1
|
High-Throughput Screening to Identify Modulators of Sarcospan. Methods Mol Biol 2022; 2587:479-493. [PMID: 36401045 DOI: 10.1007/978-1-0716-2772-3_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
High-throughput screening enables the discovery of disease-modifying small molecules. Here, we describe the development of a scalable, cell-based assay to screen for small molecules that modulate sarcospan for the treatment of Duchenne muscular dystrophy. We detail the hit validation pipeline, which includes secondary screening, gene/protein quantification, and an in vitro membrane stability assay.
Collapse
|
2
|
Natural disease history of mouse models for limb girdle muscular dystrophy types 2D and 2F. PLoS One 2017; 12:e0182704. [PMID: 28797108 PMCID: PMC5552258 DOI: 10.1371/journal.pone.0182704] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/24/2017] [Indexed: 12/22/2022] Open
Abstract
Limb-girdle muscular dystrophy types 2D and 2F (LGMD 2D and 2F) are autosomal recessive disorders caused by mutations in the alpha- and delta sarcoglycan genes, respectively, leading to severe muscle weakness and degeneration. The cause of the disease has been well characterized and a number of animal models are available for pre-clinical studies to test potential therapeutic interventions. To facilitate transition from drug discovery to clinical trials, standardized procedures and natural disease history data were collected for these mouse models. Implementing the TREAD-NMD standardized operating procedures, we here subjected LGMD2D (SGCA-null), LGMD2F (SGCD-null) and wild type (C57BL/6J) mice to five functional tests from the age of 4 to 32 weeks. To assess whether the functional test regime interfered with disease pathology, sedentary groups were taken along. Muscle physiology testing of tibialis anterior muscle was performed at the age of 34 weeks. Muscle histopathology and gene expression was analysed in skeletal muscles and heart. Muscle histopathology and gene expression was analysed in skeletal muscles and heart. Mice successfully accomplished the functional tests, which did not interfere with disease pathology. Muscle function of SGCA- and SGCD-null mice was impaired and declined over time. Interestingly, female SGCD-null mice outperformed males in the two and four limb hanging tests, which proved the most suitable non-invasive tests to assess muscle function. Muscle physiology testing of tibialis anterior muscle revealed lower specific force and higher susceptibility to eccentric-induced damage in LGMD mice. Analyzing muscle histopathology and gene expression, we identified the diaphragm as the most affected muscle in LGMD strains. Cardiac fibrosis was found in SGCD-null mice, being more severe in males than in females. Our study offers a comprehensive natural history dataset which will be useful to design standardized tests and future pre-clinical studies in LGMD2D and 2F mice.
Collapse
|
3
|
Cox ML, Evans JM, Davis AG, Guo LT, Levy JR, Starr-Moss AN, Salmela E, Hytönen MK, Lohi H, Campbell KP, Clark LA, Shelton GD. Exome sequencing reveals independent SGCD deletions causing limb girdle muscular dystrophy in Boston terriers. Skelet Muscle 2017; 7:15. [PMID: 28697784 PMCID: PMC5506588 DOI: 10.1186/s13395-017-0131-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/23/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophies (LGMDs) are a heterogeneous group of inherited autosomal myopathies that preferentially affect voluntary muscles of the shoulders and hips. LGMD has been clinically described in several breeds of dogs, but the responsible mutations are unknown. The clinical presentation in dogs is characterized by marked muscle weakness and atrophy in the shoulder and hips during puppyhood. METHODS Following clinical evaluation, the identification of the dystrophic histological phenotype on muscle histology, and demonstration of the absence of sarcoglycan-sarcospan complex by immunostaining, whole exome sequencing was performed on five Boston terriers: one affected dog and its three family members and one unrelated affected dog. RESULTS Within sarcoglycan-δ (SGCD), a two base pair deletion segregating with LGMD in the family was discovered, and a deletion encompassing exons 7 and 8 was found in the unrelated dog. Both mutations are predicted to cause an absence of SGCD protein, confirmed by immunohistochemistry. The mutations are private to each family. CONCLUSIONS Here, we describe the first cases of canine LGMD characterized at the molecular level with the classification of LGMD2F.
