, Henry C, Maves L. A novel chemical-combination screen in zebrafish identifies epigenetic small molecule candidates for the treatment of Duchenne muscular dystrophy. Skelet Muscle 2020; 10:29. [PMID: 33059738 PMCID: PMC7559456 DOI: 10.1186/s13395-020-00251-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder and is one of the most common muscular dystrophies. There are currently few effective therapies to treat the disease, although many small-molecule approaches are being pursued. Certain histone deacetylase inhibitors (HDACi) have been shown to ameliorate DMD phenotypes in mouse and zebrafish animal models. The HDACi givinostat has shown promise for DMD in clinical trials. However, beyond a small group of HDACi, other classes of epigenetic small molecules have not been broadly and systematically studied for their benefits for DMD. Methods We used an established animal model for DMD, the zebrafish dmd mutant strain sapje. A commercially available library of epigenetic small molecules was used to treat embryonic-larval stages of dmd mutant zebrafish. We used a quantitative muscle birefringence assay in order to assess and compare the effects of small-molecule treatments on dmd mutant zebrafish skeletal muscle structure. Results We performed a novel chemical-combination screen of a library of epigenetic compounds using the zebrafish dmd model. We identified candidate pools of epigenetic compounds that improve skeletal muscle structure in dmd mutant zebrafish. We then identified a specific combination of two HDACi compounds, oxamflatin and salermide, that ameliorated dmd mutant zebrafish skeletal muscle degeneration. We validated the effects of oxamflatin and salermide on dmd mutant zebrafish in an independent laboratory. Furthermore, we showed that the combination of oxamflatin and salermide caused increased levels of histone H4 acetylation in zebrafish larvae. Conclusions Our results provide novel, effective methods for performing a combination of small-molecule screen in zebrafish. Our results also add to the growing evidence that epigenetic small molecules may be promising candidates for treating DMD.
Collapse
Affiliation(s)
- Gist H Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Melanie Morris
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Medical Student Research Training Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Arianna Gomez
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Molecular Medicine and Mechanisms of Disease Program, Department of Pathology, University of Washington, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Thao Pham
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Molecular Medicine and Mechanisms of Disease Program, Department of Pathology, University of Washington, Seattle, WA, USA
| | - Elisabeth Kilroy
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Elizabeth U Parker
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Medical Student Research Training Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Shery Said
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Clarissa Henry
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA. .,Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Lasa-Fernandez H, Mosqueira-Martín L, Alzualde A, Lasa-Elgarresta J, Vallejo-Illarramendi A. A genotyping method combining primer competition PCR with HRM analysis to identify point mutations in Duchenne animal models. Sci Rep 2020; 10:17224. [PMID: 33057138 PMCID: PMC7560699 DOI: 10.1038/s41598-020-74173-y] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/28/2020] [Indexed: 11/09/2022] Open
Abstract
Dystrophin-null sapje zebrafish is an excellent model for better understanding the pathological mechanisms underlying Duchenne muscular dystrophy, and it has recently arisen as a powerful tool for high-throughput screening of therapeutic candidates for this disease. While dystrophic phenotype in sapje larvae can be easily detected by birefringence, zebrafish genotyping is necessary for drug screening experiments, where the potential rescue of larvae phenotype is the primary outcome. Genotyping is also desirable during colony husbandry since heterozygous progenitors need to be selected. Currently, sapje zebrafish are genotyped through techniques involving sequencing or multi-step PCR, which are often costly, tedious, or require special equipment. Here we report a simple, precise, cost-effective, and versatile PCR genotyping method based on primer competition. Genotypes can be resolved by standard agarose gel electrophoresis and high-resolution melt assay, the latter being especially useful for genotyping a large number of samples. Our approach has shown high sensitivity, specificity, and reproducibility in detecting the A/T point mutation in sapje zebrafish and the C/T mutation in the mdx mouse model of Duchenne. Hence, this method can be applied to other single nucleotide substitutions and may be further optimized to detect small insertions and deletions. Given its robust performance with crude DNA extracts, our strategy may be particularly well-suited for detecting single nucleotide variants in poor-quality samples such as ancient DNA or DNA from formalin-fixed, paraffin-embedded material.
Collapse
Affiliation(s)
- Haizpea Lasa-Fernandez
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Laura Mosqueira-Martín
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | | | - Jaione Lasa-Elgarresta
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain.
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain.
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain.
| |
Collapse
|
18
|
Spinazzola JM, Lambert MR, Gibbs DE, Conner JR, Krikorian GL, Pareek P, Rago C, Kunkel LM. Effect of serotonin modulation on dystrophin-deficient zebrafish. Biol Open 2020; 9:bio053363. [PMID: 32718931 PMCID: PMC7473644 DOI: 10.1242/bio.053363] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/16/2020] [Indexed: 01/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease caused by mutation of the dystrophin gene. Pharmacological therapies that function independently of dystrophin and complement strategies aimed at dystrophin restoration could significantly improve patient outcomes. Previous observations have suggested that serotonin pathway modulation ameliorates dystrophic pathology, and re-application of serotonin modulators already used clinically would potentially hasten availability to DMD patients. In our study, we used dystrophin-deficient sapje and sapje-like zebrafish models of DMD for rapid and easy screening of several classes of serotonin pathway modulators as potential therapeutics. None of the candidate drugs tested significantly decreased the percentage of zebrafish exhibiting the dystrophic muscle phenotype in the short-term birefringence assay or lengthened the lifespan in the long-term survival assay. Although we did not identify an effective drug, we believe our data is of value to the DMD research community for future studies, and there is evidence that suggests serotonin modulation may still be a viable treatment strategy with further investigation. Given the widespread clinical use of selective serotonin reuptake inhibitors, tricyclic antidepressants and reversible inhibitors of monoamine oxidase, their reapplication to DMD is an attractive strategy in the field's pursuit to identify pharmacological therapies to complement dystrophin restoration strategies.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Matthias R Lambert
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Devin E Gibbs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - James R Conner
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Georgia L Krikorian
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Prithu Pareek
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | | | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
19
|
Fabian L, Dowling JJ. Zebrafish Models of LAMA2-Related Congenital Muscular Dystrophy (MDC1A). Front Mol Neurosci 2020; 13:122. [PMID: 32742259 PMCID: PMC7364686 DOI: 10.3389/fnmol.2020.00122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/28/2023] Open
Abstract
LAMA2-related congenital muscular dystrophy (CMD; LAMA2-MD), also referred to as merosin deficient CMD (MDC1A), is a severe neonatal onset muscle disease caused by recessive mutations in the LAMA2 gene. LAMA2 encodes laminin α2, a subunit of the extracellular matrix (ECM) oligomer laminin 211. There are currently no treatments for MDC1A, and there is an incomplete understanding of disease pathogenesis. Zebrafish, due to their high degree of genetic conservation with humans, large clutch sizes, rapid development, and optical clarity, have emerged as an excellent model system for studying rare Mendelian diseases. They are particularly suitable as a model for muscular dystrophy because they contain at least one orthologue to all major human MD genes, have muscle that is similar to human muscle in structure and function, and manifest obvious and easily measured MD related phenotypes. In this review article, we present the existing zebrafish models of MDC1A, and discuss their contribution to the understanding of MDC1A pathomechanisms and therapy development.
