1
|
Emmert ME, Emmert AS, Goh Q, Cornwall R. Sexual dimorphisms in skeletal muscle: current concepts and research horizons. J Appl Physiol (1985) 2024; 137:274-299. [PMID: 38779763 PMCID: PMC11343095 DOI: 10.1152/japplphysiol.00529.2023] [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: 07/31/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
The complex compositional and functional nature of skeletal muscle makes this organ an essential topic of study for biomedical researchers and clinicians. An additional layer of complexity is added with the consideration of sex as a biological variable. Recent research advances have revealed sexual dimorphisms in developmental biology, muscle homeostasis, adaptive responses, and disorders relating to skeletal muscle. Many of the observed sex differences have hormonal and molecular mechanistic underpinnings, whereas others have yet to be elucidated. Future research is needed to investigate the mechanisms dictating sex-based differences in the various aspects of skeletal muscle. As such, it is necessary that skeletal muscle biologists ensure that both female and male subjects are represented in biomedical and clinical studies to facilitate the successful testing and development of therapeutics for all patients.
Collapse
Affiliation(s)
- Marianne E Emmert
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Andrew S Emmert
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Qingnian Goh
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Roger Cornwall
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| |
Collapse
|
2
|
Hiyoshi T, Zhao F, Baba R, Hirakawa T, Kuboki R, Suzuki K, Tomimatsu Y, O'Donnell P, Han S, Zach N, Nakashima M. Electrical impedance myography detects dystrophin-related muscle changes in mdx mice. Skelet Muscle 2023; 13:19. [PMID: 37980539 PMCID: PMC10657153 DOI: 10.1186/s13395-023-00331-1] [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: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND The lack of functional dystrophin protein in Duchenne muscular dystrophy (DMD) causes chronic skeletal muscle inflammation and degeneration. Therefore, the restoration of functional dystrophin levels is a fundamental approach for DMD therapy. Electrical impedance myography (EIM) is an emerging tool that provides noninvasive monitoring of muscle conditions and has been suggested as a treatment response biomarker in diverse indications. Although magnetic resonance imaging (MRI) of skeletal muscles has become a standard measurement in clinical trials for DMD, EIM offers distinct advantages, such as portability, user-friendliness, and reduced cost, allowing for remote monitoring of disease progression or response to therapy. To investigate the potential of EIM as a biomarker for DMD, we compared longitudinal EIM data with MRI/histopathological data from an X-linked muscular dystrophy (mdx) mouse model of DMD. In addition, we investigated whether EIM could detect dystrophin-related changes in muscles using antisense-mediated exon skipping in mdx mice. METHODS The MRI data for muscle T2, the magnetic resonance spectroscopy (MRS) data for fat fraction, and three EIM parameters with histopathology were longitudinally obtained from the hindlimb muscles of wild-type (WT) and mdx mice. In the EIM study, a cell-penetrating peptide (Pip9b2) conjugated antisense phosphorodiamidate morpholino oligomer (PPMO), designed to induce exon-skipping and restore functional dystrophin production, was administered intravenously to mdx mice. RESULTS MRI imaging in mdx mice showed higher T2 intensity at 6 weeks of age in hindlimb muscles compared to WT mice, which decreased at ≥ 9 weeks of age. In contrast, EIM reactance began to decline at 12 weeks of age, with peak reduction at 18 weeks of age in mdx mice. This decline was associated with myofiber atrophy and connective tissue infiltration in the skeletal muscles. Repeated dosing of PPMO (10 mg/kg, 4 times every 2 weeks) in mdx mice led to an increase in muscular dystrophin protein and reversed the decrease in EIM reactance. CONCLUSIONS These findings suggest that muscle T2 MRI is sensitive to the early inflammatory response associated with dystrophin deficiency, whereas EIM provides a valuable biomarker for the noninvasive monitoring of subsequent changes in skeletal muscle composition. Furthermore, EIM reactance has the potential to monitor dystrophin-deficient muscle abnormalities and their recovery in response to antisense-mediated exon skipping.
Collapse
Affiliation(s)
- Tetsuaki Hiyoshi
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Fuqiang Zhao
- Center of Excellence for Imaging, Preclinical and Translational Sciences, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Rina Baba
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Takeshi Hirakawa
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Ryosuke Kuboki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Kazunori Suzuki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yoshiro Tomimatsu
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Patricio O'Donnell
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Steve Han
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Neta Zach
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Masato Nakashima
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
3
|
Benasutti H, Maricelli JW, Seto J, Hall J, Halbert C, Wicki J, Heusgen L, Purvis N, Regnier M, Lin DC, Rodgers BD, Chamberlain JS. Efficacy and muscle safety assessment of fukutin-related protein gene therapy. Mol Ther Methods Clin Dev 2023; 30:65-80. [PMID: 37361354 PMCID: PMC10285450 DOI: 10.1016/j.omtm.2023.05.022] [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: 11/28/2022] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
Limb-girdle muscular dystrophy type R9 (LGMDR9) is a muscle-wasting disease that begins in the hip and shoulder regions of the body. This disease is caused by mutations in fukutin-related protein (FKRP), a glycosyltransferase critical for maintaining muscle cell integrity. Here we investigated potential gene therapies for LGMDR9 containing an FKRP expression construct with untranslated region (UTR) modifications. Initial studies treated an aged dystrophic mouse model (FKRPP448L) with adeno-associated virus vector serotype 6 (AAV6). Grip strength improved in a dose- and time-dependent manner, injected mice exhibited fewer central nuclei and serum creatine kinase levels were 3- and 5-fold lower compared to those in non-injected FKRPP448L mice. Treatment also partially stabilized the respiratory pattern during exercise and improved treadmill running, partially protecting muscle from exercise-induced damage. Western blotting of C2C12 myotubes using a novel rabbit antibody confirmed heightened translation with the UTR modifications. We further explored the question of FKRP toxicity in wild-type mice using high doses of two additional muscle-tropic capsids: AAV9 and AAVMYO1. No toxic effects were detected with either therapeutic agent. These data further support the feasibility of gene therapy to treat LGMDR9.
Collapse
Affiliation(s)
- Halli Benasutti
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Joseph W. Maricelli
- School of Molecular Biosciences, Washington State University College of Veterinary Medicine, Pullman, WA 99164, USA
- Washington Center for Muscle Biology, Washington State University, Pullman, WA 99164, USA
| | - Jane Seto
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Sen. Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - John Hall
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Christine Halbert
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Sen. Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Jacqueline Wicki
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lydia Heusgen
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Nicholas Purvis
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington School of Medicine, Seattle, WA, USA
| | - David C. Lin
- Department of Integrative Physiology and Neuroscience and the Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, USA
| | - Buel D. Rodgers
- School of Molecular Biosciences, Washington State University College of Veterinary Medicine, Pullman, WA 99164, USA
- Washington Center for Muscle Biology, Washington State University, Pullman, WA 99164, USA
| | - Jeffrey S. Chamberlain
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
- Sen. Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
4
|
Ritter P, Nübler S, Buttgereit A, Smith LR, Mühlberg A, Bauer J, Michael M, Kreiß L, Haug M, Barton E, Friedrich O. Myofibrillar Lattice Remodeling Is a Structural Cytoskeletal Predictor of Diaphragm Muscle Weakness in a Fibrotic mdx ( mdx Cmah-/-) Model. Int J Mol Sci 2022; 23:ijms231810841. [PMID: 36142754 PMCID: PMC9500669 DOI: 10.3390/ijms231810841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative genetic myopathy characterized by complete absence of dystrophin. Although the mdx mouse lacks dystrophin, its phenotype is milder compared to DMD patients. The incorporation of a null mutation in the Cmah gene led to a more DMD-like phenotype (i.e., more fibrosis). Although fibrosis is thought to be the major determinant of ‘structural weakness’, intracellular remodeling of myofibrillar geometry was shown to be a major cellular determinant thereof. To dissect the respective contribution to muscle weakness, we assessed biomechanics and extra- and intracellular architecture of whole muscle and single fibers from extensor digitorum longus (EDL) and diaphragm. Despite increased collagen contents in both muscles, passive stiffness in mdx Cmah−/− diaphragm was similar to wt mice (EDL muscles were twice as stiff). Isometric twitch and tetanic stresses were 50% reduced in mdx Cmah−/− diaphragm (15% in EDL). Myofibrillar architecture was severely compromised in mdx Cmah−/− single fibers of both muscle types, but more pronounced in diaphragm. Our results show that the mdx Cmah−/− genotype reproduces DMD-like fibrosis but is not associated with changes in passive visco-elastic muscle stiffness. Furthermore, detriments in active isometric force are compatible with the pronounced myofibrillar disarray of the dystrophic background.
Collapse
Affiliation(s)
- Paul Ritter
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- Correspondence:
| | - Stefanie Nübler
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Andreas Buttgereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas R. Smith
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95618, USA
| | - Alexander Mühlberg
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Julian Bauer
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Mena Michael
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas Kreiß
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Michael Haug
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Elisabeth Barton
- College of Health & Human Performance, University of Florida, Gainesville, FL 32611, USA
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- School of Medical Sciences, University of New South Wales, Wallace Wurth Building, 18 High Str., Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Assis AD, Chiarotto GB, da Silva NS, Simões GF, Oliveira ALR. Pregabalin synchronizes the regeneration of nerve and muscle fibers optimizing the gait recovery of MDX dystrophic mice. FASEB J 2022; 36:e22511. [PMID: 35998000 DOI: 10.1096/fj.202200411rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder induced by mutations in the dystrophin gene, leading to a degeneration of muscle fibers, triggering retrograde immunomodulatory, and degenerative events in the central nervous system. Thus, neuroprotective drugs such as pregabalin (PGB) can improve motor function by modulating plasticity, together with anti-inflammatory effects. The present work aimed to study the effects of PGB on axonal regeneration after axotomy in dystrophic and non-dystrophic mice. For that, MDX and C57BL/10 mouse strains were subjected to peripheral nerve damage and were treated with PGB (30 mg/kg/day, i.p.) for 28 consecutive days. The treatment was carried out in mice as soon as they completed 5 weeks of life, 1 week before the lesion, corresponding to the peak period of muscle degeneration in the MDX strain. Six-week-old mice were submitted to unilateral sciatic nerve crush and were sacrificed in the 9th week of age. The ipsi and contralateral sciatic nerves were processed for immunohistochemistry and qRT-PCR, evaluating the expression of proteins and gene transcripts related to neuronal and Schwann cell activity. Cranial tibial muscles were dissected for evaluation of neuromuscular junctions using α-bungarotoxin, and the myelinated axons of the sciatic nerve were analyzed by morphometry. The recovery of motor function was monitored throughout the treatment through tests of forced locomotion (rotarod) and spontaneous walking track test (Catwalk system). The results show that treatment with PGB reduced the retrograde cyclic effects of muscle degeneration/regeneration on the nervous system. This fact was confirmed after peripheral nerve injury, showing better adaptation and response of neurons and glia for rapid axonal regeneration, with efficient muscle targeting and regain of function. No side effects of PGB treatment were observed, and the expression of pro-regenerative proteins in neurons and Schwann cells was upregulated. Morphometry of the axons was in line with the preservation of motor endplates, resulting in enhanced performance of dystrophic animals. Overall, the present data indicate that pregabalin is protective and enhances regeneration of the SNP during the development of DMD, improving motor function, which can, in turn, be translated to the clinic.