Collapse
Affiliation(s)
- Melissa L. Cox
- CAG GmbH - Center for Animal Genetics, Paul-Ehrlich-Str. 23, 72076 Tubingen, Germany
| | - Jacquelyn M. Evans
- Department of Genetics and Biochemistry, Clemson University, 130 McGinty Ct., Clemson, SC 29634 USA
| | - Alexander G. Davis
- Department of Genetics and Biochemistry, Clemson University, 130 McGinty Ct., Clemson, SC 29634 USA
| | - Ling T. Guo
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| | - Jennifer R. Levy
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, Iowa 52242-1101 USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, Iowa 52242 USA
| | - Alison N. Starr-Moss
- Department of Genetics and Biochemistry, Clemson University, 130 McGinty Ct., Clemson, SC 29634 USA
| | - Elina Salmela
- Department of Veterinary Biosciences and Research Programs Unit, University of Helsinki, Haartmaninkatu 8, FI-00290 Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Marjo K. Hytönen
- Department of Veterinary Biosciences and Research Programs Unit, University of Helsinki, Haartmaninkatu 8, FI-00290 Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Hannes Lohi
- Department of Veterinary Biosciences and Research Programs Unit, University of Helsinki, Haartmaninkatu 8, FI-00290 Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Kevin P. Campbell
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, Iowa 52242-1101 USA
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, Iowa 52242 USA
| | - Leigh Anne Clark
- Department of Genetics and Biochemistry, Clemson University, 130 McGinty Ct., Clemson, SC 29634 USA
| | - G. Diane Shelton
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| |
Collapse
|
4
|
Cutroneo G, Bramanti P, Favaloro A, Anastasi G, Trimarchi F, Di Mauro D, Rinaldi C, Speciale F, Inferrera A, Santoro G, Arena S, Patricolo M, Magno C. Sarcoglycan complex in human normal and pathological prostatic tissue: an immunohistochemical and RT-PCR study. Anat Rec (Hoboken) 2013; 297:327-36. [PMID: 24347395 DOI: 10.1002/ar.22846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 11/03/2013] [Indexed: 11/11/2022]
Abstract
The sarcoglycan complex is a trans-membrane system playing a key role in mechano-signaling the connection from the cytoskeleton to the extracellular matrix. While b-, d-, and e-sarcoglycans are widely distributed, g- and a-sarcoglycans are expressed exclusively in skeletal and cardiac muscle. Insufficient data are available on the distribution of sarcoglycans in nonmuscular tissue. In the present study, we used immunohistochemical and RT-PCR techniques to study the sarcoglycans also in normal human glandular tissue, a type of tissue never studied in relation to the sarcoglycan complex, with the aim of verifying the real wider distribution of this complex. To understand the role of sarcoglycans, we tested specimens collected from patients affected by benign prostatic hyperplasia and adenocarcinoma. For the first time, our results showed that all sarcoglycans are detectable in normal samples both in epithelial and in myoepithelial cells; in pathological prostate, sarcoglycans appeared severely reduced in number or were absent. These data demonstrated that all sarcoglycans have a wider distribution suggesting a new unknown role for these proteins. The decreased number of sarcoglycans, containing cadherin domain homologs in samples of prostate affected by hyperplasia, and the absence of proteins in prostate biopsies, in cases affected by adenocarcinoma, could be responsible for the loss of adhesion between epithelial cells, which in turn facilitates the progression of benign tumors and the invasive potential of malignant tumors.
