1
|
Sciandra F, Bozzi M, Witt A, Goffing P, Covaceuszach S, Blaess S, Cassetta A, Bigotti MG, Huser T, Brancaccio A, Hübner W. Live cell optical super-resolution microscopy of dystroglycan mutants as a model for dystroglycanopathies in multiple cell lines. Front Mol Biosci 2025; 12:1558170. [PMID: 40248434 PMCID: PMC12003124 DOI: 10.3389/fmolb.2025.1558170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Dystroglycan (DG) is an adhesion complex comprising two subunits, α-DG and β-DG, which interact non-covalently at the plasma membrane. As a component of the dystrophin-glycoprotein complex DGC, DG plays a crucial role in linking the cytoskeleton to the surrounding basement membranes. Rare primary point mutations in the DAG1 gene have been identified in patients with various forms of neuromuscular dystrophy, ranging in phenotype from mild to severe. Methods To gain a deeper understanding of the molecular mechanisms underlying these pathologies, we have designed a series of chimeric GFP-tagged full-length α/β-DG constructs and expressed them in three different cell lines (U-2OS, HEK-293T and C2C12). Wild-type DG constructs were compared to their counterparts carrying pathologic missense mutations previously described in patients, namely, L84F, T190M and C667F and with the mutant I591D, i.e., the topological equivalent of V567D identified in zebrafish. Results Live super-resolution fluorescence microscopy showed that the C667F mutant is retained within the ER/Golgi while the T190M and wild-type proteins are correctly localized to the plasma membrane in all 3 cell lines. The L84F mutant exhibits a delay in trafficking to the plasma membrane in two of the cell lines, while localizing strongly at the plasma membrane in the high-expression HEK-293T cells. Similarly, the I591D mutant accumulated at the plasma membrane in the HEK-293T cells, in contrast to the clear retention in the endoplasmic reticulum/Golgi apparatus observed in U-2OS and C2C12 cells. Discussion Our data demonstrate the importance of using a range of different cell lines for a comprehensive study of DG mutants or variants by live cell optical super-resolution microscopy.
Collapse
Affiliation(s)
- Francesca Sciandra
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)-CNR, Rome, Italy
| | - Manuela Bozzi
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)-CNR, Rome, Italy
- Dipartimento di Scienze biotecnologiche di base, Cliniche intensivologiche e perioperatorie, Sezione di Biochimica e Biochimica Clinica, Catholic University of the Sacred Heart, Rome, Italy
| | - Alina Witt
- Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Paul Goffing
- Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Sonia Covaceuszach
- CNR - Istituto di Cristallografia – Sede Secondaria di Trieste, Trieste, Italy
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alberto Cassetta
- CNR - Istituto di Cristallografia – Sede Secondaria di Trieste, Trieste, Italy
| | - Maria Giulia Bigotti
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol, United Kingdom
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Thomas Huser
- Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Andrea Brancaccio
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)-CNR, Rome, Italy
- School of Biochemistry, Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Wolfgang Hübner
- Department of Physics, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
2
|
Karakosta C, Samiotaki M, Panayotou G, Papakonstantinou D, Moschos MM. Role of actin-binding proteins in cataract formation. Cytoskeleton (Hoboken) 2025; 82:98-110. [PMID: 38958111 PMCID: PMC11904859 DOI: 10.1002/cm.21889] [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: 02/02/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Actin has been implicated in lens opacification; however, the specific actin-related pathways involved in cataracts remain unelucidated. In this study, actin-related proteome changes and signaling pathways involved in the development of cataracts were evaluated. METHODS The anterior capsule and phacoemulsification (phaco) cassette contents were collected during cataract surgery from 11 patients with diabetic cataract (DC), 12 patients with age-related cataract (ARC), and seven patients with post-vitrectomy cataract (PVC). Untargeted, global identification and quantification of proteins was performed through liquid chromatography-mass spectrometry with the data-independent acquisition (DIA). RESULTS In phaco cassette samples, proteins with significantly lower expression in ARC than in DC and PVC were involved in various pathways, including actin binding, actin cytoskeleton reorganization, actin filament capping, cortical actin cytoskeleton organization, and small GTPase-mediated signal transduction pathways. In anterior capsules, proteins with significantly lower expression in ARC than in DC and PVC were involved in actin binding and actin cytoskeleton reorganization pathways. CONCLUSION Actin cytoskeleton and actin-binding proteins are involved in lens fiber elongation and differentiation. Rho GTPases contribute to actin cytoskeletal reorganization, and their inactivation is linked to abnormal lens fiber migration. These findings link actin binding to lens fiber integrity, lens opacification, and cataracts.
Collapse
Affiliation(s)
- Christina Karakosta
- School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | | | - George Panayotou
- Biomedical Sciences Research Center Alexander FlemingAtticaGreece
| | - Dimitrios Papakonstantinou
- 1st University Eye Clinic, G. Gennimatas General Hospital of AthensNational and Kapodistrian University of AthensAthensGreece
| | - Marilita M. Moschos
- Department of Electrophysiology of Vision1st University Eye Clinic of AthensAthensGreece
| |
Collapse
|
3
|
Anderson MJM, Hayward AN, Smiley AT, Shi K, Pawlak MR, Aird EJ, Grant E, Greenberg L, Aihara H, Evans RL, Ulens C, Gordon WR. Molecular basis of proteolytic cleavage regulation by the extracellular matrix receptor dystroglycan. Structure 2024; 32:1984-1996.e5. [PMID: 39305901 PMCID: PMC11560575 DOI: 10.1016/j.str.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Abstract
The dystrophin-glycoprotein-complex (DGC), anchored by the transmembrane protein dystroglycan, functions to mechanically link the extracellular matrix and actin cytoskeleton. Breaking this connection is associated with diseases such as muscular dystrophy, yet cleavage of dystroglycan by matrix-metalloproteinases (MMPs) remains an understudied mechanism to disrupt the DGC. We determined the crystal structure of the membrane-adjacent domain (amino acids 491-722) of E. coli expressed human dystroglycan to understand MMP cleavage regulation. The structural model includes tandem immunoglobulin-like (IGL) and sperm/enterokinase/agrin-like (SEAL) domains, which support proteolysis in diverse receptors to facilitate mechanotransduction, membrane protection, and viral entry. The structure reveals a C-terminal extension that buries the MMP site by packing into a hydrophobic pocket, a unique mechanism of MMP cleavage regulation. We further demonstrate structure-guided and disease-associated mutations disrupt proteolytic regulation using a cell-surface proteolysis assay. Thus disrupted proteolysis is a potentially relevant mechanism for "breaking" the DGC link to contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Michael J M Anderson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Adam T Smiley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Matthew R Pawlak
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Eric J Aird
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; Currently at Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Eva Grant
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Lauren Greenberg
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Robert L Evans
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Christopher Ulens
- Department of Cellular and Molecular Medicine, Karolinksa University Leuven, 3000 Leuven, Belgium
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
4
|
Karas BF, Terez KR, Mowla S, Battula N, Flannery KP, Gural BM, Aboussleman G, Mubin N, Manzini MC. Removal of pomt1 in zebrafish leads to loss of α-dystroglycan glycosylation and dystroglycanopathy phenotypes. Hum Mol Genet 2024; 33:709-723. [PMID: 38272461 PMCID: PMC11000664 DOI: 10.1093/hmg/ddae006] [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: 07/11/2023] [Revised: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Biallelic mutations in Protein O-mannosyltransferase 1 (POMT1) are among the most common causes of a severe group of congenital muscular dystrophies (CMDs) known as dystroglycanopathies. POMT1 is a glycosyltransferase responsible for the attachment of a functional glycan mediating interactions between the transmembrane glycoprotein dystroglycan and its binding partners in the extracellular matrix (ECM). Disruptions in these cell-ECM interactions lead to multiple developmental defects causing brain and eye malformations in addition to CMD. Removing Pomt1 in the mouse leads to early embryonic death due to the essential role of dystroglycan during placental formation in rodents. Here, we characterized and validated a model of pomt1 loss of function in the zebrafish showing that developmental defects found in individuals affected by dystroglycanopathies can be recapitulated in the fish. We also discovered that pomt1 mRNA provided by the mother in the oocyte supports dystroglycan glycosylation during the first few weeks of development. Muscle disease, retinal synapse formation deficits, and axon guidance defects can only be uncovered during the first week post fertilization by generating knock-out embryos from knock-out mothers. Conversely, maternal pomt1 from heterozygous mothers was sufficient to sustain muscle, eye, and brain development only leading to loss of photoreceptor synapses at 30 days post fertilization. Our findings show that it is important to define the contribution of maternal mRNA while developing zebrafish models of dystroglycanopathies and that offspring generated from heterozygous and knock-out mothers can be used to differentiate the role of dystroglycan glycosylation in tissue formation and maintenance.
Collapse
Affiliation(s)
- Brittany F Karas
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Kristin R Terez
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Shorbon Mowla
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Namarata Battula
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Kyle P Flannery
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Brian M Gural
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Grace Aboussleman
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Numa Mubin
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| |
Collapse
|
5
|
Syed OA, Tsang B, Gerlai R. The zebrafish for preclinical psilocybin research. Neurosci Biobehav Rev 2023; 153:105381. [PMID: 37689090 DOI: 10.1016/j.neubiorev.2023.105381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/11/2023]
Abstract
In this review, we discuss the possible utility of zebrafish in research on psilocybin, a psychedelic drug whose recreational use as well as possible clinical application are gaining increasing interest. First, we review behavioral tests with zebrafish, focussing on anxiety and social behavior, which have particular relevance in the context of psilocybin research. Next, we briefly consider methods of genetic manipulations with which psilocybin's phenotypical effects and underlying mechanisms may be investigated in zebrafish. We briefly review the known mechanisms of psilocybin, and also discuss what we know about its safety and toxicity profile. Last, we discuss examples of how psilocybin may be employed for testing treatment efficacy in preclinical research for affective disorders in zebrafish. We conclude that zebrafish has a promising future in preclinical research on psychedelic drugs.