Collapse
Affiliation(s)
- Alex Dias Assis
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Campinas, Brazil
| | | | | | | | | |
Collapse
|
6
|
Hrach HC, O'Brien S, Steber HS, Newbern J, Rawls A, Mangone M. Transcriptome changes during the initiation and progression of Duchenne muscular dystrophy in Caenorhabditis elegans. Hum Mol Genet 2021; 29:1607-1623. [PMID: 32227114 PMCID: PMC7322572 DOI: 10.1093/hmg/ddaa055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 02/17/2020] [Accepted: 03/23/2020] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked disease characterized by progressive muscle degeneration. The condition is driven by nonsense and missense mutations in the dystrophin gene, leading to instability of the sarcolemma and skeletal muscle necrosis and atrophy. Resulting changes in muscle-specific gene expression that take place in dystrophin's absence remain largely uncharacterized, as they are potentially obscured by the chronic inflammation elicited by muscle damage in humans. Caenorhabditis elegans possess a mild inflammatory response that is not active in the muscle, and lack a satellite cell equivalent. This allows for the characterization of the transcriptome rearrangements affecting disease progression independently of inflammation and regeneration. In effort to better understand these dynamics, we have isolated and sequenced body muscle-specific transcriptomes from C. elegans lacking functional dystrophin at distinct stages of disease progression. We have identified an upregulation of genes involved in mitochondrial function early in disease progression, and an upregulation of genes related to muscle repair in later stages. Our results suggest that in C. elegans, dystrophin may have a signaling role early in development, and its absence may activate compensatory mechanisms that counteract muscle degradation caused by loss of dystrophin. We have also developed a temperature-based screening method for synthetic paralysis that can be used to rapidly identify genetic partners of dystrophin. Our results allow for the comprehensive identification of transcriptome changes that potentially serve as independent drivers of disease progression and may in turn allow for the identification of new therapeutic targets for the treatment of DMD.
Collapse
Affiliation(s)
- Heather C Hrach
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, 427 East Tyler Mall, Tempe, AZ 85287 4501, USA.,Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281, USA
| | - Shannon O'Brien
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281, USA.,Barrett Honors College, Arizona State University, 751 E Lemon Mall, Tempe, AZ 85281, USA
| | - Hannah S Steber
- Barrett Honors College, Arizona State University, 751 E Lemon Mall, Tempe, AZ 85281, USA
| | - Jason Newbern
- School of Life Sciences, 427 East Tyler Mall, Tempe, AZ 85287 4501, USA
| | - Alan Rawls
- School of Life Sciences, 427 East Tyler Mall, Tempe, AZ 85287 4501, USA
| | - Marco Mangone
- Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute at Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85281, USA
| |
Collapse
|
7
|
Age-Dependent Dysregulation of Muscle Vasculature and Blood Flow Recovery after Hindlimb Ischemia in the mdx Model of Duchenne Muscular Dystrophy. Biomedicines 2021; 9:biomedicines9050481. [PMID: 33925757 PMCID: PMC8145677 DOI: 10.3390/biomedicines9050481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by a lack of functional dystrophin, is characterized by progressive muscle degeneration. Interestingly, dystrophin is also expressed in endothelial cells (ECs), and insufficient angiogenesis has already been hypothesized to contribute to DMD pathology, however, its status in mdx mice, a model of DMD, is still not fully clear. Our study aimed to reveal angiogenesis-related alterations in skeletal muscles of mdx mice compared to wild-type (WT) counterparts. By investigating 6- and 12-week-old mice, we sought to verify if those changes are age-dependent. We utilized a broad spectrum of methods ranging from gene expression analysis, flow cytometry, and immunofluorescence imaging to determine the level of angiogenic markers and to assess muscle blood vessel abundance. Finally, we implemented the hindlimb ischemia (HLI) model, more biologically relevant in the context of functional studies evaluating angiogenesis/arteriogenesis processes. We demonstrated that both 6- and 12-week-old dystrophic mice exhibited dysregulation of several angiogenic factors, including decreased vascular endothelial growth factor A (VEGF) in different muscle types. Nonetheless, in younger, 6-week-old mdx animals, neither the abundance of CD31+α-SMA+ double-positive blood vessels nor basal blood flow and its restoration after HLI was affected. In 12-week-old mdx mice, although a higher number of CD31+α-SMA+ double-positive blood vessels and an increased percentage of skeletal muscle ECs were found, the abundance of pericytes was diminished, and blood flow was reduced. Moreover, impeded perfusion recovery after HLI associated with a blunted inflammatory and regenerative response was evident in 12-week-old dystrophic mice. Hence, our results reinforce the hypothesis of age-dependent angiogenic dysfunction in dystrophic mice. In conclusion, we suggest that older mdx mice constitute an appropriate model for preclinical studies evaluating the effectiveness of vascular-based therapies aimed at the restoration of functional angiogenesis to mitigate DMD severity.
Collapse
|
8
|
Banks GB, Chamberlain JS, Odom GL. Microutrophin expression in dystrophic mice displays myofiber type differences in therapeutic effects. PLoS Genet 2020; 16:e1009179. [PMID: 33175853 PMCID: PMC7682874 DOI: 10.1371/journal.pgen.1009179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022] Open
Abstract
Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (μUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-μUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for μUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed μUtrn. As such, we found that μUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike μUtrn, μdystrophin (μDys) expression did not show this fiber type preference. Interestingly, μUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & μUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.
Collapse
MESH Headings
- Animals
- Dependovirus/genetics
- Disease Models, Animal
- Dystrophin/genetics
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Vectors/genetics
- HEK293 Cells
- Humans
- Mice
- Mice, Inbred mdx
- Microscopy, Electron
- Muscle Contraction
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Neuromuscular Junction/pathology
- Neuromuscular Junction/ultrastructure
- Sarcolemma/pathology
- Sarcolemma/ultrastructure
- Utrophin/genetics
- Utrophin/therapeutic use
Collapse
Affiliation(s)
- Glen B. Banks
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
- Department of BioChemistry, University of Washington, Seattle, Washington, United States of America
| | - Guy L. Odom
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
9
|
Lasa-Fernandez H, Mosqueira-Martín L, Alzualde A, Lasa-Elgarresta J, Vallejo-Illarramendi A. A genotyping method combining primer competition PCR with HRM analysis to identify point mutations in Duchenne animal models. Sci Rep 2020; 10:17224. [PMID: 33057138 PMCID: PMC7560699 DOI: 10.1038/s41598-020-74173-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/28/2020] [Indexed: 11/09/2022] Open
Abstract
Dystrophin-null sapje zebrafish is an excellent model for better understanding the pathological mechanisms underlying Duchenne muscular dystrophy, and it has recently arisen as a powerful tool for high-throughput screening of therapeutic candidates for this disease. While dystrophic phenotype in sapje larvae can be easily detected by birefringence, zebrafish genotyping is necessary for drug screening experiments, where the potential rescue of larvae phenotype is the primary outcome. Genotyping is also desirable during colony husbandry since heterozygous progenitors need to be selected. Currently, sapje zebrafish are genotyped through techniques involving sequencing or multi-step PCR, which are often costly, tedious, or require special equipment. Here we report a simple, precise, cost-effective, and versatile PCR genotyping method based on primer competition. Genotypes can be resolved by standard agarose gel electrophoresis and high-resolution melt assay, the latter being especially useful for genotyping a large number of samples. Our approach has shown high sensitivity, specificity, and reproducibility in detecting the A/T point mutation in sapje zebrafish and the C/T mutation in the mdx mouse model of Duchenne. Hence, this method can be applied to other single nucleotide substitutions and may be further optimized to detect small insertions and deletions. Given its robust performance with crude DNA extracts, our strategy may be particularly well-suited for detecting single nucleotide variants in poor-quality samples such as ancient DNA or DNA from formalin-fixed, paraffin-embedded material.
Collapse
Affiliation(s)
- Haizpea Lasa-Fernandez
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Laura Mosqueira-Martín
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | | | - Jaione Lasa-Elgarresta
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Dept. Pediatrics, University of the Basque Country UPV/EHU, Hospital Universitario Donostia, Paseo Dr. Begiristain 105, 20014, San Sebastián, Spain.
- Group of Neuromuscular Diseases, Biodonostia Health Research Institute, Paseo Dr. Begiristain s/n, 20014, San Sebastián, Spain.
- CIBERNED, Instituto de Salud Carlos III, 28031, Madrid, Spain.
| |
Collapse
|
10
|
Barthélémy I, Calmels N, Weiss RB, Tiret L, Vulin A, Wein N, Peccate C, Drougard C, Beroud C, Deburgrave N, Thibaud JL, Escriou C, Punzón I, Garcia L, Kaplan JC, Flanigan KM, Leturcq F, Blot S. X-linked muscular dystrophy in a Labrador Retriever strain: phenotypic and molecular characterisation. Skelet Muscle 2020; 10:23. [PMID: 32767978 PMCID: PMC7412789 DOI: 10.1186/s13395-020-00239-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background Canine models of Duchenne muscular dystrophy (DMD) are a valuable tool to evaluate potential therapies because they faithfully reproduce the human disease. Several cases of dystrophinopathies have been described in canines, but the Golden Retriever muscular dystrophy (GRMD) model remains the most used in preclinical studies. Here, we report a new spontaneous dystrophinopathy in a Labrador Retriever strain, named Labrador Retriever muscular dystrophy (LRMD). Methods A colony of LRMD dogs was established from spontaneous cases. Fourteen LRMD dogs were followed-up and compared to the GRMD standard using several functional tests. The disease causing mutation was studied by several molecular techniques and identified using RNA-sequencing. Results The main clinical features of the GRMD disease were found in LRMD dogs; the functional tests provided data roughly overlapping with those measured in GRMD dogs, with similar inter-individual heterogeneity. The LRMD causal mutation was shown to be a 2.2-Mb inversion disrupting the DMD gene within intron 20 and involving the TMEM47 gene. In skeletal muscle, the Dp71 isoform was ectopically expressed, probably as a consequence of the mutation. We found no evidence of polymorphism in either of the two described modifier genes LTBP4 and Jagged1. No differences were found in Pitpna mRNA expression levels that would explain the inter-individual variability. Conclusions This study provides a full comparative description of a new spontaneous canine model of dystrophinopathy, found to be phenotypically equivalent to the GRMD model. We report a novel large DNA mutation within the DMD gene and provide evidence that LRMD is a relevant model to pinpoint additional DMD modifier genes.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Nadège Calmels
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Laboratoire de Diagnostic Génétique-Institut de Génétique Médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, 1 Place de L'Hôpital, 67091, Strasbourg, France
| | - Robert B Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Adeline Vulin
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France
| | - Nicolas Wein
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Cécile Peccate
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Carole Drougard
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Christophe Beroud
- Aix Marseille Université, INSERM, MMG, Bioinformatics & Genetics, Marseille, France.,APHM, Hôpital Timone Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | - Nathalie Deburgrave
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Jean-Laurent Thibaud
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Catherine Escriou
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Isabel Punzón
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Luis Garcia
- Université de Versailles Saint-Quentin-en-Yvelines, U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France
| | - Jean-Claude Kaplan
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Kevin M Flanigan
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - France Leturcq
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Stéphane Blot
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France.
| |
Collapse
|
11
|
Miyamoto M, Tochinai R, Sekizawa SI, Shiga T, Uchida K, Tsuru Y, Kuwahara M. Cardiac lesions in Duchenne muscular dystrophy model rats with out-of-frame Dmd gene mutation mediated by CRISPR/Cas9 system. J Toxicol Pathol 2020; 33:227-236. [PMID: 33239841 PMCID: PMC7677620 DOI: 10.1293/tox.2020-0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscular disorder caused by
X-chromosomal DMD gene mutations. Recently, a new CRISPR/Cas9-mediated
DMD rat model (cDMDR) was established and is expected to show cardiac lesions similar to
those in humans. We therefore investigated the pathological and pathophysiological
features of the cardiac lesions and their progression in cDMDR. For our cDMDR,
Dmd-mutated rats (W-Dmdem1Kykn) were
obtained. Dmd heterozygous-deficient females and wild-type (WT) males
were mated, and male offspring including WT as controls were used. (1) Hearts were
collected at 3, 5, and 10 months of age, and HE- and Masson’s trichrome-stained specimens
were observed. (2) Electrocardiogram (ECG) recordings were made and analyzed at 3, 5, and
8 months of age. (3) Echocardiography was performed at 9 months of age. In cDMDR rats, (1)
degeneration/necrosis of cardiomyocytes and myocardial fibrosis prominent in the right
ventricular wall and the outer layer of the left ventricular wall were observed. Fibrosis
became more prominent with aging. (2) Lower P wave amplitudes and greater R wave
amplitudes were detected. PR intervals tended to be shorter. QT intervals were longer at 3
months but tended to be shorter at 8 months. Sinus irregularity and premature ventricular
contraction were observed at 8 months. (3) Echocardiography indicated myocardial sclerosis
and a tendency of systolic dysfunction. Pathological and pathophysiological changes
occurred in cDMDR rat hearts and progressed with aging, which is, to some extent, similar
to what occurs in humans. Thus, cDMDR could be a valuable model for studying cardiology of
human DMD.