Collapse
|
5
|
Palma-Flores C, Ramírez-Sánchez I, Rosas-Vargas H, Canto P, Coral-Vázquez RM. Description of a utrophin associated protein complex in lipid raft domains of human artery smooth muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1047-54. [PMID: 24060563 DOI: 10.1016/j.bbamem.2013.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 09/06/2013] [Accepted: 09/12/2013] [Indexed: 01/08/2023]
Abstract
The dystrophin-associated protein complex (DAPC) is a multimeric complex that links the extracellular matrix to the actin cytoskeleton, and in some cases dystrophin can be substituted by its autosomal homologue utrophin to form the utrophin-associated protein complex (UAPC). Both complexes maintain the stability of plasma membrane during contraction process and play an important role in transmembrane signaling. Mutations in members of the DAPC are associated with muscular dystrophy and dilated cardiomyopathy. In a previous study with human umbilical cord vessels, we observed that utrophin colocalize with caveolin-1 (Cav-1) which proposed the presence of UAPC in the plasma membrane of vascular smooth muscle (VSM). In the current study, we demonstrated by immunofluorescence analysis, co-immunoprecipitation assays, and subcellular fractionation by sucrose gradients, the existence of an UAPC in lipid raft domains of human umbilical artery smooth muscle cells (HUASMC). This complex is constituted by utrophin, β-DG, ε-SG, α-smooth muscle actin, Cav-1, endothelial nitric oxide synthase (eNOS) and cavin-1. It was also observed the presence of dystrophin, utrophin Dp71, β-SG, δ-SG, δ-SG3 and sarcospan in non-lipid raft fractions. Furthermore, the knockdown of α/β-DG was associated with the decrease in both the synthesis of nitric oxide (NO) and the presence of the phosphorylated (active) form of eNOS; and with a reduction in the downstream activation of some cGMP signaling transduction pathway components. Together these results show the presence of an UAPC complex in HUASMC that may participate in the activity regulation of eNOS and in the vascular function.
Collapse
Affiliation(s)
- Carlos Palma-Flores
- División de Investigación Biomédica, Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico
| | - Israel Ramírez-Sánchez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D.F., Mexico
| | - Haydeé Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Medico Nacional Siglo XXI-IMSS, Av. Cuauhtémoc No 330, Col Doctores, Delegación Cuauhtémoc, 06725 México, D.F., Mexico
| | - Patricia Canto
- División de Investigación Biomédica, Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico
| | - Ramón Mauricio Coral-Vázquez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, D.F., Mexico; Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, México, D.F., Mexico.
| |
Collapse
|
6
|
Saxena R, Saleheen D, Been LF, Garavito ML, Braun T, Bjonnes A, Young R, Ho WK, Rasheed A, Frossard P, Sim X, Hassanali N, Radha V, Chidambaram M, Liju S, Rees SD, Ng DPK, Wong TY, Yamauchi T, Hara K, Tanaka Y, Hirose H, McCarthy MI, Morris AP, Basit A, Barnett AH, Katulanda P, Matthews D, Mohan V, Wander GS, Singh JR, Mehra NK, Ralhan S, Kamboh MI, Mulvihill JJ, Maegawa H, Tobe K, Maeda S, Cho YS, Tai ES, Kelly MA, Chambers JC, Kooner JS, Kadowaki T, Deloukas P, Rader DJ, Danesh J, Sanghera DK. Genome-wide association study identifies a novel locus contributing to type 2 diabetes susceptibility in Sikhs of Punjabi origin from India. Diabetes 2013; 62:1746-1755. [PMID: 23300278 PMCID: PMC3636649 DOI: 10.2337/db12-1077] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 11/22/2012] [Indexed: 12/16/2022]
Abstract
We performed a genome-wide association study (GWAS) and a multistage meta-analysis of type 2 diabetes (T2D) in Punjabi Sikhs from India. Our discovery GWAS in 1,616 individuals (842 case subjects) was followed by in silico replication of the top 513 independent single nucleotide polymorphisms (SNPs) (P < 10⁻³) in Punjabi Sikhs (n = 2,819; 801 case subjects). We further replicated 66 SNPs (P < 10⁻⁴) through genotyping in a Punjabi Sikh sample (n = 2,894; 1,711 case subjects). On combined meta-analysis in Sikh populations (n = 7,329; 3,354 case subjects), we identified a novel locus in association with T2D at 13q12 represented by a directly genotyped intronic SNP (rs9552911, P = 1.82 × 10⁻⁸) in the SGCG gene. Next, we undertook in silico replication (stage 2b) of the top 513 signals (P < 10⁻³) in 29,157 non-Sikh South Asians (10,971 case subjects) and de novo genotyping of up to 31 top signals (P < 10⁻⁴) in 10,817 South Asians (5,157 case subjects) (stage 3b). In combined South Asian meta-analysis, we observed six suggestive associations (P < 10⁻⁵ to < 10⁻⁷), including SNPs at HMG1L1/CTCFL, PLXNA4, SCAP, and chr5p11. Further evaluation of 31 top SNPs in 33,707 East Asians (16,746 case subjects) (stage 3c) and 47,117 Europeans (8,130 case subjects) (stage 3d), and joint meta-analysis of 128,127 individuals (44,358 case subjects) from 27 multiethnic studies, did not reveal any additional loci nor was there any evidence of replication for the new variant. Our findings provide new evidence on the presence of a population-specific signal in relation to T2D, which may provide additional insights into T2D pathogenesis.