Collapse
Affiliation(s)
- Lacramioara Fabian
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Wasala NB, Chen SJ, Duan D. Duchenne muscular dystrophy animal models for high-throughput drug discovery and precision medicine. Expert Opin Drug Discov 2020; 15:443-456. [PMID: 32000537 PMCID: PMC7065965 DOI: 10.1080/17460441.2020.1718100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/15/2020] [Indexed: 02/07/2023]
Abstract
Introduction: Duchenne muscular dystrophy (DMD) is an X-linked handicapping disease due to the loss of an essential muscle protein dystrophin. Dystrophin-null animals have been extensively used to study disease mechanisms and to develop experimental therapeutics. Despite decades of research, however, treatment options for DMD remain very limited.Areas covered: High-throughput high-content screening and precision medicine offer exciting new opportunities. Here, the authors review animal models that are suitable for these studies.Expert opinion: Nonmammalian models (worm, fruit fly, and zebrafish) are particularly attractive for cost-effective large-scale drug screening. Several promising lead compounds have been discovered using these models. Precision medicine for DMD aims at developing mutation-specific therapies such as exon-skipping and genome editing. To meet these needs, models with patient-like mutations have been established in different species. Models that harbor hotspot mutations are very attractive because the drugs developed in these models can bring mutation-specific therapies to a large population of patients. Humanized hDMD mice carry the entire human dystrophin gene in the mouse genome. Reagents developed in the hDMD mouse-based models are directly translatable to human patients.
Collapse
Affiliation(s)
- Nalinda B. Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212
| | - Shi-jie Chen
- Department of Physics, The University of Missouri, Columbia, MO 65211
- Department of Biochemistry, The University of Missouri, Columbia, MO 65211
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO 65212
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO 65212
| |
Collapse
|
21
|
Abstract
Sarcopenia - the accelerated age-related loss of muscle mass and function - is an under-diagnosed condition, and is central to deteriorating mobility, disability and frailty in older age. There is a lack of treatment options for older adults at risk of sarcopenia. Although sarcopenia's pathogenesis is multifactorial, its major phenotypes - muscle mass and muscle strength - are highly heritable. Several genome-wide association studies of muscle-related traits were published recently, providing dozens of candidate genes, many with unknown function. Therefore, animal models are required not only to identify causal mechanisms, but also to clarify the underlying biology and translate this knowledge into new interventions. Over the past several decades, small teleost fishes had emerged as powerful systems for modeling the genetics of human diseases. Owing to their amenability to rapid genetic intervention and the large number of conserved genetic and physiological features, small teleosts - such as zebrafish, medaka and killifish - have become indispensable for skeletal muscle genomic studies. The goal of this Review is to summarize evidence supporting the utility of small fish models for accelerating our understanding of human skeletal muscle in health and disease. We do this by providing a basic foundation of the (zebra)fish skeletal muscle morphology and physiology, and evidence of muscle-related gene homology. We also outline challenges in interpreting zebrafish mutant phenotypes and in translating them to human disease. Finally, we conclude with recommendations on future directions to leverage the large body of tools developed in small fish for the needs of genomic exploration in sarcopenia.
Collapse
Affiliation(s)
- Alon Daya
- The Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel
| | - Rajashekar Donaka
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 130010, Israel
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 130010, Israel
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, USA
| |
Collapse
|
22
|
Saha M, Rizzo SA, Ramanathan M, Hightower RM, Santostefano KE, Terada N, Finkel RS, Berg JS, Chahin N, Pacak CA, Wagner RE, Alexander MS, Draper I, Kang PB. Selective serotonin reuptake inhibitors ameliorate MEGF10 myopathy. Hum Mol Genet 2020; 28:2365-2377. [PMID: 31267131 DOI: 10.1093/hmg/ddz064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 02/02/2023] Open
Abstract
MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.
Collapse
Affiliation(s)
- Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Skylar A Rizzo
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Medosome Biotec, Alachua, FL, USA
| | - Manashwi Ramanathan
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rylie M Hightower
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA
| | - Katherine E Santostefano
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Naohiro Terada
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Richard S Finkel
- Division of Pediatric Neurology, Nemours Children's Hospital, Orlando, FL, USA
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nizar Chahin
- Department of Neurology, Neuromuscular Division, Oregon Health and Science University, Portland, Oregon, USA
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Matthew S Alexander
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA.,Department of Genetics, University of Alabama Birmingham, Birmingham, AL, USA.,Civitan International Research Center at University of Alabama Birmingham, Birmingham, AL, USA
| | - Isabelle Draper
- Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute, Boston, MA, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Molecular Genetics and Microbiology and Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
23
|
Widrick JJ, Kawahara G, Alexander MS, Beggs AH, Kunkel LM. Discovery of Novel Therapeutics for Muscular Dystrophies using Zebrafish Phenotypic Screens. J Neuromuscul Dis 2020; 6:271-287. [PMID: 31282429 PMCID: PMC6961982 DOI: 10.3233/jnd-190389] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The recent availability and development of mutant and transgenic zebrafish strains that model human muscular dystrophies has created new research opportunities for therapeutic development. Not only do these models mimic many pathological aspects of human dystrophies, but their small size, large clutch sizes, rapid ex utero development, body transparency, and genetic tractability enable research approaches that would be inconceivable with mammalian model systems. Here we discuss the use of zebrafish models of muscular dystrophy to rapidly screen hundreds to thousands of bioactive compounds in order to identify novel therapeutic candidates that modulate pathologic phenotypes. We review the justification and rationale behind this unbiased approach, including how zebrafish screens have identified FDA-approved drugs that are candidates for treating Duchenne and limb girdle muscular dystrophies. Not only can these drugs be re-purposed for treating dystrophies in a fraction of the time and cost of new drug development, but their identification has revealed novel, unexpected directions for future therapy development. Phenotype-driven zebrafish drug screens are an important compliment to the more established mammalian, target-based approaches for rapidly developing and validating therapeutics for muscular dystrophies.
Collapse
Affiliation(s)
- Jeffrey J Widrick
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Genri Kawahara
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology at the University of Alabama at Birmingham and Children's of Alabama; University of Alabama at Birmingham Center for Exercise Medicine; University of Alabama at Birmingham Civitan International Research Center; University of Alabama at Birmingham Department of Genetics; Birmingham, Alabama, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Darmahkasih AJ, Rybalsky I, Tian C, Shellenbarger KC, Horn PS, Lambert JT, Wong BL. Neurodevelopmental, behavioral, and emotional symptoms common in Duchenne muscular dystrophy. Muscle Nerve 2020; 61:466-474. [PMID: 31909820 DOI: 10.1002/mus.26803] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 11/11/2022]
Abstract
INTRODUCTION We studied neurodevelopmental and behavioral/emotional symptoms in patients with Duchenne muscular dystrophy (DMD). METHODS Retrospective case series of neurodevelopmental and behavioral/emotional symptoms obtained through review of systems of 700 DMD patients in relation to dystrophin gene mutations. RESULTS The most common symptoms encountered were emotional/behavioral dysregulation (38.7%), inattention/hyperactive features (31.4%), obsessive and compulsive features (25.0%), and language/speech delays (24.4%). Most patients (72.7%) had at least one symptom. Patients with mutations near the 3' end of the dystrophin gene were at higher risk for developing inattention/hyperactive features, language/speech delays, and global intellectual delays. Those with mutations between exon 31 and 79 had higher risk of clustering of symptoms when compared with those upstream of exon 30. DISCUSSION Neurodevelopmental, emotional, and behavioral symptoms are common comorbidities in DMD. There is higher prevalence of inattention/hyperactive features, language/speech delays, and global intellectual delays in genotypes affecting the 3' end of the dystrophin gene.