Collapse
Affiliation(s)
- Omer A Syed
- Department of Biology, University of Toronto Mississauga, Canada.
| | - Benjamin Tsang
- Department of Cell & Systems Biology, University of Toronto, Canada.
| | - Robert Gerlai
- Department of Cell & Systems Biology, University of Toronto, Canada; Department of Psychology, University of Toronto Mississauga, Canada.
| |
Collapse
|
6
|
Shivanna K, Astumian M, Raut P, Ngo VN, Hess ST, Henry C. Super-Resolution Imaging Reveals the Nanoscale Distributions of Dystroglycan and Integrin Itga7 in Zebrafish Muscle Fibers. Biomedicines 2023; 11:1941. [PMID: 37509580 PMCID: PMC10377463 DOI: 10.3390/biomedicines11071941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Cell signaling is determined partially by the localization and abundance of proteins. Dystroglycan and integrin are both transmembrane receptors that connect the cytoskeleton inside muscle cells to the extracellular matrix outside muscle cells, maintaining proper adhesion and function of muscle. The position and abundance of Dystroglycan relative to integrins is thought to be important for muscle adhesion and function. The subcellular localization and quantification of these receptor proteins can be determined at the nanometer scale by FPALM super-resolution microscopy. We used FPALM to determine localizations of Dystroglycan and integrin proteins in muscle fibers of intact zebrafish (Danio rerio). Results were consistent with confocal imaging data, but illuminate further details at the nanoscale and show the feasibility of using FPALM to quantify interactions of two proteins in a whole organism.
Collapse
Affiliation(s)
- Komala Shivanna
- Department of Physics & Astronomy, University of Maine, 5709 Bennett Hall, Orono, ME 04469-5709, USA; (K.S.); (P.R.); (V.-N.N.)
| | - Mary Astumian
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469-5751, USA;
| | - Prakash Raut
- Department of Physics & Astronomy, University of Maine, 5709 Bennett Hall, Orono, ME 04469-5709, USA; (K.S.); (P.R.); (V.-N.N.)
| | - Vinh-Nhan Ngo
- Department of Physics & Astronomy, University of Maine, 5709 Bennett Hall, Orono, ME 04469-5709, USA; (K.S.); (P.R.); (V.-N.N.)
| | - Samuel T. Hess
- Department of Physics & Astronomy, University of Maine, 5709 Bennett Hall, Orono, ME 04469-5709, USA; (K.S.); (P.R.); (V.-N.N.)
| | - Clarissa Henry
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469-5751, USA;
| |
Collapse
|
7
|
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
|
8
|
Roles of miR-196a and miR-196b in Zebrafish Motor Function. Biomolecules 2023; 13:biom13030554. [PMID: 36979489 PMCID: PMC10046552 DOI: 10.3390/biom13030554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Background: The exertion of motor function depends on various tissues, such as bones and muscles. miR-196 has been widely studied in cancer and other fields, but its effect on bone and skeletal muscle is rarely reported. In order to explore the role of miR-196 family in bone and skeletal muscle, we used the previously successfully constructed miR-196a-1 and miR-196b gene knockout zebrafish animal models for research. Methods: The behavioral trajectories of zebrafish from 4 days post-fertilization (dpf) to 7 dpf were detected to analyze the effect of miR-196a-1 and miR-196b on motor ability. Hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) were used to detect the dorsal muscle tissue of zebrafish. The bone tissue of zebrafish was detected by microcomputed tomography (micro-CT). Real-time PCR was used to detect the expression levels of related genes, including vcp, dpm1, acta1b, mylpfb, col1a1a, bmp8a, gdf6a, and fgfr3. Results: The behavioral test showed that the total behavioral trajectory, movement time, and movement speed of zebrafish larvae were decreased in the miR-196a-1 and miR-196b gene knockout lines. Muscle tissue analysis showed that the structure of muscle fibers in the zebrafish lacking miR-196a-1 and miR-196b was abnormal and was characterized by vacuolar degeneration of muscle fibers, intranuclear migration, melanin deposition, and inflammatory cell infiltration. Bone CT examination revealed decreased bone mineral density and trabecular bone number. The real-time PCR results showed that the expression levels of vcp, dpm1, gdf6a, fgfr3, and col1a1a were decreased in the miR-196b gene knockout group. The expression levels of dpm1, acta1b, mylpfb, gdf6a, and col1a1a were decreased, and the expression level of fgfr3 was increased in the miR-196b gene knockout group compared with the wild-type group. Conclusions: miR-196a-1 and miR-196b play an important role in muscle fiber structure, bone mineral density, and bone trabecular quantity by affecting the expression of vcp, dpm1, acta1b, mylpfb, gdf6a, fgfr3, and col1a1a and then affect the function of the motor system
Collapse
|
9
|
Pipalia TG, Sultan SHA, Koth J, Knight RD, Hughes SM. Skeletal Muscle Regeneration in Zebrafish. Methods Mol Biol 2023; 2640:227-248. [PMID: 36995599 DOI: 10.1007/978-1-0716-3036-5_17] [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] [Indexed: 03/31/2023]
Abstract
Muscle regeneration models have revealed mechanisms of inflammation, wound clearance, and stem cell-directed repair of damage, thereby informing therapy. Whereas studies of muscle repair are most advanced in rodents, the zebrafish is emerging as an additional model organism with genetic and optical advantages. Various muscle wounding protocols (both chemical and physical) have been published. Here we describe simple, cheap, precise, adaptable, and effective wounding protocols and analysis methods for two stages of a larval zebrafish skeletal muscle regeneration model. We show examples of how muscle damage, ingression of muscle stem cells, immune cells, and regeneration of fibers can be monitored over an extended timecourse in individual larvae. Such analyses have the potential to greatly enhance understanding, by reducing the need to average regeneration responses across individuals subjected to an unavoidably variable wound stimulus.
Collapse
Affiliation(s)
- Tapan G Pipalia
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Sami H A Sultan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Jana Koth
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Robert D Knight
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Simon M Hughes
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
10
|
Cubilla M, Papazoglu G, Asteggiano C. Dystroglycanopathies: Genetic Bases of Muscular Dystrophies Due to Alteration in the O-Glycosylation of α-Dystroglycan. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2023; 11. [DOI: 10.1590/2326-4594-jiems-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Affiliation(s)
- M.A. Cubilla
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| | - G.M. Papazoglu
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| | - C.G. Asteggiano
- Hospital de Niños de la Santísima Trinidad, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina; Universidad Católica de Córdoba, Argentina
| |
Collapse
|
11
|
Effect of Polydeoxyribonucleotide (PDRN) Treatment on Corneal Wound Healing in Zebrafish ( Danio rerio). Int J Mol Sci 2022; 23:ijms232113525. [PMID: 36362312 PMCID: PMC9659220 DOI: 10.3390/ijms232113525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
This study aimed to develop a corneal epithelial injury model in zebrafish (Danio rerio) and investigate the effectiveness of polydeoxyribonucleotide (PDRN) treatment on in vivo corneal epithelial regeneration and wound healing. Chemical injury to zebrafish cornea was produced by placing a small cotton swab containing 3% acetic acid solution. PDRN treatment was performed by immersing corneal-injured zebrafish in water containing PDRN (2 mg/mL) for 10 min at 0, 24, 48, and 72 h post-injury (hpi). The level of corneal healing was evaluated by fluorescein staining, histological examination, transcriptional profiling, and immunoblotting techniques. Fluorescein staining results demonstrate that PDRN treatment significantly (p < 0.05) reduced the wounded area of the zebrafish eye at 48 and 72 hpi, suggesting that PDRN may accelerate the corneal re-epithelialization. Histopathological evaluation revealed that injured corneal epithelial cells were re-organized at 72 hpi upon PDRN treatment with increased goblet cell density and size. Moreover, transcriptional analysis results demonstrate that PDRN treatment induced the mRNA expression of adora2ab (6.3-fold), pax6a (7.8-fold), pax6b (29.3-fold), klf4 (7.3-fold), and muc2.1 (5.0-fold) after the first treatment. Besides, tnf-α (2.0-fold) and heat-shock proteins (hsp70; 2.8-fold and hsp90ab1; 1.6-fold) have modulated the gene expression following the PDRN treatment. Immunoblotting results convincingly confirmed the modulation of Mmp-9, Hsp70, and Tnf-α expression levels upon PDRN treatment. Overall, our corneal injury model in zebrafish allows for understanding the morphological and molecular events of corneal epithelial healing, and ophthalmic responses for PDRN treatment following acid injury in zebrafish.
Collapse
|
12
|
Lescouzères L, Bordignon B, Bomont P. Development of a high-throughput tailored imaging method in zebrafish to understand and treat neuromuscular diseases. Front Mol Neurosci 2022; 15:956582. [PMID: 36204134 PMCID: PMC9530744 DOI: 10.3389/fnmol.2022.956582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
The zebrafish (Danio rerio) is a vertebrate species offering multitude of advantages for the study of conserved biological systems in human and has considerably enriched our knowledge in developmental biology and physiology. Being equally important in medical research, the zebrafish has become a critical tool in the fields of diagnosis, gene discovery, disease modeling, and pharmacology-based therapy. Studies on the zebrafish neuromuscular system allowed for deciphering key molecular pathways in this tissue, and established it as a model of choice to study numerous motor neurons, neuromuscular junctions, and muscle diseases. Starting with the similarities of the zebrafish neuromuscular system with the human system, we review disease models associated with the neuromuscular system to focus on current methodologies employed to study them and outline their caveats. In particular, we put in perspective the necessity to develop standardized and high-resolution methodologies that are necessary to deepen our understanding of not only fundamental signaling pathways in a healthy tissue but also the changes leading to disease phenotype outbreaks, and offer templates for high-content screening strategies. While the development of high-throughput methodologies is underway for motility assays, there is no automated approach to quantify the key molecular cues of the neuromuscular junction. Here, we provide a novel high-throughput imaging methodology in the zebrafish that is standardized, highly resolutive, quantitative, and fit for drug screening. By providing a proof of concept for its robustness in identifying novel molecular players and therapeutic drugs in giant axonal neuropathy (GAN) disease, we foresee that this new tool could be useful for both fundamental and biomedical research.