Collapse
Affiliation(s)
- Mao Miyamoto
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryota Tochinai
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shin-Ich Sekizawa
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takanori Shiga
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kazuyuki Uchida
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoshiharu Tsuru
- Primetech Corp. Life Science Laboratory, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
12
|
Lim KRQ, Nguyen Q, Dzierlega K, Huang Y, Yokota T. CRISPR-Generated Animal Models of Duchenne Muscular Dystrophy. Genes (Basel) 2020; 11:genes11030342. [PMID: 32213923 PMCID: PMC7141101 DOI: 10.3390/genes11030342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive neuromuscular disorder most commonly caused by mutations disrupting the reading frame of the dystrophin (DMD) gene. DMD codes for dystrophin, which is critical for maintaining the integrity of muscle cell membranes. Without dystrophin, muscle cells receive heightened mechanical stress, becoming more susceptible to damage. An active body of research continues to explore therapeutic treatments for DMD as well as to further our understanding of the disease. These efforts rely on having reliable animal models that accurately recapitulate disease presentation in humans. While current animal models of DMD have served this purpose well to some extent, each has its own limitations. To help overcome this, clustered regularly interspaced short palindromic repeat (CRISPR)-based technology has been extremely useful in creating novel animal models for DMD. This review focuses on animal models developed for DMD that have been created using CRISPR, their advantages and disadvantages as well as their applications in the DMD field.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Kasia Dzierlega
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Yiqing Huang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G 2H7, Canada
- Correspondence: ; Tel.: +1-780-492-1102
| |
Collapse
|
13
|
Datson NA, Bijl S, Janson A, Testerink J, van den Eijnde R, Weij R, Puoliväli J, Lehtimäki K, Bragge T, Ahtoniemi T, van Deutekom JC. Using a State-of-the-Art Toolbox to Evaluate Molecular and Functional Readouts of Antisense Oligonucleotide-Induced Exon Skipping in mdx Mice. Nucleic Acid Ther 2020; 30:50-65. [PMID: 31821107 PMCID: PMC7049912 DOI: 10.1089/nat.2019.0824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/06/2019] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe childhood muscle disease primarily caused by the lack of functional dystrophin at the muscle fiber membranes. Multiple therapeutic approaches are currently in (pre)clinical development, aimed at restoring expression of (truncated) dystrophin. Key questions in this phase relate to route of drug administration, dose regimen, and levels of dystrophin required to improve muscle function. A series of studies applying antisense oligonucleotides (AONs) in the mdx mouse model for DMD has been reported over the last two decades, claiming a variable range of exon skipping and increased dystrophin levels correlated to some functional improvement. The aim of this study was to compare the efficacy of subcutaneous (SC) versus intravenous (IV) dosing routes of an mdx-specific AON at both the molecular and functional level, using state-of-the-art quantitative technologies, including digital droplet polymerase chain reaction, capillary Western immunoassay, magnetic resonance imaging, and automated kinematic analysis. The majority of all readouts we quantified, both molecular and functional, showed that IV dosing of the AON had a more pronounced beneficial effect than SC dosing in mdx mice. Last, but not least, the more quantitative molecular and functional data obtained in this study suggest that low levels of dystrophin protein of at least 2.5% of wild type may already have a beneficial effect on muscle leakiness and may improve motor performance of mdx mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Rudie Weij
- BioMarin Nederland BV, Leiden, the Netherlands
| | | | | | - Timo Bragge
- Charles River Discovery Research Services, Kuopio, Finland
| | - Toni Ahtoniemi
- Charles River Discovery Research Services, Kuopio, Finland
| | | |
Collapse
|
14
|
Maleiner B, Tomasch J, Heher P, Spadiut O, Rünzler D, Fuchs C. The Importance of Biophysical and Biochemical Stimuli in Dynamic Skeletal Muscle Models. Front Physiol 2018; 9:1130. [PMID: 30246791 PMCID: PMC6113794 DOI: 10.3389/fphys.2018.01130] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
Classical approaches to engineer skeletal muscle tissue based on current regenerative and surgical procedures still do not meet the desired outcome for patient applications. Besides the evident need to create functional skeletal muscle tissue for the repair of volumetric muscle defects, there is also growing demand for platforms to study muscle-related diseases, such as muscular dystrophies or sarcopenia. Currently, numerous studies exist that have employed a variety of biomaterials, cell types and strategies for maturation of skeletal muscle tissue in 2D and 3D environments. However, researchers are just at the beginning of understanding the impact of different culture settings and their biochemical (growth factors and chemical changes) and biophysical cues (mechanical properties) on myogenesis. With this review we intend to emphasize the need for new in vitro skeletal muscle (disease) models to better recapitulate important structural and functional aspects of muscle development. We highlight the importance of choosing appropriate system components, e.g., cell and biomaterial type, structural and mechanical matrix properties or culture format, and how understanding their interplay will enable researchers to create optimized platforms to investigate myogenesis in healthy and diseased tissue. Thus, we aim to deliver guidelines for experimental designs to allow estimation of the potential influence of the selected skeletal muscle tissue engineering setup on the myogenic outcome prior to their implementation. Moreover, we offer a workflow to facilitate identifying and selecting different analytical tools to demonstrate the successful creation of functional skeletal muscle tissue. Ultimately, a refinement of existing strategies will lead to further progression in understanding important aspects of muscle diseases, muscle aging and muscle regeneration to improve quality of life of patients and enable the establishment of new treatment options.
Collapse
Affiliation(s)
- Babette Maleiner
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Janine Tomasch
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna, Austria.,Trauma Care Consult GmbH, Vienna, Austria
| | - Oliver Spadiut
- Institute of Chemical Engineering, Vienna University of Technology, Vienna, Austria
| | - Dominik Rünzler
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christiane Fuchs
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
15
|
Rodrigues M, Echigoya Y, Fukada SI, Yokota T. Current Translational Research and Murine Models For Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 3:29-48. [PMID: 27854202 PMCID: PMC5271422 DOI: 10.3233/jnd-150113] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscle degeneration. Mutations in the DMD gene result in the absence of dystrophin, a protein required for muscle strength and stability. Currently, there is no cure for DMD. Since murine models are relatively easy to genetically manipulate, cost effective, and easily reproducible due to their short generation time, they have helped to elucidate the pathobiology of dystrophin deficiency and to assess therapies for treating DMD. Recently, several murine models have been developed by our group and others to be more representative of the human DMD mutation types and phenotypes. For instance, mdx mice on a DBA/2 genetic background, developed by Fukada et al., have lower regenerative capacity and exhibit very severe phenotype. Cmah-deficient mdx mice display an accelerated disease onset and severe cardiac phenotype due to differences in glycosylation between humans and mice. Other novel murine models include mdx52, which harbors a deletion mutation in exon 52, a hot spot region in humans, and dystrophin/utrophin double-deficient (dko), which displays a severe dystrophic phenotype due the absence of utrophin, a dystrophin homolog. This paper reviews the pathological manifestations and recent therapeutic developments in murine models of DMD such as standard mdx (C57BL/10), mdx on C57BL/6 background (C57BL/6-mdx), mdx52, dystrophin/utrophin double-deficient (dko), mdxβgeo, Dmd-null, humanized DMD (hDMD), mdx on DBA/2 background (DBA/2-mdx), Cmah-mdx, and mdx/mTRKO murine models.
Collapse
Affiliation(s)
- Merryl Rodrigues
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Yusuke Echigoya
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada.,Muscular Dystrophy Canada Research Chair, Edmonton, Alberta, Canada
| |
Collapse
|
16
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
17
|
Abnormal carbohydrate metabolism in a canine model for muscular dystrophy. J Nutr Sci 2017; 6:e57. [PMID: 29209496 PMCID: PMC5705810 DOI: 10.1017/jns.2017.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/26/2017] [Accepted: 09/28/2017] [Indexed: 11/21/2022] Open
Abstract
The canine golden retriever muscular dystrophy (GRMD) model is the best animal model for studying Duchenne muscular dystrophy in humans. Considering the importance of glucose metabolism in the muscles, the existence of metabolic and endocrine alterations in a wide range of muscular dystrophies, and the pre-existing relationship between blood insulin concentration and muscular atrophy, the present study aimed to evaluate the postprandial glucose and insulin response in GRMD dogs. A total of eighteen golden retriever dogs were randomly distributed into three experimental groups: healthy/control (G1), female GRMD carriers (G2), and male dogs affected by GRMD (G3). Higher plasma resting glucose levels (P = 0·0047) were seen in G2 and G3 compared with G1, as was the case for minimum (P = <0·0001), mean (P = 0·0002) and maximum (P = 0·0359) glucose values for G3 compared with G1. Fructosamine concentrations were in accordance with reference values found in the literature for dogs. Insulin levels were lower in G3 compared with G1 (P = 0·0065); however, there was no evidence of insulin resistance according to the homeostasis model assessment index values obtained. As for the evaluation of postprandial responses, fluctuations of glucose (P = 0·0007) and insulin (P = 0·0149) were observed in G1 and G2, while in G3 the values remained constant. The results allowed us to identify metabolic changes related to carbohydrate metabolism in GRMD dogs, highlighting the importance of adequate food management for these animals.
Collapse
|
18
|
Vohra R, Batra A, Forbes SC, Vandenborne K, Walter GA. Magnetic Resonance Monitoring of Disease Progression in mdx Mice on Different Genetic Backgrounds. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2060-2070. [PMID: 28826559 DOI: 10.1016/j.ajpath.2017.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/04/2017] [Indexed: 12/15/2022]
Abstract
Genetic modifiers alter disease progression in both preclinical models and subjects with Duchenne muscular dystrophy (DMD). Using multiparametric magnetic resonance (MR) techniques, we compared the skeletal and cardiac muscles of two different dystrophic mouse models of DMD, which are on different genetic backgrounds, the C57BL/10ScSn-Dmdmdx (B10-mdx) and D2.B10-Dmdmdx (D2-mdx). The proton transverse relaxation constant (T2) using both MR imaging and spectroscopy revealed significant age-related differences in dystrophic skeletal and cardiac muscles as compared with their age-matched controls. D2-mdx muscles demonstrated an earlier and accelerated decrease in muscle T2 compared with age-matched B10-mdx muscles. Diffusion-weighted MR imaging indicated differences in the underlying muscle structure between the mouse strains. The fractional anisotropy, mean diffusion, and radial diffusion of water varied significantly between the two dystrophic strains. Muscle structural differences were confirmed by histological analyses of the gastrocnemius, revealing a decreased muscle fiber size and increased fibrosis in skeletal muscle fibers of D2-mdx mice compared with B10-mdx and control. Cardiac involvement was also detected in D2-mdx myocardium based on both decreased function and myocardial T2. These data indicate that MR parameters may be used as sensitive biomarkers to detect fibrotic tissue deposition and fiber atrophy in dystrophic strains.