Collapse
Affiliation(s)
- Andrew J Darmahkasih
- Pediatric Residency Program, University of California, Irvine/Children's Hospital of Orange County, Orange, California
| | - Irina Rybalsky
- Neurology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Cuixia Tian
- Division of Neurology, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Karen C Shellenbarger
- Department of Pediatrics, University of Massachusetts Medical School, Worchester, Massachusetts
| | - Paul S Horn
- Division of Neurology, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Joshua T Lambert
- Neurology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brenda L Wong
- Department of Pediatrics, University of Massachusetts Medical School, Worchester, Massachusetts
| |
Collapse
|
25
|
Qiu B, Ruston J, Granzier H, Justice MJ, Dowling JJ. Failure to identify modifiers of NEBULIN-related nemaline myopathy in two pre-clinical models of the disease. Biol Open 2019; 8:bio.044867. [PMID: 31530540 PMCID: PMC6777365 DOI: 10.1242/bio.044867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nemaline myopathy is a rare neuromuscular disorder that affects 1 in 50,000 live births, with prevalence as high as 1 in 20,000 in certain populations. 13 genes have been linked to nemaline myopathy (NM), all of which are associated with the thin filament of the muscle sarcomere. Of the 13 associated genes, mutations in NEBULIN (NEB) accounts for up to 50% of all cases. Currently, the disease is incompletely understood and there are no available therapeutics for patients. To address this urgent need for effective treatments for patients affected by NM, we conducted a large scale chemical screen in a zebrafish model of NEB-related NM and an N-ethyl-N-nitrosourea (ENU)-based genetic screen in a mouse model of NEB exon 55 deletion, the most common NEB mutation in NM patients. Neither screen was able to identify a candidate for therapy development, highlighting the need to transition from conventional chemical therapeutics to gene-based therapies for the treatment of NM. Summary: NEBULIN-related nemaline myopathy currently has no treatment. We attempted to uncover new avenues for therapy by performing modifier screens, which unfortunately failed to identify modifiers that improved disease relevant phenotypes.
Collapse
Affiliation(s)
- Boyang Qiu
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - Julie Ruston
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Henk Granzier
- Department of Physiology, University of Arizona, Tuscon, Arizona 85724, USA
| | - Monica J Justice
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada .,Department of Molecular Genetics, University of Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
26
|
Muscular and mitochondrial effects of long-term fluoxetine treatment in mice, combined with physical endurance exercise on treadmill. Life Sci 2019; 232:116508. [DOI: 10.1016/j.lfs.2019.05.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 01/11/2023]
|
27
|
Antagonists for serotonin receptors ameliorate rhabdomyolysis induced by 25D-NBOMe, a psychoactive designer drug. Forensic Toxicol 2019. [DOI: 10.1007/s11419-019-00495-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
28
|
Abbey-Lee RN, Uhrig EJ, Garnham L, Lundgren K, Child S, Løvlie H. Experimental manipulation of monoamine levels alters personality in crickets. Sci Rep 2018; 8:16211. [PMID: 30385805 PMCID: PMC6212410 DOI: 10.1038/s41598-018-34519-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/22/2018] [Indexed: 01/15/2023] Open
Abstract
Animal personality has been described in a range of species with ecological and evolutionary consequences. Factors shaping and maintaining variation in personality are not fully understood, but monoaminergic systems are consistently linked to personality variation. We experimentally explored how personality was influenced by alterations in two key monoamine systems: dopamine and serotonin. This was done using ropinirole and fluoxetine, two common human pharmaceuticals. Using the Mediterranean field cricket (Gryllus bimaculatus), we focused on the personality traits activity, exploration, and aggression, with confirmed repeatability in our study. Dopamine manipulations explained little variation in the personality traits investigated, while serotonin manipulation reduced both activity and aggression. Due to limited previous research, we created a dose-response curve for ropinirole, ranging from concentrations measured in surface waters to human therapeutic doses. No ropinirole dose level strongly influenced cricket personality, suggesting our results did not come from a dose mismatch. Our results indicate that the serotonergic system explains more variation in personality than manipulations of the dopaminergic system. Additionally, they suggest that monoamine systems differ across taxa, and confirm the importance of the mode of action of pharmaceuticals in determining their effects on behaviour.
Collapse
Affiliation(s)
- Robin N Abbey-Lee
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden.
| | - Emily J Uhrig
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden.,Department of Biology, Southern Oregon University, 1250 Siskiyou Blvd, Ashland, OR, 97520, USA
| | - Laura Garnham
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden
| | - Kristoffer Lundgren
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden
| | - Sarah Child
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden.,Faculty of Biology, Medicine, and Health, Manchester University, Michael Smith Building, Dover St, Manchester, M13 9, UK
| | - Hanne Løvlie
- Dept of Physics, Chemistry and Biology, IFM Biology, Linköping University, 58183, Linköping, Sweden
| |
Collapse
|
29
|
Serafini PR, Feyder MJ, Hightower RM, Garcia-Perez D, Vieira NM, Lek A, Gibbs DE, Moukha-Chafiq O, Augelli-Szafran CE, Kawahara G, Widrick JJ, Kunkel LM, Alexander MS. A limb-girdle muscular dystrophy 2I model of muscular dystrophy identifies corrective drug compounds for dystroglycanopathies. JCI Insight 2018; 3:120493. [PMID: 30232282 DOI: 10.1172/jci.insight.120493] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 08/07/2018] [Indexed: 01/31/2023] Open
Abstract
Zebrafish are a powerful tool for studying muscle function owing to their high numbers of offspring, low maintenance costs, evolutionarily conserved muscle functions, and the ability to rapidly take up small molecular compounds during early larval stages. Fukutin-related protein (FKRP) is a putative protein glycosyltransferase that functions in the Golgi apparatus to modify sugar chain molecules of newly translated proteins. Patients with mutations in the FKRP gene can have a wide spectrum of clinical symptoms with varying muscle, eye, and brain pathologies depending on the location of the mutation in the FKRP protein. Patients with a common L276I FKRP mutation have mild adult-onset muscle degeneration known as limb-girdle muscular dystrophy 2I (LGMD2I), whereas patients with more C-terminal pathogenic mutations develop the severe Walker-Warburg syndrome (WWS)/muscle-eye-brain (MEB) disease. We generated fkrp-mutant zebrafish that phenocopy WWS/MEB pathologies including severe muscle breakdowns, head malformations, and early lethality. We have also generated a milder LGMD2I-model zebrafish via overexpression of a heat shock-inducible human FKRP (L276I) transgene that shows milder muscle pathology. Screening of an FDA-approved drug compound library in the LGMD2I zebrafish revealed a strong propensity towards steroids, antibacterials, and calcium regulators in ameliorating FKRP-dependent pathologies. Together, these studies demonstrate the utility of the zebrafish to both study human-specific FKRP mutations and perform compound library screenings for corrective drug compounds to treat muscular dystrophies.