Collapse
Affiliation(s)
- Léa Lescouzères
- ERC Team, Institut NeuroMyoGéne-PGNM, Inserm U1315, CNRS UMR 5261, Claude Bernard University Lyon 1, Lyon, France
| | - Benoît Bordignon
- Montpellier Ressources Imagerie, BioCampus, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Pascale Bomont
- ERC Team, Institut NeuroMyoGéne-PGNM, Inserm U1315, CNRS UMR 5261, Claude Bernard University Lyon 1, Lyon, France
| |
Collapse
|
13
|
Boyd A, Montandon M, Wood AJ, Currie PD. FKRP directed fibronectin glycosylation: A novel mechanism giving insights into muscular dystrophies? Bioessays 2022; 44:e2100270. [PMID: 35229908 DOI: 10.1002/bies.202100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
Abstract
The recently uncovered role of Fukutin-related protein (FKRP) in fibronectin glycosylation has challenged our understanding of the basis of disease pathogenesis in the muscular dystrophies. FKRP is a Golgi-resident glycosyltransferase implicated in a broad spectrum of muscular dystrophy (MD) pathologies that are not fully attributable to the well-described α-Dystroglycan hypoglycosylation. By revealing a new role for FKRP in the glycosylation of fibronectin, a modification critical for the development of the muscle basement membrane (MBM) and its associated muscle linkages, new possibilities for understanding clinical phenotype arise. This modification involves an interaction between FKRP and myosin-10, a protein involved in the Golgi organization and function. These observations suggest a FKRP nexus exists that controls two critical aspects to muscle fibre integrity, both fibre stability at the MBM and its elastic properties. This review explores the new potential disease axis in the context of our current knowledge of muscular dystrophies.
Collapse
Affiliation(s)
- Andrew Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Margo Montandon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
14
|
Lo Faro V, Nolte IM, Ten Brink JB, Snieder H, Jansonius NM, Bergen AA. Mitochondrial Genome Study Identifies Association Between Primary Open-Angle Glaucoma and Variants in MT-CYB, MT-ND4 Genes and Haplogroups. Front Genet 2021; 12:781189. [PMID: 34976016 PMCID: PMC8719162 DOI: 10.3389/fgene.2021.781189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022] Open
Abstract
Background and purpose: Primary open-angle glaucoma (POAG) is an optic neuropathy characterized by death of retinal ganglion cells and atrophy of the optic nerve head. The susceptibility of the optic nerve to damage has been shown to be mediated by mitochondrial dysfunction. In this study, we aimed to determine a possible association between mitochondrial SNPs or haplogroups and POAG. Methods: Mitochondrial DNA single nucleotide polymorphisms (mtSNPs) were genotyped using the Illumina Infinium Global Screening Array-24 (GSA) 700K array set. Genetic analyses were performed in a POAG case-control study involving the cohorts, Groningen Longitudinal Glaucoma Study-Lifelines Cohort Study and Amsterdam Glaucoma Study, including 721 patients and 1951 controls in total. We excluded samples not passing quality control for nuclear genotypes and samples with low call rate for mitochondrial variation. The mitochondrial variants were analyzed both as SNPs and haplogroups. These were determined with the bioinformatics software HaploGrep, and logistic regression analysis was used for the association, as well as for SNPs. Results: Meta-analysis of the results from both cohorts revealed a significant association between POAG and the allele A of rs2853496 [odds ratio (OR) = 0.64; p = 0.006] within the MT-ND4 gene, and for the T allele of rs35788393 (OR = 0.75; p = 0.041) located in the MT-CYB gene. In the mitochondrial haplogroup analysis, the most significant p-value was reached by haplogroup K (p = 1.2 × 10−05), which increases the risk of POAG with an OR of 5.8 (95% CI 2.7–13.1). Conclusion: We identified an association between POAG and polymorphisms in the mitochondrial genes MT-ND4 (rs2853496) and MT-CYB (rs35788393), and with haplogroup K. The present study provides further evidence that mitochondrial genome variations are implicated in POAG. Further genetic and functional studies are required to substantiate the association between mitochondrial gene polymorphisms and POAG and to define the pathophysiological mechanisms of mitochondrial dysfunction in glaucoma.
Collapse
Affiliation(s)
- Valeria Lo Faro
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jacoline B. Ten Brink
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nomdo M. Jansonius
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Arthur A. Bergen
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, Netherlands
- *Correspondence: Arthur A. Bergen,
| |
Collapse
|
15
|
Sun CC, Zhou ZQ, Chen ZL, Zhu RK, Yang D, Peng XY, Zheng L, Tang CF. Identification of Potentially Related Genes and Mechanisms Involved in Skeletal Muscle Atrophy Induced by Excessive Exercise in Zebrafish. BIOLOGY 2021; 10:biology10080761. [PMID: 34439993 PMCID: PMC8389602 DOI: 10.3390/biology10080761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 01/02/2023]
Abstract
Long-term imbalance between fatigue and recovery may eventually lead to muscle weakness or even atrophy. We previously reported that excessive exercise induces pathological cardiac hypertrophy. However, the effect of excessive exercise on the skeletal muscles remains unclear. In the present study, we successfully established an excessive-exercise-induced skeletal muscle atrophy zebrafish model, with decreased muscle fiber size, critical swimming speed, and maximal oxygen consumption. High-throughput RNA-seq analysis identified differentially expressed genes in the model system compared with control zebrafish. Gene ontology and KEGG enrichment analysis revealed that the upregulated genes were enriched in autophagy, homeostasis, circadian rhythm, response to oxidative stress, apoptosis, the p53 signaling pathway, and the FoxO signaling pathway. Protein-protein interaction network analysis identified several hub genes, including keap1b, per3, ulk1b, socs2, esrp1, bcl2l1, hsp70, igf2r, mdm2, rab18a, col1a1a, fn1a, ppih, tpx2, uba5, nhlrc2, mcm4, tac1, b3gat3, and ddost, that correlate with the pathogenesis of skeletal muscle atrophy induced by excessive exercise. The underlying regulatory pathways and muscle-pressure-response-related genes identified in the present study will provide valuable insights for prescribing safe and accurate exercise programs for athletes and the supervision and clinical treatment of muscle atrophy induced by excessive exercise.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lan Zheng
- Correspondence: (X.-Y.P.); (L.Z.); (C.-F.T.)
| | | |
Collapse
|
16
|
Zhou Z, Zheng L, Tang C, Chen Z, Zhu R, Peng X, Wu X, Zhu P. Identification of Potentially Relevant Genes for Excessive Exercise-Induced Pathological Cardiac Hypertrophy in Zebrafish. Front Physiol 2020; 11:565307. [PMID: 33329019 PMCID: PMC7734032 DOI: 10.3389/fphys.2020.565307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022] Open
Abstract
Exercise-induced cardiac remodeling has aroused public concern for some time, as sudden cardiac death is known to occur in athletes; however, little is known about the underlying mechanism of exercise-induced cardiac injury. In the present study, we established an excessive exercise-induced pathologic cardiac hypertrophy model in zebrafish with increased myocardial fibrosis, myofibril disassembly, mitochondrial degradation, upregulated expression of the pathological hypertrophy marker genes in the heart, contractile impairment, and cardiopulmonary function impairment. High-throughput RNA-seq analysis revealed that the differentially expressed genes were enriched in the regulation of autophagy, protein folding, and degradation, myofibril development, angiogenesis, metabolic reprogramming, and insulin and FoxO signaling pathways. FOXO proteins may be the core mediator of the regulatory network needed to promote the pathological response. Further, PPI network analysis showed that pik3c3, gapdh, fbox32, fzr1, ubox5, lmo7a, kctd7, fbxo9, lonrf1l, fbxl4, nhpb2l1b, nhp2, fbl, hsp90aa1.1, snrpd3l, dhx15, mrto4, ruvbl1, hspa8b, and faub are the hub genes that correlate with the pathogenesis of pathological cardiac hypertrophy. The underlying regulatory pathways and cardiac pressure-responsive molecules identified in the present study will provide valuable insights for the supervision and clinical treatment of pathological cardiac hypertrophy induced by excessive exercise.
Collapse
Affiliation(s)
- Zuoqiong Zhou
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Changfa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zhanglin Chen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Runkang Zhu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiushan Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
17
|
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
|
18
|
Li Y, Yang L, Yang J, Shi J, Chai P, Ge S, Wang Y, Fan X, Jia R. A novel variant in GPAA1, encoding a GPI transamidase complex protein, causes inherited vascular anomalies with various phenotypes. Hum Genet 2020; 139:1499-1511. [PMID: 32533362 DOI: 10.1007/s00439-020-02192-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Vascular anomalies (VAs), comprising wide subtypes of tumors and malformations, are often caused by variants in multiple tyrosine kinase (TK) receptor signaling pathways including TIE2, PIK3CA and GNAQ/11. Yet, a portion of individuals with clinical features of VA do not have variants in these genes, suggesting that there are undiscovered pathogenic factors underlying these patients and possibly with overlapping phenotypes. Here, we identified one rare non-synonymous variant (c.968A > G) in the seventh exon of GPAA1 (Glycosylphosphatidylinositol Anchor Attachment Protein 1), shared by the four affected members of a large pedigree with multiple types of VA using whole-exome sequencing. GPAA1 encodes a glycosylphosphatidylinositol (GPI) transamidase complex protein. This complex orchestrates the attachment of the GPI anchor to the C terminus of precursor proteins in the endoplasmic reticulum (ER). We showed such variant led to scarce expression of GPAA1 protein in vascular endothelium and induced a localization change from ER membrane to cytoplasm and nucleus. In addition, expressing wild-type GPAA1 in endothelial cells had an effect to inhibit cell proliferation and migration, while expressing variant GPAA1 led to overgrowth and overmigration, indicating a loss of the quiescent status. Finally, a gpaa1-deficient zebrafish model displayed several types of developmental defects as well as vascular dysplasia, demonstrating that GPAA1 is involved in angiogenesis and vascular remodeling. Altogether, our results indicate that the rare coding variant in GPAA1 (c.968A > G) is causally related to familial forms of VAs.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Liu Yang
- The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yefei Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Huangpu District, Shanghai, 200001, China. .,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
19
|
Liu Y, Yu M, Shang X, Nguyen MHH, Balakrishnan S, Sager R, Hu H. Eyes shut homolog (EYS) interacts with matriglycan of O-mannosyl glycans whose deficiency results in EYS mislocalization and degeneration of photoreceptors. Sci Rep 2020; 10:7795. [PMID: 32385361 PMCID: PMC7210881 DOI: 10.1038/s41598-020-64752-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in eyes shut homolog (EYS), a secreted extracellular matrix protein containing multiple laminin globular (LG) domains, and in protein O-mannose β1, 2-N-acetylglucosaminyl transferase 1 (POMGnT1), an enzyme involved in O-mannosyl glycosylation, cause retinitis pigmentosa (RP), RP25 and RP76, respectively. How EYS and POMGnT1 regulate photoreceptor survival is poorly understood. Since some LG domain-containing proteins function by binding to the matriglycan moiety of O-mannosyl glycans, we hypothesized that EYS interacted with matriglycans as well. To test this hypothesis, we performed EYS Far-Western blotting assay and generated pomgnt1 mutant zebrafish. The results showed that EYS bound to matriglycans. Pomgnt1 mutation in zebrafish resulted in a loss of matriglycan, retention of synaptotagmin-1-positive EYS secretory vesicles within the outer nuclear layer, and diminished EYS protein near the connecting cilia. Photoreceptor density in 2-month old pomgnt1 mutant retina was similar to the wild-type animals but was significantly reduced at 6-months. These results indicate that EYS protein localization to the connecting cilia requires interaction with the matriglycan and that O-mannosyl glycosylation is required for photoreceptor survival in zebrafish. This study identified a novel interaction between EYS and matriglycan demonstrating that RP25 and RP76 are mechanistically linked in that O-mannosyl glycosylation controls targeting of EYS protein.