Collapse
Affiliation(s)
- Ravneet Vohra
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
19
|
Li Z, Li Y, Zhang L, Zhang X, Sullivan R, Ai X, Szeto C, Cai A, Liu L, Xiao W, Li Q, Ge S, Chen X. Reduced Myocardial Reserve in Young X-Linked Muscular Dystrophy Mice Diagnosed by Two-Dimensional Strain Analysis Combined with Stress Echocardiography. J Am Soc Echocardiogr 2017; 30:815-827.e9. [PMID: 28511858 DOI: 10.1016/j.echo.2017.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Early, sensitive, and reproducible evaluation of left ventricular function is imperative for the diagnosis of cardiac dysfunction in patients with Duchene muscular dystrophy. The aim of this study was to test the hypothesis that combining two-dimensional strain analysis with catecholamine stress could be a sensitive method for detecting early cardiac dysfunction. METHODS Mdx (C57BL/10ScSn-Dmdmdx/J, a mouse model of DMD) and control (C57BL/10ScSn) mice were studied with conventional M-mode and high-frequency ultrasound-based two-dimensional speckle-tracking echocardiography using long- and short-axis images of the left ventricle at baseline and after intraperitoneal isoprenaline (ISO) administration (2 μg/g body weight). RESULTS Conventional M-mode analysis showed no differences in left ventricular fractional shortening, wall thickness, or internal diameter at diastole between mdx and control mice before the age of 6 months. ISO increased left ventricular ejection fraction and fractional shortening to the same extent in mdx and control mice at young ages (3, 4, and 5 months). No differences in basal peak systolic strain (PSS) but increased SDs of times to PSS between young mdx and control mice were found. After ISO, PSS and percentile changes of PSS were significantly diminished in mdx mice compared with control mice at young ages. ISO increased the normalized maximum difference of times to PSS in young mdx mice but not in young control mice, suggesting that ISO reduces cardiac contractile synchrony in young mdx mice. CONCLUSIONS This study suggests that catecholamine stress coupled with two-dimensional strain analysis is a feasible and sensitive approach for detecting early onset of cardiac dysfunction, which is instrumental for early diagnosis of cardiac dysfunction and early treatment.
Collapse
Affiliation(s)
- Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, Shenzhen, China; Drexel University College of Medicine, Philadelphia, Pennsylvania; Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ying Li
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; The General Hospital of The PLA Rocket Force, Beijing, China
| | - Li Zhang
- Drexel University College of Medicine, Philadelphia, Pennsylvania; Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoying Zhang
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Rebecca Sullivan
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Xiaojie Ai
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; College of Biological Sciences, Shanghai Jiaotong University, Shanghai, China
| | - Christopher Szeto
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Angela Cai
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Longjian Liu
- Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Weidong Xiao
- Department of Microbiology and Immunology and Sol Sherry Thrombosis Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Quanshui Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Shuping Ge
- Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Xiongwen Chen
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Huang SC, Zhou A, Nguyen DT, Zhang HS, Benz EJ. Protein 4.1R Influences Myogenin Protein Stability and Skeletal Muscle Differentiation. J Biol Chem 2016; 291:25591-25607. [PMID: 27780863 DOI: 10.1074/jbc.m116.761296] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Indexed: 01/28/2023] Open
Abstract
Protein 4.1R (4.1R) isoforms are expressed in both cardiac and skeletal muscle. 4.1R is a component of the contractile apparatus. It is also associated with dystrophin at the sarcolemma in skeletal myofibers. However, the expression and function of 4.1R during myogenesis have not been characterized. We now report that 4.1R expression increases during C2C12 myoblast differentiation into myotubes. Depletion of 4.1R impairs skeletal muscle differentiation and is accompanied by a decrease in the levels of myosin heavy and light chains and caveolin-3. Furthermore, the expression of myogenin at the protein, but not mRNA, level is drastically decreased in 4.1R knockdown myocytes. Similar results were obtained using MyoD-induced differentiation of 4.1R-/- mouse embryonic fibroblast cells. von Hippel-Lindau (VHL) protein is known to destabilize myogenin via the ubiquitin-proteasome pathway. We show that 4.1R associates with VHL and, when overexpressed, reverses myogenin ubiquitination and stability. This suggests that 4.1R may influence myogenesis by preventing VHL-mediated myogenin degradation. Together, our results define a novel biological function for 4.1R in muscle differentiation and provide a molecular mechanism by which 4.1R promotes myogenic differentiation.
Collapse
Affiliation(s)
- Shu-Ching Huang
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, .,the Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115.,the Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Anyu Zhou
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Dan T Nguyen
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Henry S Zhang
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | - Edward J Benz
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115.,the Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115.,the Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, and.,the Dana-Farber/Harvard Cancer Center, Boston, Massachusetts 02115
| |
Collapse
|
21
|
Yu HH, Zhao H, Qing YB, Pan WR, Jia BY, Zhao HY, Huang XX, Wei HJ. Porcine Zygote Injection with Cas9/sgRNA Results in DMD-Modified Pig with Muscle Dystrophy. Int J Mol Sci 2016; 17:E1668. [PMID: 27735844 PMCID: PMC5085701 DOI: 10.3390/ijms17101668] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 12/16/2022] Open
Abstract
Dystrophinopathy, including Duchenne muscle dystrophy (DMD) and Becker muscle dystrophy (BMD) is an incurable X-linked hereditary muscle dystrophy caused by a mutation in the DMD gene in coding dystrophin. Advances in further understanding DMD/BMD for therapy are expected. Studies on mdx mice and dogs with muscle dystrophy provide limited insight into DMD disease mechanisms and therapeutic testing because of the different pathological manifestations. Miniature pigs share similar physiology and anatomy with humans and are thus an excellent animal model of human disease. Here, we successfully achieved precise DMD targeting in Chinese Diannan miniature pigs by co-injecting zygotes with Cas9 mRNA and sgRNA targeting DMD. Two piglets were obtained after embryo transfer, one of piglets was identified as DMD-modified individual via traditional cloning, sequencing and T7EN1 cleavage assay. An examination of targeting rates in the DMD-modified piglet revealed that sgRNA:Cas9-mediated on-target mosaic mutations were 70% and 60% of dystrophin alleles in skeletal and smooth muscle, respectively. Meanwhile, no detectable off-target mutations were found, highlighting the high specificity of genetic modification using CRISPR/Cas9. The DMD-modified piglet exhibited degenerative and disordered phenotypes in skeletal and cardiac muscle, and declining thickness of smooth muscle in the stomach and intestine. In conclusion, we successfully generated myopathy animal model by modifying the DMD via CRISPR/Cas9 system in a miniature pig.
Collapse
MESH Headings
- Alleles
- Animals
- Base Sequence
- CRISPR-Cas Systems/genetics
- Disease Models, Animal
- Dystrophin/genetics
- Dystrophin/metabolism
- Embryo Transfer
- Genotype
- Immunohistochemistry
- Microscopy, Fluorescence
- Muscle, Skeletal/metabolism
- Muscle, Smooth/metabolism
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mutation
- Phenotype
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- RNA, Messenger/metabolism
- Sequence Analysis, DNA
- Swine
- Swine, Miniature
- Zygote/metabolism
Collapse
Affiliation(s)
- Hong-Hao Yu
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Rd., Pudong New Area, Shanghai 201210, China.
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
- Research Center of Life Science, Yulin University, Yulin 719000, China.
| | - Heng Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Yu-Bo Qing
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Wei-Rong Pan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Bao-Yu Jia
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Hong-Ye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
| | - Xing-Xu Huang
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Rd., Pudong New Area, Shanghai 201210, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming 650201, China.
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
22
|
Bettadapur A, Suh GC, Geisse NA, Wang ER, Hua C, Huber HA, Viscio AA, Kim JY, Strickland JB, McCain ML. Prolonged Culture of Aligned Skeletal Myotubes on Micromolded Gelatin Hydrogels. Sci Rep 2016; 6:28855. [PMID: 27350122 PMCID: PMC4924097 DOI: 10.1038/srep28855] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/10/2016] [Indexed: 12/19/2022] Open
Abstract
In vitro models of skeletal muscle are critically needed to elucidate disease mechanisms, identify therapeutic targets, and test drugs pre-clinically. However, culturing skeletal muscle has been challenging due to myotube delamination from synthetic culture substrates approximately one week after initiating differentiation from myoblasts. In this study, we successfully maintained aligned skeletal myotubes differentiated from C2C12 mouse skeletal myoblasts for three weeks by utilizing micromolded (μmolded) gelatin hydrogels as culture substrates, which we thoroughly characterized using atomic force microscopy (AFM). Compared to polydimethylsiloxane (PDMS) microcontact printed (μprinted) with fibronectin (FN), cell adhesion on gelatin hydrogel constructs was significantly higher one week and three weeks after initiating differentiation. Delamination from FN-μprinted PDMS precluded robust detection of myotubes. Compared to a softer blend of PDMS μprinted with FN, myogenic index, myotube width, and myotube length on μmolded gelatin hydrogels was similar one week after initiating differentiation. However, three weeks after initiating differentiation, these parameters were significantly higher on μmolded gelatin hydrogels compared to FN-μprinted soft PDMS constructs. Similar results were observed on isotropic versions of each substrate, suggesting that these findings are independent of substrate patterning. Our platform enables novel studies into skeletal muscle development and disease and chronic drug testing in vitro.
Collapse
Affiliation(s)
- Archana Bettadapur
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Gio C Suh
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | | | - Evelyn R Wang
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.,Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Clara Hua
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Holly A Huber
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Alyssa A Viscio
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Joon Young Kim
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Julie B Strickland
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
23
|
Abstract
Duchenne muscular dystrophy is a progressive, fatal, X-linked disease caused by a failure to accumulate the cytoskeletal protein dystrophin. This disease has been studied using a variety of animal models including fish, mice, rats, and dogs. While these models have contributed substantially to our mechanistic understanding of the disease and disease progression, limitations inherent to each model have slowed the clinical advancement of therapies, which necessitates the development of novel large-animal models. Several porcine dystrophin-deficient models have been identified, although disease severity may be so severe as to limit their potential contributions to the field. We have recently identified and completed the initial characterization of a natural porcine model of dystrophin insufficiency. Muscles from these animals display characteristic focal necrosis concomitant with decreased abundance and localization of dystrophin-glycoprotein complex components. These pigs recapitulate many of the cardinal features of muscular dystrophy, have elevated serum creatine kinase activity, and preliminarily appear to display altered locomotion. They also suffer from sudden death preceded by EKG abnormalities. Pig dystrophinopathy models could allow refinement of dosing strategies in human-sized animals in preparation for clinical trials. From an animal handling perspective, these pigs can generally be treated normally, with the understanding that acute stress can lead to sudden death. In summary, the ability to create genetically modified pig models and the serendipitous discovery of genetic disease in the swine industry has resulted in the emergence of new animal tools to facilitate the critical objective of improving the quality and length of life for boys afflicted with such a devastating disease.