Collapse
Affiliation(s)
- Peter R Serafini
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michael J Feyder
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rylie M Hightower
- Department of Pediatrics, Division of Neurology at the University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA.,UAB Center for Exercise Medicine at the University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daniela Garcia-Perez
- Department of Pediatrics, Division of Neurology at the University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA
| | - Natássia M Vieira
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Angela Lek
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Devin E Gibbs
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | | | | | - Genri Kawahara
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Jeffrey J Widrick
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, Massachusetts, USA
| | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology at the University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA.,UAB Center for Exercise Medicine at the University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Genetics at the University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
30
|
Koutsoulidou A, Photiades M, Kyriakides TC, Georgiou K, Prokopi M, Kapnisis K, Lusakowska A, Nearchou M, Christou Y, Papadimas GK, Anayiotos A, Kyriakou K, Kararizou E, Zamba Papanicolaou E, Phylactou LA. Identification of exosomal muscle-specific miRNAs in serum of myotonic dystrophy patients relating to muscle disease progress. Hum Mol Genet 2018. [PMID: 28637233 DOI: 10.1093/hmg/ddx212] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common form of adult-onset muscular dystrophy, which is characterised by progressive muscle wasting and the discovery of reliable blood-based biomarkers could be useful for the disease progress monitoring. There have been some reports showing that the presence of specific miRNAs in blood correlates with DM1. In one of these, our group identified four muscle-specific miRNAs, miR-1, miR-133a, miR-133b and miR-206, which correlated with the progression of muscle wasting observed in DM1 patients. The levels of the four muscle-specific miRNAs were elevated in the serum of DM1 patients compared to healthy participants and were also elevated in the serum of progressive muscle wasting DM1 patients compared to disease-stable DM1 patients. The aim of this work was to characterise the ontology of these four muscle-specific miRNAs in the blood circulation of DM1 patients. Here we show that the four muscle-specific miRNAs are encapsulated within exosomes isolated from DM1 patients. Our results show for the first time, the presence of miRNAs encapsulated within exosomes in blood circulation of DM1 patients. More interestingly, the levels of the four exosomal muscle-specific miRNAs are associated with the progression of muscle wasting in DM1 patients. We propose that exosomal muscle-specific miRNAs may be useful molecular biomarkers for monitoring the progress of muscle wasting in DM1 patients. There has been a growing interest regarding the clinical applications of exosomes and their role in prognosis and therapy of various diseases and the above results contribute towards this way.
Collapse
Affiliation(s)
- Andrie Koutsoulidou
- Department of Molecular Genetics, Function & Therapy, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Marinos Photiades
- Department of Molecular Genetics, Function & Therapy, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Tassos C Kyriakides
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Kristia Georgiou
- Department of Molecular Genetics, Function & Therapy, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Marianna Prokopi
- Department of Mechanical Engineering and Materials Science and Engineering, Cyprus University of Technology, Lemesos, Cyprus.,Theramir Ltd, Limassol, Cyprus
| | | | - Anna Lusakowska
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Marianna Nearchou
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Yiolanda Christou
- Neurology Clinic D, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - George K Papadimas
- Department of Neurology, Eginitio Hospital, Medical School of Athens, Athens, Greece
| | - Andreas Anayiotos
- Department of Mechanical Engineering and Materials Science and Engineering, Cyprus University of Technology, Lemesos, Cyprus
| | - Kyriakos Kyriakou
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Evangelia Kararizou
- Department of Neurology, Eginitio Hospital, Medical School of Athens, Athens, Greece
| | | | - Leonidas A Phylactou
- Department of Molecular Genetics, Function & Therapy, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| |
Collapse
|
31
|
Small molecule screen in embryonic zebrafish using modular variations to target segmentation. Nat Commun 2017; 8:1901. [PMID: 29196645 PMCID: PMC5711842 DOI: 10.1038/s41467-017-01469-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/19/2017] [Indexed: 01/19/2023] Open
Abstract
Small molecule in vivo phenotypic screening is used to identify drugs or biological activities by directly assessing effects in intact organisms. However, current screening designs may not exploit the full potential of chemical libraries due to false negatives. Here, we demonstrate a modular small molecule screen in embryonic zebrafish that varies concentration, genotype and timing to target segmentation disorders, birth defects that affect the spinal column. By testing each small molecule in multiple interrelated ways, this screen recovers compounds that a standard screening design would have missed, increasing the hit frequency from the chemical library three-fold. We identify molecular pathways and segmentation phenotypes, which we share in an open-access annotated database. These hits provide insight into human vertebral segmentation disorders and myopathies. This modular screening strategy is applicable to other developmental questions and disease models, highlighting the power of relatively small chemical libraries to accelerate gene discovery and disease study.
Collapse
|
32
|
Goody M, Jurczyszak D, Kim C, Henry C. Influenza A Virus Infection Damages Zebrafish Skeletal Muscle and Exacerbates Disease in Zebrafish Modeling Duchenne Muscular Dystrophy. PLOS CURRENTS 2017; 9. [PMID: 29188128 PMCID: PMC5693338 DOI: 10.1371/currents.md.8a7e35c50fa2b48156799d3c39788175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Both genetic and infectious diseases can result in skeletal muscle degeneration, inflammation, pain, and/or weakness. Duchenne muscular dystrophy (DMD) is the most common congenital muscle disease. DMD causes progressive muscle wasting due to mutations in Dystrophin. Influenza A and B viruses are frequently associated with muscle complications, especially in children. Infections activate an immune response and immunosuppressant drugs reduce DMD symptoms. These data suggest that the immune system may contribute to muscle pathology. However, roles of the immune response in DMD and Influenza muscle complications are not well understood. Zebrafish with dmd mutations are a well-characterized model in which to study the molecular and cellular mechanisms of DMD pathology. We recently showed that zebrafish can be infected by human Influenza A virus (IAV). Thus, the zebrafish is a powerful system with which to ask questions about the etiology and mechanisms of muscle damage due to genetic and/or infectious diseases. METHODS We infected zebrafish with IAV and assayed muscle tissue structure, sarcolemma integrity, cell-extracellular matrix (ECM) attachment, and molecular and cellular markers of inflammation in response to IAV infection alone or in the context of DMD. RESULTS We find that IAV-infected zebrafish display mild muscle degeneration with sarcolemma damage and compromised ECM adhesion. An innate immune response is elicited in muscle in IAV-infected zebrafish: NFkB signaling is activated, pro-inflammatory cytokine expression is upregulated, and neutrophils localize to sites of muscle damage. IAV-infected dmd mutants display more severe muscle damage than would be expected from an additive effect of dmd mutation and IAV infection, suggesting that muscle damage caused by Dystrophin-deficiency and IAV infection is synergistic. DISCUSSION These data demonstrate the importance of preventing IAV infections in individuals with genetic muscle diseases. Elucidating the mechanisms of immune-mediated muscle damage will not only apply to DMD and IAV, but also to other conditions where the immune system, inflammation, and muscle tissue are known to be affected, such as autoimmune diseases, cancer, and aging.
Collapse
Affiliation(s)
| | - Denise Jurczyszak
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA
| | | | - Clarissa Henry
- Graduate School for Biomedical Sciences and Engineering, School of Biology and Ecology, University of Maine. Orono, Main, USA
| |
Collapse
|
33
|
Kawahara G, Maeda H, Kikura-Hanajiri R, Yoshida KI, Hayashi YK. The psychoactive drug 25B-NBOMe recapitulates rhabdomyolysis in zebrafish larvae. Forensic Toxicol 2017; 35:369-375. [PMID: 28890736 PMCID: PMC5552826 DOI: 10.1007/s11419-017-0366-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/06/2017] [Indexed: 11/25/2022]
Abstract
N-Benzyl-substituted 2C class phenethylamines (NBOMes) are psychoactive designer drugs, with strong hallucinogenic and stimulant effects, even at low doses. The designer drug, 2-(4-bromo-2, 5-dimethoxyphenyl)-N-(2-methoxybenzyl) ethanamine (25B-NBOMe) is considered to be one of the most potent agonists of the serotonin-2A (5-HT2A) receptor. Recently, we reported the first lethal case of 25B-NBOMe intoxication with severe rhabdomyolysis, concluded by clinical, pathological and toxicological analyses. There are currently no good animal models that closely recapitulate serotonin receptor-dependent rhabdomyolysis. In the present study, we created animal models of rhabdomyolysis using zebrafish larvae to study the pathomechanism of rhabdomyolysis, and demonstrated that 25B-NBOMe can simulate lethal rhabdomyolysis in this animal. Treatment of the larvae with 25B-NBOMe decreased their survival rate, locomotion, altered birefringence of the skeletal muscle and immunostainings for dystroglycan (a myoseptal protein) and myosin heavy chain (a myofibril protein), which were consistent with rhabdomyolysis. This 25B-NBOMe-induced rhabdomyolysis was inhibited by the 5-HT2A receptor antagonists ritanserin and aripirazole, but not by the 5-HT1A + 5-HT1B receptor antagonist propranolol and the 5-HT3 receptor antagonist granisetron, indicating 5-HT2A-dependent rhabdomyolysis. The 25B-NBOMe-treated zebrafish is, therefore, a highly useful model of rhabdomyolysis for studying the pathomechanism of rhabdomyolysis as well as for therapeutic drug screening.