Collapse
Affiliation(s)
- Yu Liu
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Miao Yu
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Xuanze Shang
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
| | - My Hong Hoai Nguyen
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Biological Sciences, State University of New York at Plattsburgh, 101 Broad St., Plattsburgh, New York, 12901, USA
| | - Shanmuganathan Balakrishnan
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Rachel Sager
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Huaiyu Hu
- Center for Vision Research, Departments of Neuroscience and Physiology and of Ophthalmology and Visual Sciences, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
20
|
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
|
21
|
Rojas-Benítez D, L. Allende M. Elongator Subunit 3 (Elp3) Is Required for Zebrafish Trunk Development. Int J Mol Sci 2020; 21:E925. [PMID: 32023806 PMCID: PMC7036906 DOI: 10.3390/ijms21030925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Transfer RNAs (tRNAs) are the most post-transcriptionally modified RNA species. Some of these modifications, especially the ones located in the anti-codon loop, are required for decoding capabilities of tRNAs. Such is the case for 5-methoxy-carbonyl-methyl-2-thio-uridine (mcm5s2U), synthetized by the Elongator complex. Mutants for its sub-units display pleiotropic phenotypes. In this paper, we analyze the role of elp3 (Elongator catalytic sub-unit) in zebrafish development. We found that it is required for trunk development; elp3 knock-down animals presented diminished levels of mcm5s2U and sonic hedgehog (Shh) signaling activity. Activation of this pathway was sufficient to revert the phenotype caused by elp3 knockdown, indicating a functional relationship between Elongator and Shh through a yet unknown molecular mechanism.
Collapse
Affiliation(s)
- Diego Rojas-Benítez
- FONDAP Center for Genome Regulation (CGR), Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile;
| | | |
Collapse
|
22
|
Zebrafish structural development in Mueller-matrix scanning microscopy. Sci Rep 2019; 9:19974. [PMID: 31882853 PMCID: PMC6934882 DOI: 10.1038/s41598-019-56610-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/16/2019] [Indexed: 11/18/2022] Open
Abstract
Zebrafish are powerful animal models for understanding biological processes and the molecular mechanisms involved in different human diseases. Advanced optical techniques based on fluorescence microscopy have become the main imaging method to characterize the development of these organisms at the microscopic level. However, the need for fluorescence probes and the consequent high light doses required to excite fluorophores can affect the biological process under observation including modification of metabolic function or phototoxicity. Here, without using any labels, we propose an implementation of a Mueller-matrix polarimeter into a commercial optical scanning microscope to characterize the polarimetric transformation of zebrafish preserved at different embryonic developmental stages. By combining the full polarimetric measurements with statistical analysis of the Lu and Chipman mathematical decomposition, we demonstrate that it is possible to quantify the structural changes of the biological organization of fixed zebrafish embryos and larvae at the cellular scale. This convenient implementation, with low light intensity requirement and cheap price, coupled with the quantitative nature of Mueller-matrix formalism, can pave the way for a better understanding of developmental biology, in which label-free techniques become a standard tool to study organisms.
Collapse
|
23
|
Ma J, Lwigale P. Transformation of the Transcriptomic Profile of Mouse Periocular Mesenchyme During Formation of the Embryonic Cornea. Invest Ophthalmol Vis Sci 2019; 60:661-676. [PMID: 30786278 PMCID: PMC6383728 DOI: 10.1167/iovs.18-26018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Defects in neural crest development are a major contributing factor in corneal dysgenesis, but little is known about the genetic landscape during corneal development. The purpose of this study was to provide a detailed transcriptome profile and evaluate changes in gene expression during mouse corneal development. Methods RNA sequencing was used to uncover the transcriptomic profile of periocular mesenchyme (pNC) isolated at embryonic day (E) 10.5 and corneas isolated at E14.5 and E16.5. The spatiotemporal expression of several differentially expressed genes was validated by in situ hybridization. Results Analysis of the whole-transcriptome profile between pNC and embryonic corneas identified 3815 unique differentially expressed genes. Pathway analysis revealed an enrichment of differentially expressed genes involved in signal transduction (retinoic acid, transforming growth factor-β, and Wnt pathways) and transcriptional regulation. Conclusions Our analyses, for the first time, identify a large number of differentially expressed genes during progressive stages of mouse corneal development. Our data provide a comprehensive transcriptomic profile of the developing cornea. Combined, these data serve as a valuable resource for the identification of novel regulatory networks crucial for the advancement of studies in congenital defects, stem cell therapy, bioengineering, and adult corneal diseases.
Collapse
Affiliation(s)
- Justin Ma
- BioSciences Department, Rice University, Houston, Texas, United States
| | - Peter Lwigale
- BioSciences Department, Rice University, Houston, Texas, United States
| |
Collapse
|
24
|
Brancaccio A. A molecular overview of the primary dystroglycanopathies. J Cell Mol Med 2019; 23:3058-3062. [PMID: 30838779 PMCID: PMC6484290 DOI: 10.1111/jcmm.14218] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 01/17/2023] Open
Abstract
Dystroglycan is a major non-integrin adhesion complex that connects the cytoskeleton to the surrounding basement membranes, thus providing stability to skeletal muscle. In Vertebrates, hypoglycosylation of α-dystroglycan has been strongly linked to muscular dystrophy phenotypes, some of which also show variable degrees of cognitive impairments, collectively termed dystroglycanopathies. Only a small number of mutations in the dystroglycan gene, leading to the so called primary dystroglycanopathies, has been described so far, as opposed to the ever-growing number of identified secondary or tertiary dystroglycanopathies (caused by genetic abnormalities in glycosyltransferases or in enzymes involved in the synthesis of the carbohydrate building blocks). The few mutations found within the autonomous N-terminal domain of α-dystroglycan seem to destabilise it to different degrees, without influencing the overall folding and targeting of the dystroglycan complex. On the contrary other mutations, some located at the α/β interface of the dystroglycan complex, seem to be able to interfere with its maturation, thus compromising its stability and eventually leading to the intracellular engulfment and/or partial or even total degradation of the dystroglycan uncleaved precursor.
Collapse
Affiliation(s)
- Andrea Brancaccio
- School of Biochemistry, University of Bristol, Bristol, UK.,Istituto di Chimica del Riconoscimento Molecolare - CNR c/o Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
25
|
Kálmán M, Lőrincz DL, Sebők OM, Ari C, Oszwald E, Somiya H, Jancsik V. Cerebrovascular β-dystroglycan immunoreactivity in vertebrates: not detected in anurans and in the teleosts Ostariophysi and Euteleostei. Integr Zool 2019; 15:16-31. [PMID: 30811839 DOI: 10.1111/1749-4877.12384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The aim of the present paper was to check for the presence of cerebrovascular dystroglycan in vertebrates, because dystroglycan, which is localized in the vascular astroglial end-feet, has a pivotal function in glio-vascular connections. In mammalian brains, the immunoreactivity of β-dystroglycan subunit delineates the vessels. The results of the present study demonstrate similar patterns in other vertebrates, except for anurans and the teleost groups Ostariophysi and Euteleostei. In this study, we investigated 1 or 2 representative species of the main groups of Chondrichthyes, teleost and non-teleost ray-finned fishes, urodeles, anurans, and reptiles. We also investigated 5 mammalian and 3 bird species. Animals were obtained from breeders or fishermen. The presence of β-dystroglycan was investigated immunohistochemically in free-floating sections. Pre-embedding electron microscopical immunohistochemistry on Heterodontus japonicus shark brains demonstrated that in Elasmobranchii, β-dystroglycan is also localized in the perivascular glial end-feet despite the different construction of their blood-brain barrier. The results indicated that the cerebrovascular β-dystroglycan immunoreactivity disappeared separately in anurans, and in teleosts, in the latter group before its division to Ostariophysi and Euteleostei. Immunohistochemistry in muscles and western blots from brain homogenates, however, detected the presence of β-dystroglycan, even in anurans and all teleosts. A possible explanation is that in the glial end-feet, β-dystroglycan is masked in these animals, or disappeared during adaptation to the freshwater habitat.