Collapse
Affiliation(s)
- Joshua T Selsby
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Jason W Ross
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Dan Nonneman
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| | - Katrin Hollinger
- Joshua T. Selsby, PhD, and Jason W. Ross, PhD are associate professors of Animal Science at Iowa State University, Ames, IA 50011. Dan Nonneman, PhD, is a research molecular biologist at the USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933. Katrin Hollinger, PhD, was a graduate student in Genetics at Iowa State University, Ames, IA 50011
| |
Collapse
|
24
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
25
|
Cardiac Niche Influences the Direct Reprogramming of Canine Fibroblasts into Cardiomyocyte-Like Cells. Stem Cells Int 2015; 2016:4969430. [PMID: 26681949 PMCID: PMC4670879 DOI: 10.1155/2016/4969430] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/16/2015] [Indexed: 12/23/2022] Open
Abstract
The Duchenne and Becker muscular dystrophies are caused by mutation of dystrophin gene and primarily affect skeletal and cardiac muscles. Cardiac involvement in dystrophic GRMD dogs has been demonstrated by electrocardiographic studies with the onset of a progressive cardiomyopathy similar to the cardiac disease in DMD patients. In this respect, GRMD is a useful model to explore cardiac and skeletal muscle pathogenesis and for developing new therapeutic protocols. Here we describe a protocol to convert GRMD canine fibroblasts isolated from heart and skin into induced cardiac-like myocytes (ciCLMs). We used a mix of transcription factors (GATA4, HAND2, TBX5, and MEF2C), known to be able to differentiate mouse and human somatic cells into ciCLMs. Exogenous gene expression was obtained using four lentiviral vectors carrying transcription factor genes and different resistance genes. Our data demonstrate a direct switch from fibroblast into ciCLMs with no activation of early cardiac genes. ciCLMs were unable to contract spontaneously, suggesting, differently from mouse and human cells, an incomplete differentiation process. However, when transplanted in neonatal hearts of SCID/Beige mice, ciCLMs participate in cardiac myogenesis.
Collapse
|
26
|
Tamayo T, Eno E, Madrigal C, Heydemann A, García K, García J. Functional in situ assessment of muscle contraction in wild-type and mdx mice. Muscle Nerve 2015; 53:260-8. [PMID: 26012778 DOI: 10.1002/mus.24714] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/13/2015] [Accepted: 05/20/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Reports of muscle testing are frequently limited to maximal force alone. The experiments reported here show that force generation and relaxation rates can be obtained from the same experiments and provide a more complete functional characterization. METHODS Partial in situ testing was performed on the tibialis anterior of young wild-type (WT) mice, young mdx mice, and old mdx mice. Force, force generation rate, and relaxation rates were measured during a fatigue test, 2 frequency-force tests, and a passive tension test. RESULTS We measured increased force but decreased force generation rate in WT compared with mdx muscles, and increased force but decreased relaxation rate of old compared with young mdx muscles. Young mdx muscles were the most sensitive to increases in passive tension. CONCLUSIONS These measurements offer an improved understanding of muscle capability and are readily acquired by further analysis of the same tests used to obtain force measurements.
Collapse
Affiliation(s)
- Tammy Tamayo
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| | - Eben Eno
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| | - Carlos Madrigal
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| | - Ahlke Heydemann
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| | - Kelly García
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| | - Jesús García
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, Illinois, 60612, USA
| |
Collapse
|
27
|
Biochemical and Functional Comparisons of mdx and Sgcg(-/-) Muscular Dystrophy Mouse Models. BIOMED RESEARCH INTERNATIONAL 2015; 2015:131436. [PMID: 26064876 PMCID: PMC4433636 DOI: 10.1155/2015/131436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 12/31/2022]
Abstract
Mouse models have provided an essential platform to investigate facets of human diseases, from etiology, diagnosis, and prognosis, to potential treatments. Muscular dystrophy (MD) is the most common human genetic disease occurring in approximately 1 in 2500 births. The mdx mouse, which is dystrophin-deficient, has long been used to model this disease. However, this mouse strain displays a rather mild disease course compared to human patients. The mdx mice have been bred to additional genetically engineered mice to worsen the disease. Alternatively, other genes which cause human MD have been genetically disrupted in mice. We are now comparing disease progression from one of these alternative gene disruptions, the γ-sarcoglycan null mouse Sgcg−/− on the DBA2/J background, to the mdx mouse line. This paper aims to assess the time-course severity of the disease in the mouse models and determine which is best for MD research. The Sgcg−/− mice have a more severe phenotype than the mdx mice. Muscle function was assessed by plethysmography and echocardiography. Histologically the Sgcg−/− mice displayed increased fibrosis and variable fiber size. By quantitative Evan's blue dye uptake and hydroxyproline content two key disease determinants, membrane permeability and fibrosis respectively, were also proven worse in the Sgcg−/− mice.
Collapse
|
28
|
Vieira NM, Guo LT, Estrela E, Kunkel LM, Zatz M, Shelton GD. Muscular dystrophy in a family of Labrador Retrievers with no muscle dystrophin and a mild phenotype. Neuromuscul Disord 2015; 25:363-70. [DOI: 10.1016/j.nmd.2015.02.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 11/28/2022]
|
29
|
Chen Y, Zheng Y, Kang Y, Yang W, Niu Y, Guo X, Tu Z, Si C, Wang H, Xing R, Pu X, Yang SH, Li S, Ji W, Li XJ. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum Mol Genet 2015; 24:3764-74. [PMID: 25859012 DOI: 10.1093/hmg/ddv120] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022] Open
Abstract
CRISPR/Cas9 has been used to genetically modify genomes in a variety of species, including non-human primates. Unfortunately, this new technology does cause mosaic mutations, and we do not yet know whether such mutations can functionally disrupt the targeted gene or cause the pathology seen in human disease. Addressing these issues is necessary if we are to generate large animal models of human diseases using CRISPR/Cas9. Here we used CRISPR/Cas9 to target the monkey dystrophin gene to create mutations that lead to Duchenne muscular dystrophy (DMD), a recessive X-linked form of muscular dystrophy. Examination of the relative targeting rate revealed that Crispr/Cas9 targeting could lead to mosaic mutations in up to 87% of the dystrophin alleles in monkey muscle. Moreover, CRISPR/Cas9 induced mutations in both male and female monkeys, with the markedly depleted dystrophin and muscle degeneration seen in early DMD. Our findings indicate that CRISPR/Cas9 can efficiently generate monkey models of human diseases, regardless of inheritance patterns. The presence of degenerated muscle cells in newborn Cas9-targeted monkeys suggests that therapeutic interventions at the early disease stage may be effective at alleviating the myopathy.
Collapse
Affiliation(s)
- Yongchang Chen
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Yinghui Zheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China
| | - Yu Kang
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Weili Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China
| | - Yuyu Niu
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Xiangyu Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China
| | - Zhuchi Tu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China
| | - Chenyang Si
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Hong Wang
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Ruxiao Xing
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China
| | - Xiuqiong Pu
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Shang-Hsun Yang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China, Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China and
| | - Xiao-Jiang Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 10010, China, Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA,
| |
Collapse
|
30
|
Duan D. Duchenne muscular dystrophy gene therapy in the canine model. HUM GENE THER CL DEV 2015; 26:57-69. [PMID: 25710459 PMCID: PMC4442571 DOI: 10.1089/humc.2015.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked lethal muscle disease caused by dystrophin deficiency. Gene therapy has significantly improved the outcome of dystrophin-deficient mice. Yet, clinical translation has not resulted in the expected benefits in human patients. This translational gap is largely because of the insufficient modeling of DMD in mice. Specifically, mice lacking dystrophin show minimum dystrophic symptoms, and they do not respond to the gene therapy vector in the same way as human patients do. Further, the size of a mouse is hundredfolds smaller than a boy, making it impossible to scale-up gene therapy in a mouse model. None of these limitations exist in the canine DMD (cDMD) model. For this reason, cDMD dogs have been considered a highly valuable platform to test experimental DMD gene therapy. Over the last three decades, a variety of gene therapy approaches have been evaluated in cDMD dogs using a number of nonviral and viral vectors. These studies have provided critical insight for the development of an effective gene therapy protocol in human patients. This review discusses the history, current status, and future directions of the DMD gene therapy in the canine model.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology, Department of Neurology School of Medicine, University of Missouri , Columbia, MO 65212
| |
Collapse
|
31
|
Wasala NB, Zhang K, Wasala LP, Hakim CH, Duan D. The FVB Background Does Not Dramatically Alter the Dystrophic Phenotype of Mdx Mice. PLOS CURRENTS 2015; 7. [PMID: 25737807 PMCID: PMC4339318 DOI: 10.1371/currents.md.28266819ca0ec5fefcac767ea9a3461c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The mdx mouse is the most frequently used animal model for Duchenne muscular dystrophy (DMD), a fatal muscle disease caused by the loss of dystrophin. Mdx mice are naturally occurring dystrophin-null mice on the C57BL/10 (BL10) background. We crossed black mdx to the white FVB background and generated mdx/FVB mice. Compared to that of age- and sex-matched FVB mice, mdx/FVB mice showed characteristic limb muscle pathology similar to that of original mdx mice. Further, the forelimb grip strength and limb muscle (tibialis anterior and extensor digitorum longus) specific force of mdx/FVB mice were significantly lower than that of wild type FVB mice. Consistent with what has been reported in original mdx mice, mdx/FVB mice also showed increased susceptibility to eccentric contraction-induced force loss and elevated serum creatine kinase. Our results suggest that the FVB background does not dramatically alter the dystrophic phenotype of mdx mice.
Collapse
Affiliation(s)
- Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Lakmini P Wasala
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
32
|
Yang X. Stem cell transplantation for treating Duchenne muscular dystrophy: A Web of Science-based literature analysis. Neural Regen Res 2015; 7:1744-51. [PMID: 25624797 PMCID: PMC4302457 DOI: 10.3969/j.issn.1673-5374.2012.22.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE: To identify global research trends in stem cell transplantation for treating Duchenne muscular dystrophy using a bibliometric analysis of Web of Science. DATA RETRIEVAL: We performed a bibliometric analysis of studies on stem cell transplantation for treating Duchenne muscular dystrophy from 2002 to 2011 retrieved from Web of Science. SELECTION CRITERIA: Inclusion criteria: (a) peer-reviewed published articles on stem cell transplantation for treating Duchenne muscular dystrophy indexed in Web of Science; (b) original research articles, reviews, meeting abstracts, proceedings papers, book chapters, editorial material, and news items; and (c) publication between 2002 and 2011. Exclusion criteria: (a) articles that required manual searching or telephone access; (b) documents that were not published in the public domain; and (c) corrected papers. MAIN OUTCOME MEASURES: (1) Annual publication output; (2) distribution according to subject areas; (3) distribution according to journals; (4) distribution according to country; (5) distribution according to institution; (6) distribution according to institution in China; (7) distribution according to institution that cooperated with Chinese institutions; (8) top-cited articles from 2002 to 2006; (9) top-cited articles from 2007 to 2011. RESULTS: A total of 318 publications on stem cell transplantation for treating Duchenne muscular dystrophy were retrieved from Web of Science from 2002 to 2011, of which almost half derived from American authors and institutes. The number of publications has gradually increased over the past 10 years. Most papers appeared in journals with a focus on gene and molecular research, such as Molecular Therapy, Neuromuscular Disorders, and PLoS One. The 10 most-cited papers from 2002 to 2006 were mostly about different kinds of stem cell transplantation for muscle regeneration, while the 10 most-cited papers from 2007 to 2011 were mostly about new techniques of stem cell transplantation for treating Duchenne muscular dystrophy. CONCLUSION: The publications on stem cell transplantation for treating Duchenne muscular dystrophy were relatively few. It also needs more research to confirm that stem cell therapy is a reliable treatment for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Cell Therapy Center, Chinese PLA 463 Hospital, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
33
|
Liu N, Bassel-Duby R. Regulation of skeletal muscle development and disease by microRNAs. Results Probl Cell Differ 2015; 56:165-90. [PMID: 25344671 DOI: 10.1007/978-3-662-44608-9_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The identification of microRNAs (miRNA) in vertebrates has uncovered new mechanisms regulating skeletal muscle development and disease. miRNAs are inhibitors and act by silencing specific mRNAs or by repressing protein translation. In many cases, miRNAs are involved in physiological or pathological stress, suggesting they function to exacerbate or protect the organism during stress or disease. Although many skeletal muscle diseases differ in clinical and pathological manifestations, they all have a common feature of dysregulation of miRNA expression. In particular, analysis of miRNA expression patterns in skeletal muscle diseases reveals miRNA signatures, showing many miRNAs are dysregulated during disease. Emerging identification of miRNA targets and involvement in genetic regulatory networks serve to reveal new regulatory pathways in skeletal muscle biology. This chapter features the findings pertaining to skeletal muscle miRNAs in skeletal muscle development and disease and highlights therapeutic applications of miRNA-based technology in diagnosis and treatment of skeletal muscle myopathies.