Collapse
Affiliation(s)
- Genri Kawahara
- Department of Pathophysiology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402 Japan
| | - Hideyuki Maeda
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, Japan
| | - Ruri Kikura-Hanajiri
- Division of Pharmacognosy, Phytochemistry and Narcotics, National Institute of Health Sciences, Tokyo, Japan
| | - Ken-ichi Yoshida
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, Japan
| | - Yukiko K. Hayashi
- Department of Pathophysiology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402 Japan
| |
Collapse
|
34
|
A novel early onset phenotype in a zebrafish model of merosin deficient congenital muscular dystrophy. PLoS One 2017; 12:e0172648. [PMID: 28241031 PMCID: PMC5328290 DOI: 10.1371/journal.pone.0172648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/07/2017] [Indexed: 11/19/2022] Open
Abstract
Merosin deficient congenital muscular dystrophy (MDC1A) is a severe neuromuscular disorder with onset in infancy that is associated with severe morbidities (particularly wheelchair dependence) and early mortality. It is caused by recessive mutations in the LAMA2 gene that encodes a subunit of the extracellular matrix protein laminin 211. At present, there are no treatments for this disabling disease. The zebrafish has emerged as a powerful model system for the identification of novel therapies. However, drug discovery in the zebrafish is largely dependent on the identification of phenotypes suitable for chemical screening. Our goal in this study was to elucidate novel, early onset abnormalities in the candyfloss (caf) zebrafish, a model of MDC1A. We uncovered and characterize abnormalities in spontaneous coiling, the earliest motor movement in the zebrafish, as a fully penetrant change specific to caf mutants that is ideal for future drug testing.
Collapse
|
35
|
Huang SSY, Benskin JP, Veldhoen N, Chandramouli B, Butler H, Helbing CC, Cosgrove JR. A multi-omic approach to elucidate low-dose effects of xenobiotics in zebrafish (Danio rerio) larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 182:102-112. [PMID: 27886581 DOI: 10.1016/j.aquatox.2016.11.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 06/06/2023]
Abstract
Regulatory-approved toxicity assays such as the OECD Fish Embryo Toxicity Assay (TG236) allow correlation of chemical exposure to adverse morphological phenotypes. However, these assays are ineffective in assessing sub-lethal (i.e. low-dose) effects, or differentiating between similar phenotypes induced by different chemicals. Inclusion of multi-omic analyses in studies investigating xenobiotic action provides improved characterization of biological response, thereby enhancing prediction of toxicological outcomes in whole animals in the absence of morphological effects. In the current study, we assessed perturbations in both the metabolome and transcriptome of zebrafish (Danio rerio; ZF) larvae exposed from 96 to 120h post fertilization to environmental concentrations of acetaminophen (APAP), diphenhydramine (DH), carbamazepine (CBZ), and fluoxetine (FLX); common pharmaceuticals with known mechanisms of action. Multi-omic responses were evaluated independently and integrated to identify molecular interactions and biological relevance of the responses. Results indicated chemical- and dose-specific changes suggesting differences in the time scale of transcript abundance and metabolite production. Increased impact on the metabolome relative to the transcriptome in FLX-treated animals suggests a stronger post-translational effect of the treatment. In contrast, the transcriptome showed higher sensitivity to perturbation in DH-exposed animals. Integration of 'omic' responses using multivariate approaches provided additional insights not obtained by independent 'omic' analyses and demonstrated that the most distinct overall response profiles were induced following low-dose exposure for all 4 pharmaceuticals. Importantly, changes in transcript abundance corroborated with predictions from metabolomic enrichment analyses and the identified perturbed biological pathways aligned with known xenobiotic mechanisms of action. This work demonstrates that a multi-omic toxicological approach, coupled with a sensitive animal model such as ZF larvae, can help characterize the toxicological relevance of acute low-dose chemical exposures.
Collapse
Affiliation(s)
- Susie S Y Huang
- SGS AXYS, Sidney, BC, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
| | - Jonathan P Benskin
- Department of Environmental Science and Analytical Chemistry (ACES), Stockholm University, Stockholm, Sweden
| | - Nik Veldhoen
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | | | | | - Caren C Helbing
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | | |
Collapse
|
36
|
|
37
|
Abstract
Skeletal muscle performs an essential function in human physiology with defects in genes encoding a variety of cellular components resulting in various types of inherited muscle disorders. Muscular dystrophies (MDs) are a severe and heterogeneous type of human muscle disease, manifested by progressive muscle wasting and degeneration. The disease pathogenesis and therapeutic options for MDs have been investigated for decades using rodent models, and considerable knowledge has been accumulated on the cause and pathogenetic mechanisms of this group of human disorders. However, due to some differences between disease severity and progression, what is learned in mammalian models does not always transfer to humans, prompting the desire for additional and alternative models. More recently, zebrafish have emerged as a novel and robust animal model for the study of human muscle disease. Zebrafish MD models possess a number of distinct advantages for modeling human muscle disorders, including the availability and ease of generating mutations in homologous disease-causing genes, the ability to image living muscle tissue in an intact animal, and the suitability of zebrafish larvae for large-scale chemical screens. In this chapter, we review the current understanding of molecular and cellular mechanisms involved in MDs, the process of myogenesis in zebrafish, and the structural and functional characteristics of zebrafish larval muscles. We further discuss the insights gained from the key zebrafish MD models that have been so far generated, and we summarize the attempts that have been made to screen for small molecules inhibitors of the dystrophic phenotypes using these models. Overall, these studies demonstrate that zebrafish is a useful in vivo system for modeling aspects of human skeletal muscle disorders. Studies using these models have contributed both to the understanding of the pathogenesis of muscle wasting disorders and demonstrated their utility as highly relevant models to implement therapeutic screening regimens.