Collapse
Affiliation(s)
- Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - David L Lőrincz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.,University of Leicester, Dept. of Neuroscience, Psychology and Behaviour, Leicester, United Kingdom
| | - Olivér M Sebők
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csilla Ari
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.,Hyperbaric Neuroscience Research Lab., Dept of Psychology, University of South Florida, Tampa, Florida, USA
| | - Erzsébet Oszwald
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Hiroaki Somiya
- Department of Environmental Biology, Chubu University, Chubu, Japan
| | | |
Collapse
|
26
|
Nickolls AR, Bönnemann CG. The roles of dystroglycan in the nervous system: insights from animal models of muscular dystrophy. Dis Model Mech 2018; 11:11/12/dmm035931. [PMID: 30578246 PMCID: PMC6307911 DOI: 10.1242/dmm.035931] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dystroglycan is a cell membrane protein that binds to the extracellular matrix in a variety of mammalian tissues. The α-subunit of dystroglycan (αDG) is heavily glycosylated, including a special O-mannosyl glycoepitope, relying upon this unique glycosylation to bind its matrix ligands. A distinct group of muscular dystrophies results from specific hypoglycosylation of αDG, and they are frequently associated with central nervous system involvement, ranging from profound brain malformation to intellectual disability without evident morphological defects. There is an expanding literature addressing the function of αDG in the nervous system, with recent reports demonstrating important roles in brain development and in the maintenance of neuronal synapses. Much of these data are derived from an increasingly rich array of experimental animal models. This Review aims to synthesize the information from such diverse models, formulating an up-to-date understanding about the various functions of αDG in neurons and glia of the central and peripheral nervous systems. Where possible, we integrate these data with our knowledge of the human disorders to promote translation from basic mechanistic findings to clinical therapies that take the neural phenotypes into account. Summary: Dystroglycan is a ubiquitous matrix receptor linked to brain and muscle disease. Unraveling the functions of this protein will inform basic and translational research on neural development and muscular dystrophies.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Ma RC, Jacobs CT, Sharma P, Kocha KM, Huang P. Stereotypic generation of axial tenocytes from bipartite sclerotome domains in zebrafish. PLoS Genet 2018; 14:e1007775. [PMID: 30388110 PMCID: PMC6235400 DOI: 10.1371/journal.pgen.1007775] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/14/2018] [Accepted: 10/17/2018] [Indexed: 12/17/2022] Open
Abstract
Development of a functional musculoskeletal system requires coordinated generation of muscles, bones, and tendons. However, how axial tendon cells (tenocytes) are generated during embryo development is still poorly understood. Here, we show that axial tenocytes arise from the sclerotome in zebrafish. In contrast to mouse and chick, the zebrafish sclerotome consists of two separate domains: a ventral domain and a previously undescribed dorsal domain. While dispensable for sclerotome induction, Hedgehog (Hh) signaling is required for the migration and maintenance of sclerotome derived cells. Axial tenocytes are located along the myotendinous junction (MTJ), extending long cellular processes into the intersomitic space. Using time-lapse imaging, we show that both sclerotome domains contribute to tenocytes in a dynamic and stereotypic manner. Tenocytes along a given MTJ always arise from the sclerotome of the adjacent anterior somite. Inhibition of Hh signaling results in loss of tenocytes and enhanced sensitivity to muscle detachment. Together, our work shows that axial tenocytes in zebrafish originate from the sclerotome and are essential for maintaining muscle integrity. The coordinated generation of bones, muscles and tendons at the correct time and location is critical for the development of a functional musculoskeletal system. Although it is well known that tendon is the connective tissue that attaches muscles to bones, it is still poorly understood how tendon cells, or tenocytes, are generated during embryo development. Using the zebrafish model, we identify trunk tenocytes located along the boundary of muscle segments. Using cell tracing in live animals, we find that tenocytes originate from the sclerotome, an embryonic structure that is previously known to generate the trunk skeleton. In contrast to higher vertebrates, the zebrafish sclerotome consists of two separate domains, a ventral domain and a novel dorsal domain. Both domains give rise to trunk tenocytes in a dynamic and stereotypic manner. Hedgehog (Hh) signaling, an important cell signaling pathway, is not required for sclerotome induction but essential for the generation of sclerotome derived cells. Inhibition of Hh signaling leads to loss of tenocytes and increased sensitivity to muscle detachment. Thus, our work shows that tenocytes develop from the sclerotome and play an important role in maintaining muscle integrity.
Collapse
Affiliation(s)
- Roger C. Ma
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Craig T. Jacobs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Priyanka Sharma
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Katrinka M. Kocha
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- * E-mail:
| |
Collapse
|
28
|
Widrick JJ, Gibbs DE, Sanchez B, Gupta VA, Pakula A, Lawrence C, Beggs AH, Kunkel LM. An open source microcontroller based flume for evaluating swimming performance of larval, juvenile, and adult zebrafish. PLoS One 2018; 13:e0199712. [PMID: 29944715 PMCID: PMC6019105 DOI: 10.1371/journal.pone.0199712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 11/25/2022] Open
Abstract
Zebrafish are a preferred vertebrate model for delineating genotype-phenotype relationships. One of the most studied features of zebrafish is their exceptional swimming ability. By 7 days postfertilization (dpf), zebrafish spend over two-thirds of their time engaged in spontaneous swimming activity and several months later they are capable of attaining some of the fastest swimming velocities relative to body length ever recorded in the laboratory. However, laboratory-assembled flumes capable of achieving the slow flow velocities characteristics of larvae as well as the relatively fast maximal velocities of adults have not been described in sufficient detail to allow easy replication. Here we describe an easily assembled, open-source zebrafish-scaled flume for assessing swimming performance. The flume uses two independent spherical-impeller pumps modulated by a microcontroller to achieve flow velocities ranging from 1 to 70 cm s−1. The microcontroller also monitors water temperature and flow velocity and sends these data to a personal computer for real-time display and storage. Incremental protocols for assessing maximal swimming speed (Umax) were developed, stored in custom software, and then uploaded to the microcontroller in order to assess performance of larval (14, 21, 28 dpf), juvenile (35, 42 dpf), and adult (8, 22 month) zebrafish. The flume had sufficient range and sensitivity to detect developmental changes in Umax of larvae and juveniles, an 18–24% faster Umax of adult males vs. females, and a 14–20% age-related reduction in Umax for the oldest zebrafish. Detailed information is provided to assemble and operate this low-cost, versatile, and reliable tool for assessing zebrafish swimming performance.
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, MA, United States of America
- * E-mail:
| | - Devin E. Gibbs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Benjamin Sanchez
- Department of Neurology, Division of Neuromuscular Diseases, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Vandana A. Gupta
- Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Anna Pakula
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Christian Lawrence
- Aquatic Resources Program, Boston Children’s Hospital, Boston, MA, United States of America
| | - Alan H. Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Louis M. Kunkel
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
29
|
Vergara HM, Ramirez J, Rosing T, Nave C, Blandino R, Saw D, Saraf P, Piexoto G, Coombes C, Adams M, Domingo CR. miR-206 is required for changes in cell adhesion that drive muscle cell morphogenesis in Xenopus laevis. Dev Biol 2018; 438:94-110. [PMID: 29596841 DOI: 10.1016/j.ydbio.2018.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are highly conserved small non-coding RNA molecules that post-transcriptionally regulate gene expression in multicellular organisms. Within the set of muscle-specific miRNAs, miR-206 expression is largely restricted to skeletal muscle and is found exclusively within the bony fish lineage. Although many studies have implicated miR-206 in muscle maintenance and disease, its role in skeletal muscle development remains largely unknown. Here, we examine the role of miR-206 during Xenopus laevis somitogenesis. In Xenopus laevis, miR-206 expression coincides with the onset of somitogenesis. We show that both knockdown and over-expression of miR-206 result in abnormal somite formation affecting muscle cell rotation, attachment, and elongation. In particular, our data suggests that miR-206 regulates changes in cell adhesion that affect the ability of newly formed somites to adhere to the notochord as well as to the intersomitic boundaries. Additionally, we show that β-dystroglycan and F-actin expression levels are significantly reduced, suggesting that knockdown of miR-206 levels affects cellular mechanics necessary for cell shape changes and attachments that are required for proper muscle formation.
Collapse
Affiliation(s)
- Hernando Martínez Vergara
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Julio Ramirez
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Trista Rosing
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Ceazar Nave
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Rebecca Blandino
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Daniel Saw
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Parag Saraf
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Gabriel Piexoto
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Coohleen Coombes
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Melissa Adams
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Carmen R Domingo
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA.
| |
Collapse
|
30
|
Bennett AH, O’Donohue MF, Gundry SR, Chan AT, Widrick J, Draper I, Chakraborty A, Zhou Y, Zon LI, Gleizes PE, Beggs AH, Gupta VA. RNA helicase, DDX27 regulates skeletal muscle growth and regeneration by modulation of translational processes. PLoS Genet 2018. [PMID: 29518074 PMCID: PMC5843160 DOI: 10.1371/journal.pgen.1007226] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gene expression in a tissue-specific context depends on the combined efforts of epigenetic, transcriptional and post-transcriptional processes that lead to the production of specific proteins that are important determinants of cellular identity. Ribosomes are a central component of the protein biosynthesis machinery in cells; however, their regulatory roles in the translational control of gene expression in skeletal muscle remain to be defined. In a genetic screen to identify critical regulators of myogenesis, we identified a DEAD-Box RNA helicase, DDX27, that is required for skeletal muscle growth and regeneration. We demonstrate that DDX27 regulates ribosomal RNA (rRNA) maturation, and thereby the ribosome biogenesis and the translation of specific transcripts during myogenesis. These findings provide insight into the translational regulation of gene expression in myogenesis and suggest novel functions for ribosomes in regulating gene expression in skeletal muscles.
Collapse
Affiliation(s)
- Alexis H. Bennett
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marie-Francoise O’Donohue
- Laboratoire de Biologie Moléculaire Eucaryote, Centre de Biologie Intégrative (CBI), Université de Toulouse, UPS, CNRS, France
| | - Stacey R. Gundry
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aye T. Chan
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeffrey Widrick
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Anirban Chakraborty
- Laboratoire de Biologie Moléculaire Eucaryote, Centre de Biologie Intégrative (CBI), Université de Toulouse, UPS, CNRS, France
- Division of Molecular Genetics and Cancer, NU Centre for Science Education and Research, Nitte University, Mangalore, India
| | - Yi Zhou
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Leonard I. Zon
- Stem Cell Program and Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Pierre-Emmanuel Gleizes
- Laboratoire de Biologie Moléculaire Eucaryote, Centre de Biologie Intégrative (CBI), Université de Toulouse, UPS, CNRS, France
| | - Alan H. Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vandana A. Gupta
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
31
|
Thomasi ABD, Sonntag C, Pires DF, Zuidema D, Benci A, Currie PD, Wood AJ. A Low-Cost Pulse Generator for Exacerbating Muscle Fiber Detachment Phenotypes in Zebrafish. Zebrafish 2018; 15:420-424. [PMID: 29381425 DOI: 10.1089/zeb.2017.1512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Muscle fiber detachment from myoseptal boundaries is a common finding in zebrafish models of muscular dystrophies. In some instances, there is a weakening of the interaction between muscle fiber and myosepta, which is yet to manifest as a fiber detachment phenotype. Therefore, to push the fiber detachment of muscle, mutant fish but not their wild-type siblings, beyond their binding threshold, a series of small electrical pulses can be applied to the larvae to create a maximal force contraction and ultimately fiber detachment. To do this, we built a digital pulse generator which delivers four 8 ms 30 V pulses in quick succession, and it has the advantage over older analog approaches to pulse generation because it improves accuracy and is appreciably less expensive. Our pulse generator significantly increases fiber detachment in the laminin-α2 deficient, congenital muscular dystrophy type 1a (MDC1a) model lama2-/- fish when compared with controls.