Collapse
Affiliation(s)
- Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA,
| | | |
Collapse
|
34
|
The miR-206/133b cluster is dispensable for development, survival and regeneration of skeletal muscle. Skelet Muscle 2014; 4:23. [PMID: 25530839 PMCID: PMC4272821 DOI: 10.1186/s13395-014-0023-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/24/2014] [Indexed: 01/16/2023] Open
Abstract
Background Three different gene clusters code for the muscle-specific miRNAs miR-206, miR-1 and miR-133a/b. The two miR-1/133a clusters generate identical mature miR-1 and miR-133a miRNAs in heart and skeletal muscle, while the cognate miR-206/133b cluster is exclusively expressed in skeletal muscle. Since sequences of the miRNAs miR-133a and miR-133b are almost identical, it seems likely that they share potential targets. Similarly, miR-1 and miR-206 are structurally related and contain identical seed sequences important for miRNA-target recognition. In the past, different functions of these miRNAs were suggested for development, function and regeneration of skeletal muscle using different in vivo and in vitro models; however, mutants lacking the complete miR-206/133b cluster, which generates a single pri-miRNA constituting a functional unit, have not been analyzed. Methods We generated miR-206/133b knock-out mice and analyzed these mice morphologically; at the transcriptome and proteome level to elucidate the contribution of this miRNA cluster for skeletal muscle development, differentiation, regeneration in vivo; and by systematic analysis. In addition, we studied the consequences of a genetic loss of miR-206/133b for expression of Pax7 and satellite cell differentiation in vitro. Results Deletion of the miR-206/133b cluster did not reveal any obvious essential function of the miRNA-cluster for development and differentiation of skeletal muscle. Careful examination of skeletal muscles of miR-206/133b mutants revealed no structural alterations or molecular changes at the transcriptome and proteome level. In contrast to previous studies, deletion of the miR-206/133b cluster did not impair regeneration of skeletal muscle in mdx mice. Likewise, differentiation of miR-206/133b deficient satellite cells in vitro was unaffected and no change in Pax7 protein concentration was apparent. Conclusions We conclude that the miR-206/133b cluster is dispensable for development, function and regeneration of skeletal muscle, probably due to overlapping functions of the related miR-1/133a clusters, which are strongly expressed in skeletal muscle. We reason that the miR-206/133b cluster alone is not an essential regulator of skeletal muscle regeneration, although more subtle functions might exist that are not apparent under laboratory conditions.
Collapse
|
35
|
Brolio MP, Cima DS, Miglino MA, Ambrósio CE. Histological comparison of the smooth uterine muscle of healthy golden retriever bitches, carriers of the progressive muscular dystrophy (GRMD) gene, and GRMD-affected bitches. Anim Reprod Sci 2014; 150:56-61. [PMID: 25200710 DOI: 10.1016/j.anireprosci.2014.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 07/25/2014] [Accepted: 08/15/2014] [Indexed: 10/24/2022]
Abstract
There is evidence to suggest that weakness of the pelvic and/or uterine musculature may negatively affect the obstetric performance of women who carry the gene for Duchenne muscular dystrophy (DMD). The golden retriever dog is the ideal animal model for preclinical studies of progressive muscular dystrophy, and this model is referred to as "golden retriever muscular dystrophy (GRMD)". This study evaluated and compared the histopathological aspects of the uterine muscle of eleven dogs: health, n=4; carriers of GRMD gene, n=5; and affected females, n=2. The obtained results showed that the uterine muscle of healthy dogs was exclusively composed of type III collagen, while a predominance of type I collagen and small amounts of type III were observed in the uterine muscle of the carriers. The myometrium of the affected bitches showed small quantities of both collagen types. The differences noted in the three evaluated groups suggest that female carrier and those individuals affected by muscular dystrophy had collagen alteration and muscle fiber commitment in the uterine muscle, a deficiency which could directly influence the composition and function of this tissue. In addition, this information is highly relevant to the reproductive management of these animals. This data open important venues for translate reproductive protocols for women, who carry the dystrophin gene.
Collapse
Affiliation(s)
- M P Brolio
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science of the University of São Paulo - FMVZ-USP, Orlando Marques de Paiva, 87 street, Sao Paulo, SP 05508-270, Brazil.
| | - D S Cima
- Paulista University - UNIP, Tenente Júlio Prado Neves, 965 street, São Paulo, SP 02370-000, Brazil.
| | - M A Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science of the University of São Paulo - FMVZ-USP, Orlando Marques de Paiva, 87 street, Sao Paulo, SP 05508-270, Brazil.
| | - C E Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering of the University of São Paulo - FZEA-USP, Duque de Caxias Norte, 225, Pirassunuga, SP 13635-900, Brazil.
| |
Collapse
|
36
|
Le Guiner C, Montus M, Servais L, Cherel Y, Francois V, Thibaud JL, Wary C, Matot B, Larcher T, Guigand L, Dutilleul M, Domenger C, Allais M, Beuvin M, Moraux A, Le Duff J, Devaux M, Jaulin N, Guilbaud M, Latournerie V, Veron P, Boutin S, Leborgne C, Desgue D, Deschamps JY, Moullec S, Fromes Y, Vulin A, Smith RH, Laroudie N, Barnay-Toutain F, Rivière C, Bucher S, Le TH, Delaunay N, Gasmi M, Kotin RM, Bonne G, Adjali O, Masurier C, Hogrel JY, Carlier P, Moullier P, Voit T. Forelimb treatment in a large cohort of dystrophic dogs supports delivery of a recombinant AAV for exon skipping in Duchenne patients. Mol Ther 2014; 22:1923-35. [PMID: 25200009 PMCID: PMC4429735 DOI: 10.1038/mt.2014.151] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/14/2014] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by mutations in the dystrophin gene, without curative treatment yet available. Our study provides, for the first time, the overall safety profile and therapeutic dose of a recombinant adeno-associated virus vector, serotype 8 (rAAV8) carrying a modified U7snRNA sequence promoting exon skipping to restore a functional in-frame dystrophin transcript, and injected by locoregional transvenous perfusion of the forelimb. Eighteen Golden Retriever Muscular Dystrophy (GRMD) dogs were exposed to increasing doses of GMP-manufactured vector. Treatment was well tolerated in all, and no acute nor delayed adverse effect, including systemic and immune toxicity was detected. There was a dose relationship for the amount of exon skipping with up to 80% of myofibers expressing dystrophin at the highest dose. Similarly, histological, nuclear magnetic resonance pathological indices and strength improvement responded in a dose-dependent manner. The systematic comparison of effects using different independent methods, allowed to define a minimum threshold of dystrophin expressing fibers (>33% for structural measures and >40% for strength) under which there was no clear-cut therapeutic effect. Altogether, these results support the concept of a phase 1/2 trial of locoregional delivery into upper limbs of nonambulatory DMD patients.
Collapse
Affiliation(s)
- Caroline Le Guiner
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
- Généthon, Evry, France
| | | | - Laurent Servais
- Institut de Myologie, Service of Clinical Trials and Databases, Paris, France
| | - Yan Cherel
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Virginie Francois
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Jean-Laurent Thibaud
- Institut de Myologie, Laboratoire RMN, AIM & CEA, Paris, France
- UPR de Neurobiologie, Ecole Nationale Vétérinaire d'Alfort, Maisons Alfort, France
| | - Claire Wary
- Institut de Myologie, Laboratoire RMN, AIM & CEA, Paris, France
| | - Béatrice Matot
- Institut de Myologie, Laboratoire RMN, AIM & CEA, Paris, France
| | - Thibaut Larcher
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Lydie Guigand
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Maeva Dutilleul
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Claire Domenger
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Marine Allais
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Maud Beuvin
- Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Université Pierre and Marie Curie Paris 6 UPMC-INSERM UMR 974, CNRS FRE 3617, Paris, France
| | - Amélie Moraux
- Institut de Myologie, Neuromuscular Physiology and Evaluation Laboratory, Paris, France
| | - Johanne Le Duff
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Marie Devaux
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Nicolas Jaulin
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Mickaël Guilbaud
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | | | | | | | | | | | - Jack-Yves Deschamps
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
- Atlantic Gene Therapies, Centre de Boisbonne, ONIRIS, Nantes, France
| | - Sophie Moullec
- Atlantic Gene Therapies, Centre de Boisbonne, ONIRIS, Nantes, France
| | - Yves Fromes
- Atlantic Gene Therapies, Centre de Boisbonne, ONIRIS, Nantes, France
| | - Adeline Vulin
- Research Institute, Center for Gene Therapy, Nationwide Childrens Hospital, Columbus, Ohio, USA
| | - Richard H Smith
- Laboratory of Molecular Virology and Gene Therapy, National Heart Lung and Blood Institute, National Institute of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | - Robert M Kotin
- Laboratory of Molecular Virology and Gene Therapy, National Heart Lung and Blood Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Gisèle Bonne
- Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Université Pierre and Marie Curie Paris 6 UPMC-INSERM UMR 974, CNRS FRE 3617, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, U.F. Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Paris, France
| | - Oumeya Adjali
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | | | - Jean-Yves Hogrel
- Institut de Myologie, Neuromuscular Physiology and Evaluation Laboratory, Paris, France
| | - Pierre Carlier
- Institut de Myologie, Laboratoire RMN, AIM & CEA, Paris, France
| | - Philippe Moullier
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
- Généthon, Evry, France
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| | - Thomas Voit
- Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Université Pierre and Marie Curie Paris 6 UPMC-INSERM UMR 974, CNRS FRE 3617, Paris, France
| |
Collapse
|
37
|
Jeanson-Leh L, Lameth J, Krimi S, Buisset J, Amor F, Le Guiner C, Barthélémy I, Servais L, Blot S, Voit T, Israeli D. Serum Profiling Identifies Novel Muscle miRNA and Cardiomyopathy-Related miRNA Biomarkers in Golden Retriever Muscular Dystrophy Dogs and Duchenne Muscular Dystrophy Patients. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2885-98. [DOI: 10.1016/j.ajpath.2014.07.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/01/2014] [Accepted: 07/08/2014] [Indexed: 10/24/2022]
|
38
|
Barthélémy I, Pinto-Mariz F, Yada E, Desquilbet L, Savino W, Silva-Barbosa SD, Faussat AM, Mouly V, Voit T, Blot S, Butler-Browne G. Predictive markers of clinical outcome in the GRMD dog model of Duchenne muscular dystrophy. Dis Model Mech 2014; 7:1253-61. [PMID: 25261568 PMCID: PMC4213729 DOI: 10.1242/dmm.016014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In the translational process of developing innovative therapies for DMD (Duchenne muscular dystrophy), the last preclinical validation step is often carried out in the most relevant animal model of this human disease, namely the GRMD (Golden Retriever muscular dystrophy) dog. The disease in GRMD dogs mimics human DMD in many aspects, including the inter-individual heterogeneity. This last point can be seen as a drawback for an animal model but is inherently related to the disease in GRMD dogs closely resembling that of individuals with DMD. In order to improve the management of this inter-individual heterogeneity, we have screened a combination of biomarkers in sixty-one 2-month-old GRMD dogs at the onset of the disease and a posteriori we addressed their predictive value on the severity of the disease. Three non-invasive biomarkers obtained at early stages of the disease were found to be highly predictive for the loss of ambulation before 6 months of age. An elevation in the number of circulating CD4+CD49dhi T cells and a decreased stride frequency resulting in a reduced spontaneous speed were found to be strongly associated with the severe clinical form of the disease. These factors can be used as predictive tests to screen dogs to separate them into groups with slow or fast disease progression before their inclusion into a therapeutic preclinical trial, and therefore improve the reliability and translational value of the trials carried out on this invaluable large animal model. These same biomarkers have also been described to be predictive for the time to loss of ambulation in boys with DMD, strengthening the relevance of GRMD dogs as preclinical models of this devastating muscle disease.