Collapse
Affiliation(s)
- Richa Saxena
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Pakistan
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
- Departments of Biostatistics and Epidemiology and Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Latonya F. Been
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Martha L. Garavito
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Timothy Braun
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew Bjonnes
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Robin Young
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Weang Kee Ho
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | - Xueling Sim
- Center for Statistical Genetics and Department of Statistics, University of Michigan, Ann Arbor, Michigan
- Centre for Molecular Epidemiology, National University of Singapore, Singapore
| | - Neelam Hassanali
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | | | | | - Samuel Liju
- Madras Diabetes Research Foundation, Chennai, India
| | - Simon D. Rees
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
- Diabetes Centre, Heart of England National Health Service Foundation Trust, Birmingham, U.K
| | - Daniel Peng-Keat Ng
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuo Hara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Integrated Molecular Science on Metabolic Diseases, 22nd Century Medical and Research Center, The University of Tokyo, Tokyo, Japan
| | - Yasushi Tanaka
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroshi Hirose
- Health Center, Keio University School of Medicine, Tokyo, Japan
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
| | | | | | | | - Abdul Basit
- Baqai Institute of Diabetology and Endocrinology, Karachi, Pakistan
| | - Anthony H. Barnett
- Diabetes Centre, Heart of England National Health Service Foundation Trust, Birmingham, U.K
| | - Prasad Katulanda
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Diabetes Research Unit, Department of Clinical Medicine, University of Colombo, Colombo, Sri Lanka
| | - David Matthews
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Chennai, India
- Dr. Mohan's Diabetes Specialities Centre, Chennai, India
| | - Gurpreet S. Wander
- Hero Dayanand Medical College and Heart Institute, Ludhiana, Punjab, India
| | - Jai Rup Singh
- Central University of Punjab, Bathinda, Punjab, India
| | - Narinder K. Mehra
- All India Institute of Medical Sciences and Research, New Delhi, India
| | - Sarju Ralhan
- Hero Dayanand Medical College and Heart Institute, Ludhiana, Punjab, India
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John J. Mulvihill
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Shiro Maeda
- Laboratory for Endocrinology and Metabolism, RIKEN Center for Genomic Medicine, Kanagawa, Japan
| | - Yoon S. Cho
- Department of Biomedical Science, Hallym University, Chuncheon, Gangwon-do 200-702, Republic of Korea
| | - E. Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Graduate Medical School Singapore, Singapore
| | - M. Ann Kelly
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
- Diabetes Centre, Heart of England National Health Service Foundation Trust, Birmingham, U.K
| | - John C. Chambers
- Ealing Hospital National Health Service Trust, Middlesex, U.K
- Imperial College Healthcare National Health Service Trust, London, U.K
- Epidemiology and Biostatistics, Imperial College London, London, U.K
| | - Jaspal S. Kooner
- Ealing Hospital National Health Service Trust, Middlesex, U.K
- Imperial College Healthcare National Health Service Trust, London, U.K
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital, London, U.K
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Daniel J. Rader
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Dharambir K. Sanghera
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
7
|
Marshall JL, Crosbie-Watson RH. Sarcospan: a small protein with large potential for Duchenne muscular dystrophy. Skelet Muscle 2013; 3:1. [PMID: 23282144 PMCID: PMC3599653 DOI: 10.1186/2044-5040-3-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 11/27/2012] [Indexed: 01/09/2023] Open