Collapse
Affiliation(s)
| | - Carmen Sonntag
- 1 Australian Regenerative Medicine Institute, Monash University , Clayton, Victoria, Australia
| | - Danilo Fernando Pires
- 1 Australian Regenerative Medicine Institute, Monash University , Clayton, Victoria, Australia
| | - David Zuidema
- 2 Monash Facility for Instrumentation and Technology Development, Monash University , Clayton, Victoria, Australia
| | - Antonio Benci
- 2 Monash Facility for Instrumentation and Technology Development, Monash University , Clayton, Victoria, Australia
| | - Peter David Currie
- 1 Australian Regenerative Medicine Institute, Monash University , Clayton, Victoria, Australia
| | - Alasdair John Wood
- 1 Australian Regenerative Medicine Institute, Monash University , Clayton, Victoria, Australia
| |
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
|
Covaceuszach S, Bozzi M, Bigotti MG, Sciandra F, Konarev PV, Brancaccio A, Cassetta A. The effect of the pathological V72I, D109N and T190M missense mutations on the molecular structure of α-dystroglycan. PLoS One 2017; 12:e0186110. [PMID: 29036200 PMCID: PMC5643065 DOI: 10.1371/journal.pone.0186110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022] Open
Abstract
Dystroglycan (DG) is a highly glycosylated protein complex that links the cytoskeleton with the extracellular matrix, mediating fundamental physiological functions such as mechanical stability of tissues, matrix organization and cell polarity. A crucial role in the glycosylation of the DG α subunit is played by its own N-terminal region that is required by the glycosyltransferase LARGE. Alteration in this O-glycosylation deeply impairs the high affinity binding to other extracellular matrix proteins such as laminins. Recently, three missense mutations in the gene encoding DG, mapped in the α-DG N-terminal region, were found to be responsible for hypoglycosylated states, causing congenital diseases of different severity referred as primary dystroglycanopaties.To gain insight on the molecular basis of these disorders, we investigated the crystallographic and solution structures of these pathological point mutants, namely V72I, D109N and T190M. Small Angle X-ray Scattering analysis reveals that these mutations affect the structures in solution, altering the distribution between compact and more elongated conformations. These results, supported by biochemical and biophysical assays, point to an altered structural flexibility of the mutant α-DG N-terminal region that may have repercussions on its interaction with LARGE and/or other DG-modifying enzymes, eventually reducing their catalytic efficiency.
Collapse
Affiliation(s)
| | - Manuela Bozzi
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Roma, Italy
- Istituto di Chimica del Riconoscimento Molecolare—CNR c/o Università Cattolica del Sacro Cuore, Roma, Italy
| | | | - Francesca Sciandra
- Istituto di Chimica del Riconoscimento Molecolare—CNR c/o Università Cattolica del Sacro Cuore, Roma, Italy
| | - Petr V. Konarev
- A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, Moscow, Russia
- National Research Centre “Kurchatov Institute”, Moscow, Russia
| | - Andrea Brancaccio
- Istituto di Chimica del Riconoscimento Molecolare—CNR c/o Università Cattolica del Sacro Cuore, Roma, Italy
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Alberto Cassetta
- Istituto di Cristallografia–CNR, Trieste Outstation, Trieste, Italy
| |
Collapse
|
34
|
Xiyuan Z, Fink RHA, Mosqueira M. NO-sGC Pathway Modulates Ca 2+ Release and Muscle Contraction in Zebrafish Skeletal Muscle. Front Physiol 2017; 8:607. [PMID: 28878687 PMCID: PMC5572320 DOI: 10.3389/fphys.2017.00607] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/07/2017] [Indexed: 11/26/2022] Open
Abstract
Vertebrate skeletal muscle contraction and relaxation is a complex process that depends on Ca2+ ions to promote the interaction of actin and myosin. This process can be modulated by nitric oxide (NO), a gas molecule synthesized endogenously by (nitric oxide synthase) NOS isoforms. At nanomolar concentrations NO activates soluble guanylate cyclase (sGC), which in turn activates protein kinase G via conversion of GTP into cyclic GMP. Alternatively, NO post-translationally modifies proteins via S-nitrosylation of the thiol group of cysteine. However, the mechanisms of action of NO on Ca2+ homeostasis during muscle contraction are not fully understood and we hypothesize that NO exerts its effects on Ca2+ homeostasis in skeletal muscles mainly through negative modulation of Ca2+ release and Ca2+ uptake via the NO-sGC-PKG pathway. To address this, we used 5–7 days-post fecundation-larvae of zebrafish, a well-established animal model for physiological and pathophysiological muscle activity. We evaluated the response of muscle contraction and Ca2+ transients in presence of SNAP, a NO-donor, or L-NAME, an unspecific NOS blocker in combination with specific blockers of key proteins of Ca2+ homeostasis. We also evaluate the expression of NOS in combination with dihydropteridine receptor, ryanodine receptor and sarco/endoplasmic reticulum Ca2+ ATPase. We concluded that endogenous NO reduced force production through negative modulation of Ca2+ transients via the NO-sGC pathway. This effect could be reversed using an unspecific NOS blocker or sGC blocker.
Collapse
Affiliation(s)
- Zhou Xiyuan
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, Heidelberg University HospitalHeidelberg, Germany.,Department of Traumatic Surgery, TongJi Hospital affiliated to TongJi Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Rainer H A Fink
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, Heidelberg University HospitalHeidelberg, Germany
| | - Matias Mosqueira
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, Heidelberg University HospitalHeidelberg, Germany
| |
Collapse
|
35
|
Osborn DPS, Pond HL, Mazaheri N, Dejardin J, Munn CJ, Mushref K, Cauley ES, Moroni I, Pasanisi MB, Sellars EA, Hill RS, Partlow JN, Willaert RK, Bharj J, Malamiri RA, Galehdari H, Shariati G, Maroofian R, Mora M, Swan LE, Voit T, Conti FJ, Jamshidi Y, Manzini MC. Mutations in INPP5K Cause a Form of Congenital Muscular Dystrophy Overlapping Marinesco-Sjögren Syndrome and Dystroglycanopathy. Am J Hum Genet 2017; 100:537-545. [PMID: 28190459 PMCID: PMC5339112 DOI: 10.1016/j.ajhg.2017.01.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/05/2017] [Indexed: 02/01/2023] Open
Abstract
Congenital muscular dystrophies display a wide phenotypic and genetic heterogeneity. The combination of clinical, biochemical, and molecular genetic findings must be considered to obtain the precise diagnosis and provide appropriate genetic counselling. Here we report five individuals from four families presenting with variable clinical features including muscular dystrophy with a reduction in dystroglycan glycosylation, short stature, intellectual disability, and cataracts, overlapping both the dystroglycanopathies and Marinesco-Sjögren syndrome. Whole-exome sequencing revealed homozygous missense and compound heterozygous mutations in INPP5K in the affected members of each family. INPP5K encodes the inositol polyphosphate-5-phosphatase K, also known as SKIP (skeletal muscle and kidney enriched inositol phosphatase), which is highly expressed in the brain and muscle. INPP5K localizes to both the endoplasmic reticulum and to actin ruffles in the cytoplasm. It has been shown to regulate myoblast differentiation and has also been implicated in protein processing through its interaction with the ER chaperone HSPA5/BiP. We show that morpholino-mediated inpp5k loss of function in the zebrafish results in shortened body axis, microphthalmia with disorganized lens, microcephaly, reduced touch-evoked motility, and highly disorganized myofibers. Altogether these data demonstrate that mutations in INPP5K cause a congenital muscular dystrophy syndrome with short stature, cataracts, and intellectual disability.
Collapse
Affiliation(s)
- Daniel P S Osborn
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Heather L Pond
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Science, Washington, DC 20037, USA
| | - Neda Mazaheri
- Department of Genetics, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran; Narges Medical Genetics and Prenatal Diagnosis Laboratory, East Mihan Ave., Kianpars, Ahvaz 6155689467, Iran
| | - Jeremy Dejardin
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Christopher J Munn
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Khaloob Mushref
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Science, Washington, DC 20037, USA
| | - Edmund S Cauley
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Science, Washington, DC 20037, USA
| | - Isabella Moroni
- Pediatric Neurology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20133 Milan, Italy
| | - Maria Barbara Pasanisi
- Pediatric Neurology Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20133 Milan, Italy; Division of Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico C. Besta, 20126 Milan, Italy
| | - Elizabeth A Sellars
- Department of Pediatrics, Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - R Sean Hill
- Program in Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jennifer N Partlow
- Program in Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Jaipreet Bharj
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Reza Azizi Malamiri
- Department of Paediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6163764648, Iran
| | - Hamid Galehdari
- Department of Genetics, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran; Narges Medical Genetics and Prenatal Diagnosis Laboratory, East Mihan Ave., Kianpars, Ahvaz 6155689467, Iran
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, East Mihan Ave., Kianpars, Ahvaz 6155689467, Iran; Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Reza Maroofian
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK; University of Exeter Medical School, RILD Wellcome Wolfson Centre, Royal Devon & Exeter NHS Foundation Trust, Exeter EX1 2LU, UK
| | - Marina Mora
- Division of Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico C. Besta, 20126 Milan, Italy
| | - Laura E Swan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Thomas Voit
- NIHR GOSH Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Francesco J Conti
- NIHR GOSH Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Yalda Jamshidi
- Cardiovascular and Cell Sciences Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - M Chiara Manzini
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Science, Washington, DC 20037, USA.
| |
Collapse
|
36
|
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
|
37
|
Congenital myopathy results from misregulation of a muscle Ca2+ channel by mutant Stac3. Proc Natl Acad Sci U S A 2016; 114:E228-E236. [PMID: 28003463 DOI: 10.1073/pnas.1619238114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle contractions are initiated by an increase in Ca2+ released during excitation-contraction (EC) coupling, and defects in EC coupling are associated with human myopathies. EC coupling requires communication between voltage-sensing dihydropyridine receptors (DHPRs) in transverse tubule membrane and Ca2+ release channel ryanodine receptor 1 (RyR1) in the sarcoplasmic reticulum (SR). Stac3 protein (SH3 and cysteine-rich domain 3) is an essential component of the EC coupling apparatus and a mutation in human STAC3 causes the debilitating Native American myopathy (NAM), but the nature of how Stac3 acts on the DHPR and/or RyR1 is unknown. Using electron microscopy, electrophysiology, and dynamic imaging of zebrafish muscle fibers, we find significantly reduced DHPR levels, functionality, and stability in stac3 mutants. Furthermore, stac3NAM myofibers exhibited increased caffeine-induced Ca2+ release across a wide range of concentrations in the absence of altered caffeine sensitivity as well as increased Ca2+ in internal stores, which is consistent with increased SR luminal Ca2+ These findings define critical roles for Stac3 in EC coupling and human disease.