Collapse
Affiliation(s)
- Inès Barthélémy
- Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UPR de Neurobiologie, 94704 Maisons-Alfort, France
| | - Fernanda Pinto-Mariz
- Université Pierre et Marie Curie-Paris 06 UM76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France. Institute of Pediatrics/Federal University of Rio de Janeiro (IPPMG/UFRJ), Rio de Janeiro 21941-912, Brazil. Laboratory of Thymus Research, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Erica Yada
- Université Pierre et Marie Curie-Paris 06 UM76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France
| | - Loïc Desquilbet
- Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, Unité D'Épidémiologie clinique et de Biostatistique, 94704 Maisons-Alfort, France. CNRS UMR 7179, MNHN, Brunoy 91800, France
| | - Wilson Savino
- Laboratory of Thymus Research, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Suse Dayse Silva-Barbosa
- Department of Clinical Research, National Cancer Institute (INCA), Rio de Janeiro 20230-130, Brazil
| | - Anne-Marie Faussat
- Université Pierre et Marie Curie-Paris 06 IFR 65 Saint-Antoine, Paris 75005, France
| | - Vincent Mouly
- Université Pierre et Marie Curie-Paris 06 UM76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France
| | - Thomas Voit
- Université Pierre et Marie Curie-Paris 06 UM76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France
| | - Stéphane Blot
- Université Paris-Est, Ecole Nationale Vétérinaire d'Alfort, UPR de Neurobiologie, 94704 Maisons-Alfort, France.
| | - Gillian Butler-Browne
- Université Pierre et Marie Curie-Paris 06 UM76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France.
| |
Collapse
|
39
|
Waugh TA, Horstick E, Hur J, Jackson SW, Davidson AE, Li X, Dowling JJ. Fluoxetine prevents dystrophic changes in a zebrafish model of Duchenne muscular dystrophy. Hum Mol Genet 2014; 23:4651-62. [PMID: 24760771 PMCID: PMC4119416 DOI: 10.1093/hmg/ddu185] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a common and relentlessly progressive muscle disease. Some interventions have been identified that modestly slow progression and prolong survival, but more meaningful therapies are lacking. The goal of this study is to identify new therapeutic pathways for DMD using a zebrafish model of the disease. To accomplish this, we performed a non-biased drug screen in sapje, a zebrafish line with a recessive nonsense mutation in dystrophin. We identified 6 positive hits (out of 640 total drugs tested) by their ability to prevent abnormal birefringence in sapje. Follow-up analyses demonstrated that fluoxetine, a selective serotonin reuptake inhibitor (SSRI), provided the most substantial benefit. Morpholino-based experimentation confirmed that modulation of the serotonin pathway alone can prevent the dystrophic phenotype, and transcriptomic analysis revealed changes in calcium homeostasis as a potential mechanism. In all, we demonstrate that monoamine agonists can prevent disease in a vertebrate model of DMD. Given the safe and widespread use of SSRIs in clinical practice, our study identifies an attractive target pathway for therapy development.
Collapse
MESH Headings
- Animals
- Base Sequence
- Birefringence
- Calcium/metabolism
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Dystrophin/metabolism
- Embryo, Nonmammalian/drug effects
- Embryo, Nonmammalian/metabolism
- Evans Blue/metabolism
- Fluoxetine/pharmacology
- Fluoxetine/therapeutic use
- Gene Expression Profiling
- Gene Knockdown Techniques
- Homeostasis/drug effects
- Molecular Sequence Data
- Morpholinos/pharmacology
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Oligonucleotide Array Sequence Analysis
- Phenotype
- Serotonin Plasma Membrane Transport Proteins/metabolism
- Stress, Mechanical
- Survival Analysis
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/physiology
Collapse
Affiliation(s)
| | | | | | | | - Ann E Davidson
- Division of Neurology and Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | | | - James J Dowling
- Department of Pediatrics, Department of Neurology, and Department of Neuroscience, University of Michigan, Ann Arbor, MI 48109-2200, USA Division of Neurology and Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada Department of Paediatrics, and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Brinkmeier H, Ohlendieck K. Chaperoning heat shock proteins: Proteomic analysis and relevance for normal and dystrophin-deficient muscle. Proteomics Clin Appl 2014; 8:875-95. [DOI: 10.1002/prca.201400015] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/24/2014] [Accepted: 05/28/2014] [Indexed: 12/15/2022]
Affiliation(s)
| | - Kay Ohlendieck
- Department of Biology; National University of Ireland; Maynooth Co. Kildare Ireland
| |
Collapse
|
41
|
Nakamura K, Fujii W, Tsuboi M, Tanihata J, Teramoto N, Takeuchi S, Naito K, Yamanouchi K, Nishihara M. Generation of muscular dystrophy model rats with a CRISPR/Cas system. Sci Rep 2014; 4:5635. [PMID: 25005781 PMCID: PMC4088098 DOI: 10.1038/srep05635] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/23/2014] [Indexed: 12/28/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked lethal muscle disorder caused by mutations in the Dmd gene encoding Dystrophin12. DMD model animals, such as mdx mice and canine X-linked muscular dystrophy dogs, have been widely utilized in the development of a treatment for DMD3. Here, we demonstrate the generation of Dmd-mutated rats using a clustered interspaced short palindromic repeats (CRISPR)/Cas system, an RNA-based genome engineering technique that is also adaptive to rats. We simultaneously targeted two exons in the rat Dmd gene, which resulted in the absence of Dystrophin expression in the F0 generation. Dmd-mutated rats exhibited a decline in muscle strength, and the emergence of degenerative/regenerative phenotypes in the skeletal muscle, heart, and diaphragm. These mutations were heritable by the next generation, and F1 male rats exhibited similar phenotypes in their skeletal muscles. These model rats should prove to be useful for developing therapeutic methods to treat DMD.
Collapse
Affiliation(s)
- Katsuyuki Nakamura
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Wataru Fujii
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masaya Tsuboi
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Naomi Teramoto
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shiho Takeuchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kunihiko Naito
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
42
|
Banks GB, Combs AC, Odom GL, Bloch RJ, Chamberlain JS. Muscle structure influences utrophin expression in mdx mice. PLoS Genet 2014; 10:e1004431. [PMID: 24922526 PMCID: PMC4055409 DOI: 10.1371/journal.pgen.1004431] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/24/2014] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by mutations in the dystrophin gene. To examine the influence of muscle structure on the pathogenesis of DMD we generated mdx4cv:desmin double knockout (dko) mice. The dko male mice died of apparent cardiorespiratory failure at a median age of 76 days compared to 609 days for the desmin−/− mice. An ∼2.5 fold increase in utrophin expression in the dko skeletal muscles prevented necrosis in ∼91% of 1a, 2a and 2d/x fiber-types. In contrast, utrophin expression was reduced in the extrasynaptic sarcolemma of the dko fast 2b fibers leading to increased membrane fragility and dystrophic pathology. Despite lacking extrasynaptic utrophin, the dko fast 2b fibers were less dystrophic than the mdx4cv fast 2b fibers suggesting utrophin-independent mechanisms were also contributing to the reduced dystrophic pathology. We found no overt change in the regenerative capacity of muscle stem cells when comparing the wild-type, desmin−/−, mdx4cv and dko gastrocnemius muscles injured with notexin. Utrophin could form costameric striations with α-sarcomeric actin in the dko to maintain the integrity of the membrane, but the lack of restoration of the NODS (nNOS, α-dystrobrevin 1 and 2, α1-syntrophin) complex and desmin coincided with profound changes to the sarcomere alignment in the diaphragm, deposition of collagen between the myofibers, and impaired diaphragm function. We conclude that the dko mice may provide new insights into the structural mechanisms that influence endogenous utrophin expression that are pertinent for developing a therapy for DMD. Duchenne muscular dystrophy (DMD) is a severe muscle wasting disorder caused by mutations in the dystrophin gene. Utrophin is structurally similar to dystrophin and improving its expression can prevent skeletal muscle necrosis in the mdx mouse model of DMD. Consequently, improving utrophin expression is a primary therapeutic target for treating DMD. While the downstream mechanisms that influence utrophin expression and stability are well described, the upstream mechanisms are less clear. Here, we found that perturbing the highly ordered structure of striated muscle by genetically deleting desmin from mdx mice increased utrophin expression to levels that prevented skeletal muscle necrosis. Thus, the mdx:desmin double knockout mice may prove valuable in determining the upstream mechanisms that influence utrophin expression to develop a therapy for DMD.
Collapse
Affiliation(s)
- Glen B. Banks
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Ariana C. Combs
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Guy L. Odom
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
43
|
Holland A, Dowling P, Ohlendieck K. New pathobiochemical insights into dystrophinopathy from the proteomics of senescent mdx mouse muscle. Front Aging Neurosci 2014; 6:109. [PMID: 24917816 PMCID: PMC4042888 DOI: 10.3389/fnagi.2014.00109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/18/2014] [Indexed: 01/08/2023] Open
Abstract
Primary abnormalities in the dystrophin gene cause X-linked muscular dystrophy, a highly progressive muscle wasting disorder of childhood. A spontaneous animal model of Duchenne muscular dystrophy is the mdx mouse, which presents a highly interesting phenotype that exhibits considerable variations in the degree of fiber degeneration in different subtypes of muscles. The idea that aging exacerbates the dystrophic mdx phenotype, as previously indicated by a large number of biochemical and cell biological studies, was clearly confirmed by comparative muscle proteomics. Here we outline recent findings of age-dependent changes in the dystrophin-deficient muscle proteome and contrast these results with the previously established proteomic profile of sarcopenic muscle. Besides comparable perturbations of various biochemical functions, especially striking are similarities in the cellular stress response associated with a drastic up-regulation of small αB-crystallin-like heat shock proteins. Hence, the comparison of large-scale proteomic data sets of natural muscle aging with dystrophic sarcopenia promises to shed light on the differential effect of sarcopenia of old age vs. senescent abnormalities on a mutant dystrophic background.
Collapse
Affiliation(s)
- Ashling Holland
- Department of Biology, National University of Ireland Maynooth, Ireland
| | - Paul Dowling
- Department of Biology, National University of Ireland Maynooth, Ireland
| | - Kay Ohlendieck
- Department of Biology, National University of Ireland Maynooth, Ireland
| |
Collapse
|
44
|
Faber RM, Hall JK, Chamberlain JS, Banks GB. Myofiber branching rather than myofiber hyperplasia contributes to muscle hypertrophy in mdx mice. Skelet Muscle 2014; 4:10. [PMID: 24910770 PMCID: PMC4047439 DOI: 10.1186/2044-5040-4-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 05/06/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle hypertrophy in the mdx mouse model of Duchenne muscular dystrophy (DMD) can partially compensate for the loss of dystrophin by maintaining peak force production. Histopathology examination of the hypertrophic muscles suggests the hypertrophy primarily results from the addition of myofibers, and is accompanied by motor axon branching. However, it is unclear whether an increased number of innervated myofibers (myofiber hyperplasia) contribute to muscle hypertrophy in the mdx mice. METHODS To better understand the cellular mechanisms of muscle hypertrophy in mdx mice, we directly compared the temporal progression of the dystrophic pathology in the extensor digitorum longus (EDL) muscle to myofiber number, myofiber branching, and innervation, from 3 to 20 weeks of age. RESULTS We found that a 28% increase in the number of fibers in transverse sections of muscle correlated with a 31% increase in myofiber branching. Notably, the largest increases in myofiber number and myofiber branching occurred after 12 weeks of age when the proportion of myofibers with central nuclei had stabilized and the mdx mouse had reached maturity. The dystrophic pathology coincided with profound changes to innervation of the muscles that included temporary denervation of necrotic fibers, fragmentation of synapses, and ultra-terminal axon sprouting. However, there was little evidence of synapse formation in the mdx mice from 3 to 20 weeks of age. Only 4.4% of neuromuscular junctions extended ultra-terminal synapses, which failed to mature, and the total number of neuromuscular junctions remained constant. CONCLUSIONS Muscle hypertrophy in mdx mice results from myofiber branching rather than myofiber hyperplasia.