Abstract
Purification of the proteins associated with dystrophin, the gene product responsible for Duchenne muscular dystrophy, led to the discovery of the dystrophin-glycoprotein complex. Sarcospan, a 25-kDa transmembrane protein, was the last component to be identified and its function in skeletal muscle has been elusive. This review will focus on progress over the last decade revealing that sarcospan is an important regulator of muscle cell adhesion, strength, and regeneration. Investigations using several transgenic mouse models demonstrate that overexpression of sarcospan in the mouse model for Duchenne muscular dystrophy ameliorates pathology and restores muscle cell binding to laminin. Sarcospan improves cell surface expression of the dystrophin- and utrophin-glycoprotein complexes as well as α7β1 integrin, which are the three major laminin-binding complexes in muscle. Utrophin and α7β1 integrin compensate for the loss of dystrophin and the finding that sarcospan increases their abundance at the extra-synaptic sarcolemma supports the use of sarcospan as a therapeutic target. Newly discovered phenotypes in sarcospan-deficient mice, including a reduction in specific force output and increased drop in force in the diaphragm muscle, result from decreased utrophin and dystrophin expression and further reveal sarcospan’s role in determining abundance of these complexes. Dystrophin protein levels and the specific force output of the diaphragm muscle are further reduced upon genetic removal of α7 integrin (Itga7) in SSPN-deficient mice, demonstrating that interactions between integrin and sarcospan are critical for maintenance of the dystrophin-glycoprotein complex and force production of the diaphragm muscle. Sarcospan is a major regulator of Akt signaling pathways and sarcospan-deficiency significantly impairs muscle regeneration, a process that is dependent on Akt activation. Intriguingly, sarcospan regulates glycosylation of a specific subpopulation of α-dystroglycan, the laminin-binding receptor associated with dystrophin and utrophin, localized to the neuromuscular junction. Understanding the basic mechanisms responsible for assembly and trafficking of the dystrophin- and utrophin-glycoprotein complexes to the cell surface is lacking and recent studies suggest that sarcospan plays a role in these essential processes.
Collapse
Affiliation(s)
- Jamie L Marshall
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E, Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
| | | |
Collapse
|
8
|
Rauch U, Shami A, Zhang F, Carmignac V, Durbeej M, Hultgårdh-Nilsson A. Increased neointimal thickening in dystrophin-deficient mdx mice. PLoS One 2012; 7:e29904. [PMID: 22238670 PMCID: PMC3251593 DOI: 10.1371/journal.pone.0029904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 12/08/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The dystrophin gene, which is mutated in Duchenne muscular dystrophy (DMD), encodes a large cytoskeletal protein present in muscle fibers. While dystrophin in skeletal muscle has been extensively studied, the function of dystrophin in vascular smooth muscle is less clear. Here, we have analyzed the role of dystrophin in injury-induced arterial neointima formation. METHODOLOGY/PRINCIPAL FINDINGS We detected a down-regulation of dystrophin, dystroglycan and β-sarcoglycan mRNA expression when vascular smooth muscle cells de-differentiate in vitro. To further mimic development of intimal lesions, we performed a collar-induced injury of the carotid artery in the mdx mouse, a model for DMD. As compared with control mice, mdx mice develop larger lesions with increased numbers of proliferating cells. In vitro experiments demonstrate increased migration of vascular smooth muscle cells from mdx mice whereas the rate of proliferation was similar in cells isolated from wild-type and mdx mice. CONCLUSIONS/SIGNIFICANCE These results show that dystrophin deficiency stimulates neointima formation and suggest that expression of dystrophin in vascular smooth muscle cells may protect the artery wall against injury-induced intimal thickening.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cells, Cultured