Collapse
|
38
|
Sztal TE, Ruparelia AA, Williams C, Bryson-Richardson RJ. Using Touch-evoked Response and Locomotion Assays to Assess Muscle Performance and Function in Zebrafish. J Vis Exp 2016. [PMID: 27842370 PMCID: PMC5226210 DOI: 10.3791/54431] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Zebrafish muscle development is highly conserved with mammalian systems making them an excellent model to study muscle function and disease. Many myopathies affecting skeletal muscle function can be quickly and easily assessed in zebrafish over the first few days of embryogenesis. By 24 hr post-fertilization (hpf), wildtype zebrafish spontaneously contract their tail muscles and by 48 hpf, zebrafish exhibit controlled swimming behaviors. Reduction in the frequency of, or other alterations in, these movements may indicate a skeletal muscle dysfunction. To analyze swimming behavior and assess muscle performance in early zebrafish development, we utilize both touch-evoked escape response and locomotion assays. Touch-evoked escape response assays can be used to assess muscle performance during short burst movements resulting from contraction of fast-twitch muscle fibers. In response to an external stimulus, which in this case is a tap on the head, wildtype zebrafish at 2 days post-fertilization (dpf) typically exhibit a powerful burst swim, accompanied by sharp turns. Our method quantifies skeletal muscle function by measuring the maximum acceleration during a burst swimming motion, the acceleration being directly proportional to the force produced by muscle contraction. In contrast, locomotion assays during early zebrafish larval development are used to assess muscle performance during sustained periods of muscle activity. Using a tracking system to monitor swimming behavior, we obtain an automated calculation of the frequency of activity and distance in 6-day old zebrafish, reflective of their skeletal muscle function. Measurements of swimming performance are valuable for phenotypic assessment of disease models and high-throughput screening of mutations or chemical treatments affecting skeletal muscle function.
Collapse
|
39
|
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: 27] [Impact Index Per Article: 3.0] [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
|
40
|
Pipalia TG, Koth J, Roy SD, Hammond CL, Kawakami K, Hughes SM. Cellular dynamics of regeneration reveals role of two distinct Pax7 stem cell populations in larval zebrafish muscle repair. Dis Model Mech 2016; 9:671-84. [PMID: 27149989 PMCID: PMC4920144 DOI: 10.1242/dmm.022251] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Heterogeneity of stem cells or their niches is likely to influence tissue regeneration. Here we reveal stem/precursor cell diversity during wound repair in larval zebrafish somitic body muscle using time-lapse 3D confocal microscopy on reporter lines. Skeletal muscle with incision wounds rapidly regenerates both slow and fast muscle fibre types. A swift immune response is followed by an increase in cells at the wound site, many of which express the muscle stem cell marker Pax7. Pax7(+) cells proliferate and then undergo terminal differentiation involving Myogenin accumulation and subsequent loss of Pax7 followed by elongation and fusion to repair fast muscle fibres. Analysis of pax7a and pax7b transgenic reporter fish reveals that cells expressing each of the duplicated pax7 genes are distinctly localised in uninjured larvae. Cells marked by pax7a only or by both pax7a and pax7b enter the wound rapidly and contribute to muscle wound repair, but each behaves differently. Low numbers of pax7a-only cells form nascent fibres. Time-lapse microscopy revealed that the more numerous pax7b-marked cells frequently fuse to pre-existing fibres, contributing more strongly than pax7a-only cells to repair of damaged fibres. pax7b-marked cells are more often present in rows of aligned cells that are observed to fuse into a single fibre, but more rarely contribute to nascent regenerated fibres. Ablation of a substantial portion of nitroreductase-expressing pax7b cells with metronidazole prior to wounding triggered rapid pax7a-only cell accumulation, but this neither inhibited nor augmented pax7a-only cell-derived myogenesis and thus altered the cellular repair dynamics during wound healing. Moreover, pax7a-only cells did not regenerate pax7b cells, suggesting a lineage distinction. We propose a modified founder cell and fusion-competent cell model in which pax7a-only cells initiate fibre formation and pax7b cells contribute to fibre growth. This newly discovered cellular complexity in muscle wound repair raises the possibility that distinct populations of myogenic cells contribute differentially to repair in other vertebrates.
Collapse
Affiliation(s)
- Tapan G Pipalia
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Jana Koth
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, UK
| | - Shukolpa D Roy
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Christina L Hammond
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| |
Collapse
|
41
|
Where do we stand in trial readiness for autosomal recessive limb girdle muscular dystrophies? Neuromuscul Disord 2015; 26:111-25. [PMID: 26810373 DOI: 10.1016/j.nmd.2015.11.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/27/2015] [Accepted: 11/29/2015] [Indexed: 12/20/2022]
Abstract
Autosomal recessive limb girdle muscular dystrophies (LGMD2) are a group of genetically heterogeneous diseases that are typically characterised by progressive weakness and wasting of the shoulder and pelvic girdle muscles. Many of the more than 20 different conditions show overlapping clinical features with other forms of muscular dystrophy, congenital, myofibrillar or even distal myopathies and also with acquired muscle diseases. Although individually extremely rare, all types of LGMD2 together form an important differential diagnostic group among neuromuscular diseases. Despite improved diagnostics and pathomechanistic insight, a curative therapy is currently lacking for any of these diseases. Medical care consists of the symptomatic treatment of complications, aiming to improve life expectancy and quality of life. Besides well characterised pre-clinical tools like animal models and cell culture assays, the determinants of successful drug development programmes for rare diseases include a good understanding of the phenotype and natural history of the disease, the existence of clinically relevant outcome measures, guidance on care standards, up to date patient registries, and, ideally, biomarkers that can help assess disease severity or drug response. Strong patient organisations driving research and successful partnerships between academia, advocacy, industry and regulatory authorities can also help accelerate the elaboration of clinical trials. All these determinants constitute aspects of translational research efforts and influence patient access to therapies. Here we review the current status of determinants of successful drug development programmes for LGMD2, and the challenges of translating promising therapeutic strategies into effective and accessible treatments for patients.
Collapse
|
42
|
Schindler RFR, Scotton C, Zhang J, Passarelli C, Ortiz-Bonnin B, Simrick S, Schwerte T, Poon KL, Fang M, Rinné S, Froese A, Nikolaev VO, Grunert C, Müller T, Tasca G, Sarathchandra P, Drago F, Dallapiccola B, Rapezzi C, Arbustini E, Di Raimo FR, Neri M, Selvatici R, Gualandi F, Fattori F, Pietrangelo A, Li W, Jiang H, Xu X, Bertini E, Decher N, Wang J, Brand T, Ferlini A. POPDC1(S201F) causes muscular dystrophy and arrhythmia by affecting protein trafficking. J Clin Invest 2015; 126:239-53. [PMID: 26642364 DOI: 10.1172/jci79562] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 10/29/2015] [Indexed: 01/16/2023] Open
Abstract
The Popeye domain-containing 1 (POPDC1) gene encodes a plasma membrane-localized cAMP-binding protein that is abundantly expressed in striated muscle. In animal models, POPDC1 is an essential regulator of structure and function of cardiac and skeletal muscle; however, POPDC1 mutations have not been associated with human cardiac and muscular diseases. Here, we have described a homozygous missense variant (c.602C>T, p.S201F) in POPDC1, identified by whole-exome sequencing, in a family of 4 with cardiac arrhythmia and limb-girdle muscular dystrophy (LGMD). This allele was absent in known databases and segregated with the pathological phenotype in this family. We did not find the allele in a further screen of 104 patients with a similar phenotype, suggesting this mutation to be family specific. Compared with WT protein, POPDC1(S201F) displayed a 50% reduction in cAMP affinity, and in skeletal muscle from patients, both POPDC1(S201F) and WT POPDC2 displayed impaired membrane trafficking. Forced expression of POPDC1(S201F) in a murine cardiac muscle cell line (HL-1) increased hyperpolarization and upstroke velocity of the action potential. In zebrafish, expression of the homologous mutation (popdc1(S191F)) caused heart and skeletal muscle phenotypes that resembled those observed in patients. Our study therefore identifies POPDC1 as a disease gene causing a very rare autosomal recessive cardiac arrhythmia and LGMD, expanding the genetic causes of this heterogeneous group of inherited rare diseases.
Collapse
|
43
|
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
|
44
|
Ryckebüsch L. [Potential of the zebrafish model to study congenital muscular dystrophies]. Med Sci (Paris) 2015; 31:912-9. [PMID: 26481031 DOI: 10.1051/medsci/20153110018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In order to better understand the complexity of congenital muscular dystrophies (CMD) and develop new strategies to cure them, it is important to establish new disease models. Due to its numerous helpful attributes, the zebrafish has recently become a very powerful animal model for the study of CMD. For some CMD, this vertebrate model is phenotypically closer to human pathology than the murine model. Over the last few years, researchers have developed innovative techniques to screen rapidly and on a large scale for muscle defects in zebrafish. Furthermore, new genome editing techniques in zebrafish make possible the identification of new disease models. In this review, the major attributes of zebrafish for CMD studies are discussed and the principal models of CMD in zebrafish are highlighted.