Collapse
Affiliation(s)
- Rachel M Faber
- Department of Neurology, University of Washington, Seattle, WA 98195-7720, USA
| | - John K Hall
- Department of Neurology, University of Washington, Seattle, WA 98195-7720, USA
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington, Seattle, WA 98195-7720, USA ; Department of Biochemistry, University of Washington, Seattle, WA 98195-7720, USA ; Department of Medicine, University of Washington, Seattle, WA 98195-7720, USA
| | - Glen B Banks
- Department of Neurology, University of Washington, Seattle, WA 98195-7720, USA
| |
Collapse
|
45
|
Kharraz Y, Guerra J, Pessina P, Serrano AL, Muñoz-Cánoves P. Understanding the process of fibrosis in Duchenne muscular dystrophy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:965631. [PMID: 24877152 PMCID: PMC4024417 DOI: 10.1155/2014/965631] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/08/2014] [Indexed: 02/06/2023]
Abstract
Fibrosis is the aberrant deposition of extracellular matrix (ECM) components during tissue healing leading to loss of its architecture and function. Fibrotic diseases are often associated with chronic pathologies and occur in a large variety of vital organs and tissues, including skeletal muscle. In human muscle, fibrosis is most readily associated with the severe muscle wasting disorder Duchenne muscular dystrophy (DMD), caused by loss of dystrophin gene function. In DMD, skeletal muscle degenerates and is infiltrated by inflammatory cells and the functions of the muscle stem cells (satellite cells) become impeded and fibrogenic cells hyperproliferate and are overactivated, leading to the substitution of skeletal muscle with nonfunctional fibrotic tissue. Here, we review new developments in our understanding of the mechanisms leading to fibrosis in DMD and several recent advances towards reverting it, as potential treatments to attenuate disease progression.
Collapse
Affiliation(s)
- Yacine Kharraz
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Joana Guerra
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Patrizia Pessina
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Antonio L. Serrano
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| |
Collapse
|
46
|
The Dynamics of Compound, Transcript, and Protein Effects After Treatment With 2OMePS Antisense Oligonucleotides in mdx Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e148. [PMID: 24549299 PMCID: PMC3950770 DOI: 10.1038/mtna.2014.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 12/24/2013] [Indexed: 12/28/2022]
Abstract
Antisense-mediated exon skipping is currently in clinical development for Duchenne muscular dystrophy (DMD) to amend the consequences of the underlying genetic defect and restore dystrophin expression. Due to turnover of compound, transcript, and protein, chronic treatment with effector molecules (antisense oligonucleotides) will be required. To investigate the dynamics and persistence of antisense 2′-O-methyl phosphorothioate oligonucleotides, exon skipping, and dystrophin expression after dosing was concluded, mdx mice were treated subcutaneously for 8 weeks with 100 mg/kg oligonucleotides twice weekly. Thereafter, mice were sacrificed at different time points after the final injection (36 hours–24 weeks). Oligonucleotide half-life was longer in heart (~65 days) compared with that in skeletal muscle, liver, and kidney (~35 days). Exon skipping half-lives varied between 33 and 53 days, whereas dystrophin protein showed a long half-life (>100 days). Oligonucleotide and exon-skipping levels peaked in the first week and declined thereafter. By contrast, dystrophin expression peaked after 3–8 weeks and then slowly declined, remaining detectable after 24 weeks. Concordance between levels of oligonucleotides, exon skipping, and proteins was observed, except in heart, wherein high oligonucleotide levels but low exon skipping and dystrophin expression were seen. Overall, these results enhance our understanding of the pharmacokinetics and pharmacodynamics of 2′-O-methyl phosphorothioate oligos used for the treatment of DMD.
Collapse
|
47
|
Brinkmeyer-Langford C, Kornegay JN. Comparative Genomics of X-linked Muscular Dystrophies: The Golden Retriever Model. Curr Genomics 2014; 14:330-42. [PMID: 24403852 PMCID: PMC3763684 DOI: 10.2174/13892029113149990004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 12/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease that dramatically decreases the lifespan and abilities of affected young people. The primary molecular cause of the disease is the absence of functional dystrophin protein, which is critical to proper muscle function. Those with DMD vary in disease presentation and dystrophin mutation; the same causal mutation may be associated with drastically different levels of disease severity. Also contributing to this variation are the influences of additional modifying genes and/or changes in functional elements governing such modifiers. This genetic heterogeneity complicates the efficacy of treatment methods and to date medical interventions are limited to treating symptoms. Animal models of DMD have been instrumental in teasing out the intricacies of DMD disease and hold great promise for advancing knowledge of its variable presentation and treatment. This review addresses the utility of comparative genomics in elucidating the complex background behind phenotypic variation in a canine model of DMD, Golden Retriever muscular dystrophy (GRMD). This knowledge can be exploited in the development of improved, more personalized treatments for DMD patients, such as therapies that can be tailor-matched to the disease course and genomic background of individual patients.
Collapse
Affiliation(s)
- Candice Brinkmeyer-Langford
- Texas A&M University College of Veterinary Medicine, Dept. of Veterinary Integrative Biosciences - Mailstop 4458, College Station, Texas, U.S.A. 77843-4458
| | - Joe N Kornegay
- Texas A&M University College of Veterinary Medicine, Dept. of Veterinary Integrative Biosciences - Mailstop 4458, College Station, Texas, U.S.A. 77843-4458
| |
Collapse
|
48
|
Arnett AL, Konieczny P, Ramos JN, Hall J, Odom G, Yablonka-Reuveni Z, Chamberlain JR, Chamberlain JS. Adeno-associated viral (AAV) vectors do not efficiently target muscle satellite cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:S2329-0501(16)30105-X. [PMID: 25580445 PMCID: PMC4288464 DOI: 10.1038/mtm.2014.38] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adeno-associated viral (AAV) vectors are becoming an important tool for gene therapy of numerous genetic and other disorders. Several recombinant AAV vectors (rAAV) have the ability to transduce striated muscles in a variety of animals following intramuscular and intravascular administration, and have attracted widespread interest for therapy of muscle disorders such as the muscular dystrophies. However, most studies have focused on the ability to transduce mature muscle cells, and have not examined the ability to target myogenic stem cells such as skeletal muscle satellite cells. Here we examined the relative ability of rAAV vectors derived from AAV6 to target myoblasts, myocytes, and myotubes in culture and satellite cells and myofibers in vivo. AAV vectors are able to transduce proliferating myoblasts in culture, albeit with reduced efficiency relative to postmitotic myocytes and myotubes. In contrast, quiescent satellite cells are refractory to transduction in adult mice. These results suggest that while muscle disorders characterized by myofiber regeneration can be slowed or halted by AAV transduction, little if any vector transduction can be obtained in myogenic stems cells that might other wise support ongoing muscle regeneration.
Collapse
Affiliation(s)
- Andrea Lh Arnett
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, USA ; Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Molecular and Cellular Biology Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Patryk Konieczny
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Julian N Ramos
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Molecular and Cellular Biology Program, University of Washington School of Medicine, Seattle, WA, USA
| | - John Hall
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Guy Odom
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA, USA
| | - Joel R Chamberlain
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA ; Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
49
|
Hollinger K, Yang CX, Montz RE, Nonneman D, Ross JW, Selsby JT. Dystrophin insufficiency causes selective muscle histopathology and loss of dystrophin-glycoprotein complex assembly in pig skeletal muscle. FASEB J 2013; 28:1600-9. [PMID: 24347611 DOI: 10.1096/fj.13-241141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The purpose of this investigation was to determine the extent to which dystrophin insufficiency caused histomorphological changes in a novel pig model of Becker muscular dystrophy. In our procedures, we used a combination of biochemical approaches, including quantitative PCR and Western blots, along with a histological analysis using standard and immunohistological measures. We found that 8-wk-old male affected pigs had a 70% reduction in dystrophin protein abundance in the diaphragm, psoas major, and longissimus lumborum and a 5-fold increase in serum creatine kinase activity compared with healthy male littermates. Dystrophin insufficiency in the diaphragm and the longissimus resulted in muscle histopathology with disorganized fibrosis that often colocalized with fatty infiltration but not the psoas. Affected animals also had an 80-85% reduction in α-sarcoglycan localization in these muscles, indicating compromised assembly of the dystrophin glycoprotein complex. Controls used in this study were 4 healthy male littermates, as they are most closely related to the affected animals. We concluded that pigs with insufficient dystrophin protein expression have a phenotype consistent with human dystrophinopathy patients. Given that and their similarity in body size and physiology to humans, we further conclude that this pig line is an appropriate translational model for dystrophinopathies.
Collapse
Affiliation(s)
- Katrin Hollinger
- 1Iowa State University, Department of Animal Science, 2356 Kildee Hall, Ames, IA 50011, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Carberry S, Zweyer M, Swandulla D, Ohlendieck K. Application of fluorescence two-dimensional difference in-gel electrophoresis as a proteomic biomarker discovery tool in muscular dystrophy research. BIOLOGY 2013; 2:1438-64. [PMID: 24833232 PMCID: PMC4009800 DOI: 10.3390/biology2041438] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/05/2013] [Accepted: 11/12/2013] [Indexed: 12/31/2022]
Abstract
In this article, we illustrate the application of difference in-gel electrophoresis for the proteomic analysis of dystrophic skeletal muscle. The mdx diaphragm was used as a tissue model of dystrophinopathy. Two-dimensional gel electrophoresis is a widely employed protein separation method in proteomic investigations. Although two-dimensional gels usually underestimate the cellular presence of very high molecular mass proteins, integral membrane proteins and low copy number proteins, this method is extremely powerful in the comprehensive analysis of contractile proteins, metabolic enzymes, structural proteins and molecular chaperones. This gives rise to two-dimensional gel electrophoretic separation as the method of choice for studying contractile tissues in health and disease. For comparative studies, fluorescence difference in-gel electrophoresis has been shown to provide an excellent biomarker discovery tool. Since aged diaphragm fibres from the mdx mouse model of Duchenne muscular dystrophy closely resemble the human pathology, we have carried out a mass spectrometry-based comparison of the naturally aged diaphragm versus the senescent dystrophic diaphragm. The proteomic comparison of wild type versus mdx diaphragm resulted in the identification of 84 altered protein species. Novel molecular insights into dystrophic changes suggest increased cellular stress, impaired calcium buffering, cytostructural alterations and disturbances of mitochondrial metabolism in dystrophin-deficient muscle tissue.
Collapse
Affiliation(s)
- Steven Carberry
- Department of Biology, National University of Ireland, Maynooth, Kildare, Ireland.
| | - Margit Zweyer
- Department of Physiology II, University of Bonn, Bonn D-53115, Germany.
| | - Dieter Swandulla
- Department of Physiology II, University of Bonn, Bonn D-53115, Germany.
| | - Kay Ohlendieck
- Department of Biology, National University of Ireland, Maynooth, Kildare, Ireland.
| |
Collapse
|