Collapse
Affiliation(s)
- M Li
- Monash University, Clayton, VIC, Australia
| | - K J Hromowyk
- The Ohio State University, Columbus, OH, United States
| | - S L Amacher
- The Ohio State University, Columbus, OH, United States
| | - P D Currie
- Monash University, Clayton, VIC, Australia
| |
Collapse
|
38
|
Widrick JJ, Alexander MS, Sanchez B, Gibbs DE, Kawahara G, Beggs AH, Kunkel LM. Muscle dysfunction in a zebrafish model of Duchenne muscular dystrophy. Physiol Genomics 2016; 48:850-860. [PMID: 27764767 DOI: 10.1152/physiolgenomics.00088.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/29/2016] [Indexed: 01/10/2023] Open
Abstract
Sapje zebrafish lack the protein dystrophin and are the smallest vertebrate model of Duchenne muscular dystrophy (DMD). Their small size makes them ideal for large-scale drug discovery screens. However, the extent that sapje mimic the muscle dysfunction of higher vertebrate models of DMD is unclear. We used an optical birefringence assay to differentiate affected dystrophic sapje larvae from their unaffected siblings and then studied trunk muscle contractility at 4-7 days postfertilization. Preparation cross-sectional area (CSA) was similar for affected and unaffected larvae, yet tetanic forces of affected preparations were only 30-60% of normal. ANCOVA indicated that the linear relationship observed between tetanic force and CSA for unaffected preparations was absent in the affected population. Consequently, the average force/CSA of affected larvae was depressed 30-70%. Disproportionate reductions in twitch vs. tetanic force, and a slowing of twitch tension development and relaxation, indicated that the myofibrillar disorganization evident in the birefringence assay could not explain the entire force loss. Single eccentric contractions, in which activated preparations were lengthened 5-10%, resulted in tetanic force deficits in both groups of larvae. However, deficits of affected preparations were three- to fivefold greater at all strains and ages, even after accounting for any recovery. Based on these functional assessments, we conclude that the sapje mutant zebrafish is a phenotypically severe model of DMD. The severe contractile deficits of sapje larvae represent novel physiological endpoints for therapeutic drug screening.
Collapse
Affiliation(s)
- Jeffrey J Widrick
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts; .,Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts
| | - Matthew S Alexander
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts
| | - Benjamin Sanchez
- Department of Neurology, Division of Neuromuscular Diseases, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Devin E Gibbs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts
| | - Genri Kawahara
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts
| | - Alan H Beggs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts; and
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts; and.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
39
|
Housley MP, Njaine B, Ricciardi F, Stone OA, Hölper S, Krüger M, Kostin S, Stainier DYR. Cavin4b/Murcb Is Required for Skeletal Muscle Development and Function in Zebrafish. PLoS Genet 2016; 12:e1006099. [PMID: 27294373 PMCID: PMC4905656 DOI: 10.1371/journal.pgen.1006099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 05/10/2016] [Indexed: 01/16/2023] Open
Abstract
Skeletal muscles provide metazoans with the ability to feed, reproduce and avoid predators. In humans, a heterogeneous group of genetic diseases, termed muscular dystrophies (MD), lead to skeletal muscle dysfunction. Mutations in the gene encoding Caveolin-3, a principal component of the membrane micro-domains known as caveolae, cause defects in muscle maintenance and function; however it remains unclear how caveolae dysfunction underlies MD pathology. The Cavin family of caveolar proteins can form membrane remodeling oligomers and thus may also impact skeletal muscle function. Changes in the distribution and function of Cavin4/Murc, which is predominantly expressed in striated muscles, have been reported to alter caveolae structure through interaction with Caveolin-3. Here, we report the generation and phenotypic analysis of murcb mutant zebrafish, which display impaired swimming capacity, skeletal muscle fibrosis and T-tubule abnormalities during development. To understand the mechanistic importance of Murc loss of function, we assessed Caveolin-1 and 3 localization and found it to be abnormal. We further identified an in vivo function for Murc in Erk signaling. These data link Murc with developmental defects in T-tubule formation and progressive muscle dysfunction, thereby providing a new candidate for the etiology of muscular dystrophy.
Collapse
Affiliation(s)
- Michael P. Housley
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, California, United States of America
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (MPH); (DYRS)
| | - Brian Njaine
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Oliver A. Stone
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, California, United States of America
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Soraya Hölper
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marcus Krüger
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sawa Kostin
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), San Francisco, California, United States of America
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (MPH); (DYRS)
| |
Collapse
|
40
|
Abstract
Duchenne muscular dystrophy is a progressive, fatal, X-linked disease caused by a failure to accumulate the cytoskeletal protein dystrophin. This disease has been studied using a variety of animal models including fish, mice, rats, and dogs. While these models have contributed substantially to our mechanistic understanding of the disease and disease progression, limitations inherent to each model have slowed the clinical advancement of therapies, which necessitates the development of novel large-animal models. Several porcine dystrophin-deficient models have been identified, although disease severity may be so severe as to limit their potential contributions to the field. We have recently identified and completed the initial characterization of a natural porcine model of dystrophin insufficiency. Muscles from these animals display characteristic focal necrosis concomitant with decreased abundance and localization of dystrophin-glycoprotein complex components. These pigs recapitulate many of the cardinal features of muscular dystrophy, have elevated serum creatine kinase activity, and preliminarily appear to display altered locomotion. They also suffer from sudden death preceded by EKG abnormalities. Pig dystrophinopathy models could allow refinement of dosing strategies in human-sized animals in preparation for clinical trials. From an animal handling perspective, these pigs can generally be treated normally, with the understanding that acute stress can lead to sudden death. In summary, the ability to create genetically modified pig models and the serendipitous discovery of genetic disease in the swine industry has resulted in the emergence of new animal tools to facilitate the critical objective of improving the quality and length of life for boys afflicted with such a devastating disease.
Collapse
Affiliation(s)
- Joshua T Selsby
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Jason W Ross
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Dan Nonneman
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Katrin Hollinger
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| |
Collapse
|
41
|
Smith SJ, Horstick EJ, Davidson AE, Dowling J. Analysis of Zebrafish Larvae Skeletal Muscle Integrity with Evans Blue Dye. J Vis Exp 2015:53183. [PMID: 26649573 PMCID: PMC4692762 DOI: 10.3791/53183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The zebrafish model is an emerging system for the study of neuromuscular disorders. In the study of neuromuscular diseases, the integrity of the muscle membrane is a critical disease determinant. To date, numerous neuromuscular conditions display degenerating muscle fibers with abnormal membrane integrity; this is most commonly observed in muscular dystrophies. Evans Blue Dye (EBD) is a vital, cell permeable dye that is rapidly taken into degenerating, damaged, or apoptotic cells; in contrast, it is not taken up by cells with an intact membrane. EBD injection is commonly employed to ascertain muscle integrity in mouse models of neuromuscular diseases. However, such EBD experiments require muscle dissection and/or sectioning prior to analysis. In contrast, EBD uptake in zebrafish is visualized in live, intact preparations. Here, we demonstrate a simple and straightforward methodology for performing EBD injections and analysis in live zebrafish. In addition, we demonstrate a co-injection strategy to increase efficacy of EBD analysis. Overall, this video article provides an outline to perform EBD injection and characterization in zebrafish models of neuromuscular disease.