- Dystrophin/deficiency
- Dystrophin/genetics
- Dystrophin/metabolism
- Dystrophin/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscular Dystrophy, Animal/complications
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/genetics
- Neointima/metabolism
- Neointima/pathology
- Organ Size
- Up-Regulation
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
Collapse
Affiliation(s)
- Uwe Rauch
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Annelie Shami
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Feng Zhang
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Virginie Carmignac
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
9
|
Fine mapping of a QTL on chromosome 13 for submaximal exercise capacity training response: the HERITAGE Family Study. Eur J Appl Physiol 2011; 112:2969-78. [PMID: 22170014 DOI: 10.1007/s00421-011-2274-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 12/02/2011] [Indexed: 10/14/2022]
Abstract
Although regular exercise improves submaximal aerobic capacity, there is large variability in its response to exercise training. While this variation is thought to be partly due to genetic differences, relatively little is known about the causal genes. Submaximal aerobic capacity traits in the current report include the responses of oxygen consumption (ΔVO(2)60), power output (ΔWORK60), and cardiac output (ΔQ60) at 60% of VO2max to a standardized 20-week endurance exercise training program. Genome-wide linkage analysis in 475 HERITAGE Family Study Caucasians identified a locus on chromosome 13q for ΔVO(2)60 (LOD = 3.11). Follow-up fine mapping involved a dense marker panel of over 1,800 single-nucleotide polymorphisms (SNPs) in a 7.9-Mb region (21.1-29.1 Mb from p-terminus). Single-SNP analyses found 14 SNPs moderately associated with both ΔVO(2)60 at P ≤ 0.005 and the correlated traits of ΔWORK60 and ΔQ60 at P < 0.05. Haplotype analyses provided several strong signals (P < 1.0 × 10(-5)) for ΔVO(2)60. Overall, association analyses narrowed the target region and included potential biological candidate genes (MIPEP and SGCG). Consistent with maximal heritability estimates of 23%, up to 20% of the phenotypic variance in ΔVO(2)60 was accounted for by these SNPs. These results implicate candidate genes on chromosome 13q12 for the ability to improve submaximal exercise capacity in response to regular exercise. Submaximal exercise at 60% of maximal capacity is an exercise intensity that falls well within the range recommended in the Physical Activity Guidelines for Americans and thus has potential public health relevance.
Collapse
|
10
|
Biglycan recruits utrophin to the sarcolemma and counters dystrophic pathology in mdx mice. Proc Natl Acad Sci U S A 2010; 108:762-7. [PMID: 21187385 DOI: 10.1073/pnas.1013067108] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in dystrophin and the subsequent disruption of the dystrophin-associated protein complex (DAPC). Utrophin is a dystrophin homolog expressed at high levels in developing muscle that is an attractive target for DMD therapy. Here we show that the extracellular matrix protein biglycan regulates utrophin expression in immature muscle and that recombinant human biglycan (rhBGN) increases utrophin expression in cultured myotubes. Systemically delivered rhBGN up-regulates utrophin at the sarcolemma and reduces muscle pathology in the mdx mouse model of DMD. RhBGN treatment also improves muscle function as judged by reduced susceptibility to eccentric contraction-induced injury. Utrophin is required for the rhBGN therapeutic effect. Several lines of evidence indicate that biglycan acts by recruiting utrophin protein to the muscle membrane. RhBGN is well tolerated in animals dosed for as long as 3 months. We propose that rhBGN could be a therapy for DMD.