Collapse
Affiliation(s)
- Lucile Ryckebüsch
- Division of biological sciences, university of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0380, La Jolla, États-Unis
| |
Collapse
|
45
|
Adams JC, Brancaccio A. The evolution of the dystroglycan complex, a major mediator of muscle integrity. Biol Open 2015; 4:1163-79. [PMID: 26319583 PMCID: PMC4582122 DOI: 10.1242/bio.012468] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Basement membrane (BM) extracellular matrices are crucial for the coordination of different tissue layers. A matrix adhesion receptor that is important for BM function and stability in many mammalian tissues is the dystroglycan (DG) complex. This comprises the non-covalently-associated extracellular α-DG, that interacts with laminin in the BM, and the transmembrane β-DG, that interacts principally with dystrophin to connect to the actin cytoskeleton. Mutations in dystrophin, DG, or several enzymes that glycosylate α-DG underlie severe forms of human muscular dystrophy. Nonwithstanding the pathophysiological importance of the DG complex and its fundamental interest as a non-integrin system of cell-ECM adhesion, the evolution of DG and its interacting proteins is not understood. We analysed the phylogenetic distribution of DG, its proximal binding partners and key processing enzymes in extant metazoan and relevant outgroups. We identify that DG originated after the divergence of ctenophores from porifera and eumetazoa. The C-terminal half of the DG core protein is highly-conserved, yet the N-terminal region, that includes the laminin-binding region, has undergone major lineage-specific divergences. Phylogenetic analysis based on the C-terminal IG2_MAT_NU region identified three distinct clades corresponding to deuterostomes, arthropods, and mollusks/early-diverging metazoans. Whereas the glycosyltransferases that modify α-DG are also present in choanoflagellates, the DG-binding proteins dystrophin and laminin originated at the base of the metazoa, and DG-associated sarcoglycan is restricted to cnidarians and bilaterians. These findings implicate extensive functional diversification of DG within invertebrate lineages and identify the laminin-DG-dystrophin axis as a conserved adhesion system that evolved subsequent to integrin-ECM adhesion, likely to enhance the functional complexity of cell-BM interactions in early metazoans.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Andrea Brancaccio
- Istituto di Chimica del Riconoscimento Molecolare, CNR, Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma 00168, Italy
| |
Collapse
|
46
|
Guiraud S, Aartsma-Rus A, Vieira NM, Davies KE, van Ommen GJB, Kunkel LM. The Pathogenesis and Therapy of Muscular Dystrophies. Annu Rev Genomics Hum Genet 2015; 16:281-308. [DOI: 10.1146/annurev-genom-090314-025003] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Simon Guiraud
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Natassia M. Vieira
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Kay E. Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Gert-Jan B. van Ommen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Louis M. Kunkel
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|
47
|
Genetic Engineering of Dystroglycan in Animal Models of Muscular Dystrophy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:635792. [PMID: 26380289 PMCID: PMC4561298 DOI: 10.1155/2015/635792] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/11/2015] [Indexed: 01/24/2023]
Abstract
In skeletal muscle, dystroglycan (DG) is the central component of the dystrophin-glycoprotein complex (DGC), a multimeric protein complex that ensures a strong mechanical link between the extracellular matrix and the cytoskeleton. Several muscular dystrophies arise from mutations hitting most of the components of the DGC. Mutations within the DG gene (DAG1) have been recently associated with two forms of muscular dystrophy, one displaying a milder and one a more severe phenotype. This review focuses specifically on the animal (murine and others) model systems that have been developed with the aim of directly engineering DAG1 in order to study the DG function in skeletal muscle as well as in other tissues. In the last years, conditional animal models overcoming the embryonic lethality of the DG knock-out in mouse have been generated and helped clarifying the crucial role of DG in skeletal muscle, while an increasing number of studies on knock-in mice are aimed at understanding the contribution of single amino acids to the stability of DG and to the possible development of muscular dystrophy.
Collapse
|
48
|
Zukosky K, Meilleur K, Traynor BJ, Dastgir J, Medne L, Devoto M, Collins J, Rooney J, Zou Y, Yang ML, Gibbs JR, Meier M, Stetefeld J, Finkel RS, Schessl J, Elman L, Felice K, Ferguson TA, Ceyhan-Birsoy O, Beggs AH, Tennekoon G, Johnson JO, Bönnemann CG. Association of a Novel ACTA1 Mutation With a Dominant Progressive Scapuloperoneal Myopathy in an Extended Family. JAMA Neurol 2015; 72:689-98. [PMID: 25938801 DOI: 10.1001/jamaneurol.2015.37] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
IMPORTANCE New genomic strategies can now be applied to identify a diagnosis in patients and families with previously undiagnosed rare genetic conditions. The large family evaluated in the present study was described in 1966 and now expands the phenotype of a known neuromuscular gene. OBJECTIVE To determine the genetic cause of a slowly progressive, autosomal dominant, scapuloperoneal neuromuscular disorder by using linkage and exome sequencing. DESIGN, SETTING, AND PARTICIPANTS Fourteen affected individuals in a 6-generation family with a progressive scapuloperoneal disorder were evaluated. Participants were examined at pediatric, neuromuscular, and research clinics from March 1, 2005, to May 31, 2014. Exome and linkage were performed in genetics laboratories of research institutions. MAIN OUTCOMES AND MEASURES Examination and evaluation by magnetic resonance imaging, ultrasonography, electrodiagnostic studies, and muscle biopsies (n = 3). Genetic analysis included linkage analysis (n = 17) with exome sequencing (n = 7). RESULTS Clinical findings included progressive muscle weakness in an initially scapuloperoneal and distal distribution, including wrist extensor weakness, finger and foot drop, scapular winging, mild facial weakness, Achilles tendon contractures, and diminished or absent deep tendon reflexes. Both age at onset and progression of the disease showed clinical variability within the family. Muscle biopsy specimens demonstrated type I fiber atrophy and trabeculated fibers without nemaline rods. Analysis of exome sequences within the linkage region (4.8 megabases) revealed missense mutation c.591C>A p.Glu197Asp in a highly conserved residue in exon 4 of ACTA1. The mutation cosegregated with disease in all tested individuals and was not present in unaffected individuals. CONCLUSIONS AND RELEVANCE This family defines a new scapuloperoneal phenotype associated with an ACTA1 mutation. A highly conserved protein, ACTA1 is implicated in multiple muscle diseases, including nemaline myopathy, actin aggregate myopathy, fiber-type disproportion, and rod-core myopathy. To our knowledge, mutations in Glu197 have not been reported previously. This residue is highly conserved and located in an exposed position in the protein; the mutation affects the intermolecular and intramolecular electrostatic interactions as shown by structural modeling. The mutation in this residue does not appear to lead to rod formation or actin accumulation in vitro or in vivo, suggesting a different molecular mechanism from that of other ACTA1 diseases.
Collapse
Affiliation(s)
- Kristen Zukosky
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Katherine Meilleur
- National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Jahannaz Dastgir
- Division of Child Neurology, Columbia University Medical Center, New York, New York
| | - Livija Medne
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Marcella Devoto
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia7Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia8Department of Molecular Medicine, Universit
| | - James Collins
- Department of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jachinta Rooney
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland11Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC
| | - Yaqun Zou
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Michele L Yang
- Department of Pediatrics and Neurology, Children's Hospital Colorado, Aurora
| | - J Raphael Gibbs
- Computational Biology Core, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Markus Meier
- Department of Chemistry, Microbiology, Biochemistry, and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Joerg Stetefeld
- Department of Chemistry, Microbiology, Biochemistry, and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard S Finkel
- Department of Pediatrics, Nemours Children's Hospital, Orlando, Florida
| | - Joachim Schessl
- Department of Neurology, Friedrich-Baur Institute, Ludwig-Maximillans University of Munich, Munich, Germany
| | - Lauren Elman
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Kevin Felice
- Department of Neuromuscular Medicine, Hospital for Special Care, New Britain, Connecticut
| | - Toby A Ferguson
- Shriners Pediatric Research Center, Department of Neurology, Temple University, Philadelphia, Pennsylvania
| | - Ozge Ceyhan-Birsoy
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alan H Beggs
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gihan Tennekoon
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Janel O Johnson
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, Porter Neuroscience Research Center, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
49
|
Radev Z, Hermel JM, Elipot Y, Bretaud S, Arnould S, Duchateau P, Ruggiero F, Joly JS, Sohm F. A TALEN-Exon Skipping Design for a Bethlem Myopathy Model in Zebrafish. PLoS One 2015. [PMID: 26221953 PMCID: PMC4519248 DOI: 10.1371/journal.pone.0133986] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Presently, human collagen VI-related diseases such as Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM) remain incurable, emphasizing the need to unravel their etiology and improve their treatments. In UCMD, symptom onset occurs early, and both diseases aggravate with ageing. In zebrafish fry, morpholinos reproduced early UCMD and BM symptoms but did not allow to study the late phenotype. Here, we produced the first zebrafish line with the human mutation frequently found in collagen VI-related disorders such as UCMD and BM. We used a transcription activator-like effector nuclease (TALEN) to design the col6a1ama605003-line with a mutation within an essential splice donor site, in intron 14 of the col6a1 gene, which provoke an in-frame skipping of exon 14 in the processed mRNA. This mutation at a splice donor site is the first example of a template-independent modification of splicing induced in zebrafish using a targetable nuclease. This technique is readily expandable to other organisms and can be instrumental in other disease studies. Histological and ultrastructural analyzes of homozygous and heterozygous mutant fry and 3 months post-fertilization (mpf) fish revealed co-dominantly inherited abnormal myofibers with disorganized myofibrils, enlarged sarcoplasmic reticulum, altered mitochondria and misaligned sarcomeres. Locomotion analyzes showed hypoxia-response behavior in 9 mpf col6a1 mutant unseen in 3 mpf fish. These symptoms worsened with ageing as described in patients with collagen VI deficiency. Thus, the col6a1ama605003-line is the first adult zebrafish model of collagen VI-related diseases; it will be instrumental both for basic research and drug discovery assays focusing on this type of disorders.
Collapse
Affiliation(s)
- Zlatko Radev
- UMS 1374, AMAGEN, INRA, Jouy en Josas, Domaine de Vilvert, France
- UMS 3504, AMAGEN, CNRS, Gif-sur-Yvette, France
| | - Jean-Michel Hermel
- UMR 9197, INRA-CASBAH team, NEURO-Psi, CNRS, Gif sur Yvette, France
- * E-mail: (FS); (JMH)
| | - Yannick Elipot
- UMR 9197, DECA team, NEURO-Psi, CNRS, Gif sur Yvette, France
| | - Sandrine Bretaud
- UMR 5242, Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, CNRS, Université Lyon 1, Lyon, France
| | | | | | - Florence Ruggiero
- UMR 5242, Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, CNRS, Université Lyon 1, Lyon, France
| | | | - Frédéric Sohm
- UMS 1374, AMAGEN, INRA, Jouy en Josas, Domaine de Vilvert, France
- UMS 3504, AMAGEN, CNRS, Gif-sur-Yvette, France
- * E-mail: (FS); (JMH)
| |
Collapse
|
50
|
The dystroglycan: Nestled in an adhesome during embryonic development. Dev Biol 2015; 401:132-42. [DOI: 10.1016/j.ydbio.2014.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/23/2014] [Accepted: 07/08/2014] [Indexed: 01/11/2023]
|