Collapse
Affiliation(s)
- Sarah J Smith
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto
| | - Eric J Horstick
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development; Departments of Pediatrics and Neurology, University of Michigan
| | - Ann E Davidson
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto
| | - James Dowling
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto; Departments of Pediatrics and Neurology, University of Michigan;
| |
Collapse
|
42
|
Lipscomb L, Piggott RW, Emmerson T, Winder SJ. Dasatinib as a treatment for Duchenne muscular dystrophy. Hum Mol Genet 2015; 25:266-74. [PMID: 26604135 PMCID: PMC4706114 DOI: 10.1093/hmg/ddv469] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
Identification of a systemically acting and universal small molecule therapy for Duchenne muscular dystrophy would be an enormous advance for this condition. Based on evidence gained from studies on mouse genetic models, we have identified tyrosine phosphorylation and degradation of β-dystroglycan as a key event in the aetiology of Duchenne muscular dystrophy. Thus, preventing tyrosine phosphorylation and degradation of β-dystroglycan presents itself as a potential therapeutic strategy. Using the dystrophic sapje zebrafish, we have investigated the use of tyrosine kinase and other inhibitors to treat the dystrophic symptoms in this model of Duchenne muscular dystrophy. Dasatinib, a potent and specific Src tyrosine kinase inhibitor, was found to decrease the levels of β-dystroglycan phosphorylation on tyrosine and to increase the relative levels of non-phosphorylated β-dystroglycan in sapje zebrafish. Furthermore, dasatinib treatment resulted in the improved physical appearance of the sapje zebrafish musculature and increased swimming ability as measured by both duration and distance of swimming of dasatinib-treated fish compared with control animals. These data suggest great promise for pharmacological agents that prevent the phosphorylation of β-dystroglycan on tyrosine and subsequent steps in the degradation pathway as therapeutic targets for the treatment of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Leanne Lipscomb
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Robert W Piggott
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Tracy Emmerson
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Steve J Winder
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
43
|
Li M, Arner A. Immobilization of Dystrophin and Laminin α2-Chain Deficient Zebrafish Larvae In Vivo Prevents the Development of Muscular Dystrophy. PLoS One 2015; 10:e0139483. [PMID: 26536238 PMCID: PMC4633184 DOI: 10.1371/journal.pone.0139483] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 09/13/2015] [Indexed: 11/19/2022] Open
Abstract
Muscular dystrophies are often caused by genetic alterations in the dystrophin-dystroglycan complex or its extracellular ligands. These structures are associated with the cell membrane and provide mechanical links between the cytoskeleton and the matrix. Mechanical stress is considered a pathological mechanism and muscle immobilization has been shown to be beneficial in some mouse models of muscular dystrophy. The zebrafish enables novel and less complex models to examine the effects of extended immobilization or muscle relaxation in vivo in different dystrophy models. We have examined effects of immobilization in larvae from two zebrafish strains with muscular dystrophy, the Sapje dystrophin-deficient and the Candyfloss laminin α2-chain-deficient strains. Larvae (4 days post fertilization, dpf) of both mutants have significantly lower active force in vitro, alterations in the muscle structure with gaps between muscle fibers and altered birefringence patterns compared to their normal siblings. Complete immobilization (18 hrs to 4 dpf) was achieved using a small molecular inhibitor of actin-myosin interaction (BTS, 50 μM). This treatment resulted in a significantly weaker active contraction at 4 dpf in both mutated larvae and normal siblings, most likely reflecting a general effect of immobilization on myofibrillogenesis. The immobilization also significantly reduced the structural damage in the mutated strains, showing that muscle activity is an important pathological mechanism. Following one-day washout of BTS, muscle tension partly recovered in the Candyfloss siblings and caused structural damage in these mutants, indicating activity-induced muscle recovery and damage, respectively.
Collapse
Affiliation(s)
- Mei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
44
|
Bajanca F, Gonzalez-Perez V, Gillespie SJ, Beley C, Garcia L, Theveneau E, Sear RP, Hughes SM. In vivo dynamics of skeletal muscle Dystrophin in zebrafish embryos revealed by improved FRAP analysis. eLife 2015; 4. [PMID: 26459831 PMCID: PMC4601390 DOI: 10.7554/elife.06541] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 09/10/2015] [Indexed: 12/30/2022] Open
Abstract
Dystrophin forms an essential link between sarcolemma and cytoskeleton, perturbation of which causes muscular dystrophy. We analysed Dystrophin binding dynamics in vivo for the first time. Within maturing fibres of host zebrafish embryos, our analysis reveals a pool of diffusible Dystrophin and complexes bound at the fibre membrane. Combining modelling, an improved FRAP methodology and direct semi-quantitative analysis of bleaching suggests the existence of two membrane-bound Dystrophin populations with widely differing bound lifetimes: a stable, tightly bound pool, and a dynamic bound pool with high turnover rate that exchanges with the cytoplasmic pool. The three populations were found consistently in human and zebrafish Dystrophins overexpressed in wild-type or dmd(ta222a/ta222a) zebrafish embryos, which lack Dystrophin, and in Gt(dmd-Citrine)(ct90a) that express endogenously-driven tagged zebrafish Dystrophin. These results lead to a new model for Dystrophin membrane association in developing muscle, and highlight our methodology as a valuable strategy for in vivo analysis of complex protein dynamics.
Collapse
Affiliation(s)
- Fernanda Bajanca
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,CNRS and Université Paul Sabatier, Toulouse, France
| | | | - Sean J Gillespie
- Department of Physics, University of Surrey, Guildford, United Kingdom
| | - Cyriaque Beley
- Université Versailles Saint-Quentin, Montigny-le-Bretonneux, France.,Laboratoire International Associé-Biologie appliquée aux handicaps neuromusculaires, Centre Scientifique de Monaco, Monaco, Monaco
| | - Luis Garcia
- Université Versailles Saint-Quentin, Montigny-le-Bretonneux, France.,Laboratoire International Associé-Biologie appliquée aux handicaps neuromusculaires, Centre Scientifique de Monaco, Monaco, Monaco
| | | | - Richard P Sear
- Department of Physics, University of Surrey, Guildford, United Kingdom
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| |
Collapse
|
45
|
Abstract
Regeneration involves interactions between multiple signaling pathways acting in a spatially and temporally complex manner. As signaling pathways are highly conserved, understanding how regeneration is controlled in animal models exhibiting robust regenerative capacities should aid efforts to stimulate repair in humans. One way to discover molecular regulators of regeneration is to alter gene/protein function and quantify effect(s) on the regenerative process: dedifferentiation/reprograming, stem/progenitor proliferation, migration/remodeling, progenitor cell differentiation and resolution. A powerful approach for applying this strategy to regenerative biology is chemical genetics, the use of small-molecule modulators of specific targets or signaling pathways. Here, we review advances that have been made using chemical genetics for hypothesis-focused and discovery-driven studies aimed at furthering understanding of how regeneration is controlled.
Collapse
|
46
|
Plantié E, Migocka-Patrzałek M, Daczewska M, Jagla K. Model organisms in the fight against muscular dystrophy: lessons from drosophila and Zebrafish. Molecules 2015; 20:6237-53. [PMID: 25859781 PMCID: PMC6272363 DOI: 10.3390/molecules20046237] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 01/01/2023] Open
Abstract
Muscular dystrophies (MD) are a heterogeneous group of genetic disorders that cause muscle weakness, abnormal contractions and muscle wasting, often leading to premature death. More than 30 types of MD have been described so far; those most thoroughly studied are Duchenne muscular dystrophy (DMD), myotonic dystrophy type 1 (DM1) and congenital MDs. Structurally, physiologically and biochemically, MDs affect different types of muscles and cause individual symptoms such that genetic and molecular pathways underlying their pathogenesis thus remain poorly understood. To improve our knowledge of how MD-caused muscle defects arise and to find efficacious therapeutic treatments, different animal models have been generated and applied. Among these, simple non-mammalian Drosophila and zebrafish models have proved most useful. This review discusses how zebrafish and Drosophila MD have helped to identify genetic determinants of MDs and design innovative therapeutic strategies with a special focus on DMD, DM1 and congenital MDs.
Collapse
Affiliation(s)
- Emilie Plantié
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France; E-Mail:
| | - Marta Migocka-Patrzałek
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland; E-Mails: (M.M.-P.); (M.D.)