Collapse
|
11
|
Groh S, Zong H, Goddeeris MM, Lebakken CS, Venzke D, Pessin JE, Campbell KP. Sarcoglycan complex: implications for metabolic defects in muscular dystrophies. J Biol Chem 2009; 284:19178-82. [PMID: 19494113 PMCID: PMC2740540 DOI: 10.1074/jbc.c109.010728] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The sarcoglycans are known as an integral subcomplex of the dystrophin glycoprotein complex, the function of which is best characterized in skeletal muscle in relation to muscular dystrophies. Here we demonstrate that the white adipocytes, which share a common precursor with the myocytes, express a cell-specific sarcoglycan complex containing β-, δ-, and ϵ-sarcoglycan. In addition, the adipose sarcoglycan complex associates with sarcospan and laminin binding dystroglycan. Using multiple sarcoglycan null mouse models, we show that loss of α-sarcoglycan has no consequence on the expression of the adipocyte sarcoglycan complex. However, loss of β- or δ-sarcoglycan leads to a concomitant loss of the sarcoglycan complex as well as sarcospan and a dramatic reduction in dystroglycan in adipocytes. We further demonstrate that β-sarcoglycan null mice, which lack the sarcoglycan complex in adipose tissue and skeletal muscle, are glucose-intolerant and exhibit whole body insulin resistance specifically due to impaired insulin-stimulated glucose uptake in skeletal muscles. Thus, our data demonstrate a novel function of the sarcoglycan complex in whole body glucose homeostasis and skeletal muscle metabolism, suggesting that the impairment of the skeletal muscle metabolism influences the pathogenesis of muscular dystrophy.
Collapse
Affiliation(s)
- Séverine Groh
- Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242-1101, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Arena S, Favaloro A, Cutroneo G, Consolo A, Arena F, Anastasi G, Di Benedetto V. Sarcoglycan Subcomplex Expression in Refluxing Ureteral Endings. J Urol 2008; 179:1980-6; discussion 1986. [DOI: 10.1016/j.juro.2008.01.059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Indexed: 10/22/2022]
Affiliation(s)
- Salvatore Arena
- Department of Pediatric Surgery, Unit of Pediatric Surgery, University of Catania, Catania, Italy
| | - Angelo Favaloro
- Departments of Biomorphology and Biotechnologies, University of Messina, Messina, Italy
| | - Giuseppina Cutroneo
- Departments of Biomorphology and Biotechnologies, University of Messina, Messina, Italy
| | - Angela Consolo
- Departments of Biomorphology and Biotechnologies, University of Messina, Messina, Italy
| | - Francesco Arena
- Departments of Medical and Surgical Pediatric Sciences, Unit of Pediatric Surgery, University of Messina, Messina, Italy
| | - Giuseppe Anastasi
- Departments of Biomorphology and Biotechnologies, University of Messina, Messina, Italy
| | - Vincenzo Di Benedetto
- Department of Pediatric Surgery, Unit of Pediatric Surgery, University of Catania, Catania, Italy
| |
Collapse
|
13
|
Hjermind LE, Vissing J, Asmus F, Krag T, Lochmüller H, Walter MC, Erdal J, Blake DJ, Nielsen JE. No muscle involvement in myoclonus-dystonia caused by epsilon-sarcoglycan gene mutations. Eur J Neurol 2008; 15:525-9. [PMID: 18355305 DOI: 10.1111/j.1468-1331.2008.02116.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mutations in the epsilon-sarcoglycan gene (SGCE) can cause autosomal dominant inherited myoclonus-dystonia (M-D). Defects in other sarcoglycans; alpha-, beta-, gamma-, and delta can cause autosomal recessive inherited limb girdle muscular dystrophies. epsilon- and alpha-sarcoglycans are very homologous and may substitute for one-another in different tissues. We therefore investigated whether mutations in SGCE also cause abnormalities of skeletal and myocardial muscle. Six patients with clinically and genetically verified M-D and no signs of limb-girdle muscular dystrophy were included. Skeletal muscle biopsies were obtained from all patients, and endomyocardial muscle biopsy from one of the patients. Morphological and immunohistological investigations were performed and compared with controls. Histological and immunohistological investigations of muscle and clinical assessment of muscle strength and mass showed no difference between M-D patients and controls. Our findings indicate that patients with M-D have no signs or symptoms of muscle disease. This suggests a different role of the sarcoglycan complex epsilonbetagammadelta versus alphabetagammadelta complex in humans, as earlier suggested in rodents.