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland; E-Mails: (M.M.-P.); (M.D.)
| | - Krzysztof Jagla
- GReD (Genetics, Reproduction and Development laboratory), INSERM U1103, CNRS UMR6293, University of Clermont-Ferrand, 28 place Henri-Dunant, 63000 Clermont-Ferrand, France; E-Mail:
| |
Collapse
|
47
|
Goody MF, Sher RB, Henry CA. Hanging on for the ride: adhesion to the extracellular matrix mediates cellular responses in skeletal muscle morphogenesis and disease. Dev Biol 2015; 401:75-91. [PMID: 25592225 DOI: 10.1016/j.ydbio.2015.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 10/24/2022]
Abstract
Skeletal muscle specification and morphogenesis during early development are critical for normal physiology. In addition to mediating locomotion, skeletal muscle is a secretory organ that contributes to metabolic homeostasis. Muscle is a highly adaptable tissue, as evidenced by the ability to increase muscle cell size and/or number in response to weight bearing exercise. Conversely, muscle wasting can occur during aging (sarcopenia), cancer (cancer cachexia), extended hospital stays (disuse atrophy), and in many genetic diseases collectively known as the muscular dystrophies and myopathies. It is therefore of great interest to understand the cellular and molecular mechanisms that mediate skeletal muscle development and adaptation. Muscle morphogenesis transforms short muscle precursor cells into long, multinucleate myotubes that anchor to tendons via the myotendinous junction. This process requires carefully orchestrated interactions between cells and their extracellular matrix microenvironment. These interactions are dynamic, allowing muscle cells to sense biophysical, structural, organizational, and/or signaling changes within their microenvironment and respond appropriately. In many musculoskeletal diseases, these cell adhesion interactions are disrupted to such a degree that normal cellular adaptive responses are not sufficient to compensate for accumulating damage. Thus, one major focus of current research is to identify the cell adhesion mechanisms that drive muscle morphogenesis, with the hope that understanding how muscle cell adhesion promotes the intrinsic adaptability of muscle tissue during development may provide insight into potential therapeutic approaches for muscle diseases. Our objectives in this review are to highlight recent studies suggesting conserved roles for cell-extracellular matrix adhesion in vertebrate muscle morphogenesis and cellular adaptive responses in animal models of muscle diseases.
Collapse
Affiliation(s)
- Michelle F Goody
- School of Biology and Ecology, University of Maine, Orono, ME 04469, United States
| | - Roger B Sher
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, United States
| | - Clarissa A Henry
- School of Biology and Ecology, University of Maine, Orono, ME 04469, United States; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, United States; Institute for Molecular Biophysics, University of Maine, Orono, ME 04469, United States.
| |
Collapse
|
48
|
LOBIKIN MARIA, PARÉ JEANFRAN, KAPLAN DAVIDL, LEVIN MICHAEL. Selective depolarization of transmembrane potential alters muscle patterning and muscle cell localization in Xenopus laevis embryos. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2015; 59:303-11. [PMID: 26198143 PMCID: PMC10461602 DOI: 10.1387/ijdb.150198ml] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The correct anatomical placement and precise determination of specific cell types is required for the establishment of normal embryonic patterning. Understanding these processes is also important for progress in regenerative medicine and cancer biology. Transmembrane voltage gradients across embryonic tissues can mediate cellular communication to regulate the processes of proliferation, migration, and differentiation. Our past work showed that selective depolarization of an endogenous instructor cell population in Xenopus laevis in vivo induced a melanoma-like phenotype in the absence of genetic damage. Here, we use a hypersensitive glycine-gated chloride channel (GlyR) under control of tissue-specific promoters to show that instructor cells resident within muscle are more effective at triggering the metastatic conversion of ectodermal melanocytes than those similar cells within the nervous system. Moreover, depolarization of muscle cells results in aberrant muscle patterning and the appearance of cells expressing muscle markers within the neural tube, which impacts but does not abolish the animals' ability to learn in an associative conditioning assay. Taken together, our data reveal new details of long-range (non-cell-autonomous) reprogramming of cell behavior via alteration of the resting potential of specific embryonic subpopulations.
Collapse
Affiliation(s)
- MARIA LOBIKIN
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, Medford, MA, USA
| | - JEAN-FRANçOIS PARÉ
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, Medford, MA, USA
| | - DAVID L. KAPLAN
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - MICHAEL LEVIN
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, Medford, MA, USA
| |
Collapse
|
49
|
Developing zebrafish models relevant to PTSD and other trauma- and stressor-related disorders. Prog Neuropsychopharmacol Biol Psychiatry 2014; 55:67-79. [PMID: 25138994 DOI: 10.1016/j.pnpbp.2014.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/01/2014] [Accepted: 08/07/2014] [Indexed: 11/20/2022]
Abstract
While post-traumatic stress disorder (PTSD) and other trauma- and stress-related disorders (TSRDs) represent a serious societal and public health concern, their pathogenesis is largely unknown. Given the clinical complexity of TSRD development and susceptibility, greater investigation into candidate biomarkers and specific genetic pathways implicated in both risk and resilience to trauma becomes critical. In line with this, numerous animal models have been extensively used to better understand the pathogenic mechanisms of PTSD and related TSRD. Here, we discuss the rapidly increasing potential of zebrafish as models of these disorders, and how their use may aid researchers in uncovering novel treatments and therapies in this field.
Collapse
|
50
|
Wood AJ, Currie PD. Analysing regenerative potential in zebrafish models of congenital muscular dystrophy. Int J Biochem Cell Biol 2014; 56:30-7. [PMID: 25449259 DOI: 10.1016/j.biocel.2014.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 10/12/2014] [Accepted: 10/18/2014] [Indexed: 01/16/2023]
Abstract
The congenital muscular dystrophies (CMDs) are a clinically and genetically heterogeneous group of muscle disorders. Clinically hypotonia is present from birth, with progressive muscle weakness and wasting through development. For the most part, CMDs can mechanistically be attributed to failure of basement membrane protein laminin-α2 sufficiently binding with correctly glycosylated α-dystroglycan. The majority of CMDs therefore arise as the result of either a deficiency of laminin-α2 (MDC1A) or hypoglycosylation of α-dystroglycan (dystroglycanopathy). Here we consider whether by filling a regenerative medicine niche, the zebrafish model can address the present challenge of delivering novel therapeutic solutions for CMD. In the first instance the readiness and appropriateness of the zebrafish as a model organism for pioneering regenerative medicine therapies in CMD is analysed, in particular for MDC1A and the dystroglycanopathies. Despite the recent rapid progress made in gene editing technology, these approaches have yet to yield any novel zebrafish models of CMD. Currently the most genetically relevant zebrafish models to the field of CMD, have all been created by N-ethyl-N-nitrosourea (ENU) mutagenesis. Once genetically relevant models have been established the zebrafish has several important facets for investigating the mechanistic cause of CMD, including rapid ex vivo development, optical transparency up to the larval stages of development and relative ease in creating transgenic reporter lines. Together, these tools are well suited for use in live-imaging studies such as in vivo modelling of muscle fibre detachment. Secondly, the zebrafish's contribution to progress in effective treatment of CMD was analysed. Two approaches were identified in which zebrafish could potentially contribute to effective therapies. The first hinges on the augmentation of functional redundancy within the system, such as upregulating alternative laminin chains in the candyfloss fish, a model of MDC1A. Secondly high-throughput small molecule screens not only provide effective therapies, but also an alternative strategy for investigating CMD in zebrafish. In this instance insight into disease mechanism is derived in reverse. Zebrafish models are therefore clearly of critical importance in the advancement of regenerative medicine strategies in CMD. This article is part of a Directed Issue entitled: Regenerative Medicine: The challenge of translation.
Collapse
Affiliation(s)
- A J Wood
- Australian Regenerative Medicine Institute, Building 75, Level 1, Clayton Campus, Wellington Road, Melbourne, Victoroia 3181, Australia
| | - P D Currie
- Australian Regenerative Medicine Institute, Building 75, Level 1, Clayton Campus, Wellington Road, Melbourne, Victoroia 3181, Australia.
| |
Collapse
|