Collapse
Affiliation(s)
- L E Hjermind
- Department of Medical Genetics, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sharma P, Tran T, Stelmack GL, McNeill K, Gosens R, Mutawe MM, Unruh H, Gerthoffer WT, Halayko AJ. Expression of the dystrophin-glycoprotein complex is a marker for human airway smooth muscle phenotype maturation. Am J Physiol Lung Cell Mol Physiol 2007; 294:L57-68. [PMID: 17993586 DOI: 10.1152/ajplung.00378.2007] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Airway smooth muscle (ASM) cells may contribute to asthma pathogenesis through their capacity to switch between a synthetic/proliferative and a contractile phenotype. The multimeric dystrophin-glycoprotein complex (DGC) spans the sarcolemma, linking the actin cytoskeleton and extracellular matrix. The DGC is expressed in smooth muscle tissue, but its functional role is not fully established. We tested whether contractile phenotype maturation of human ASM is associated with accumulation of DGC proteins. We compared subconfluent, serum-fed cultures and confluent cultures subjected to serum deprivation, which express a contractile phenotype. Western blotting confirmed that beta-dystroglycan, beta-, delta-, and epsilon-sarcoglycan, and dystrophin abundance increased six- to eightfold in association with smooth muscle myosin heavy chain (smMHC) and calponin accumulation during 4-day serum deprivation. Immunocytochemistry showed that the accumulation of DGC subunits was specifically localized to a subset of cells that exhibit robust staining for smMHC. Laminin competing peptide (YIGSR, 1 microM) and phosphatidylinositol 3-kinase (PI3K) inhibitors (20 microM LY-294002 or 100 nM wortmannin) abrogated the accumulation of smMHC, calponin, and DGC proteins. These studies demonstrate that the accumulation of DGC is an integral feature for phenotype maturation of human ASM cells. This provides a strong rationale for future studies investigating the role of the DGC in ASM smooth muscle physiology in health and disease.
Collapse
Affiliation(s)
- Pawan Sharma
- Department of Physiology, Section of Respiratory Disease, Univ. of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Anastasi G, Cutroneo G, Sidoti A, Rinaldi C, Bruschetta D, Rizzo G, D'Angelo R, Tarone G, Amato A, Favaloro A. Sarcoglycan subcomplex expression in normal human smooth muscle. J Histochem Cytochem 2007; 55:831-43. [PMID: 17438352 DOI: 10.1369/jhc.6a7145.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The sarcoglycan complex (SGC) is a multimember transmembrane complex interacting with other members of the dystrophin-glycoprotein complex (DGC) to provide a mechanosignaling connection from the cytoskeleton to the extracellular matrix. The SGC consists of four proteins (alpha, beta, gamma, and delta). A fifth sarcoglycan subunit, epsilon-sarcoglycan, shows a wider tissue distribution. Recently, a novel sarcoglycan, the zeta-sarcoglycan, has been identified. All reports about the structure of SGC showed a common assumption of a tetrameric arrangement of sarcoglycans. Addressing this issue, our immunofluorescence and molecular results showed, for the first time, that all sarcoglycans are always detectable in all observed samples. Therefore, one intriguing possibility is the existence of a pentameric or hexameric complex considering zeta-sarcoglycan of SGC, which could present a higher or lower expression of a single sarcoglycan in conformity with muscle type--skeletal, cardiac, or smooth--or also in conformity with the origin of smooth muscle.
Collapse
Affiliation(s)
- Giuseppe Anastasi
- Department of Biomorphology and Biotechnologies, University of Messina, Via Consolare Valeria, 1 IT-98125, Messina, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Milner DJ, Kaufman SJ. Alpha7beta1 integrin does not alleviate disease in a mouse model of limb girdle muscular dystrophy type 2F. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:609-19. [PMID: 17255329 PMCID: PMC1851849 DOI: 10.2353/ajpath.2007.060686] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transgenic expression of the alpha7beta1 integrin in the dystrophic mdx/utr-/- mouse decreases development of muscular dystrophy and enhances longevity. To explore the possibility that elevating alpha7beta1 integrin expression could also ameliorate different forms of muscular dystrophy, we used transgenic technology to enhance integrin expression in mice lacking delta-sarcoglycan (delta sgc), a mouse model for human limb girdle muscular dystrophy type 2F. Unlike alpha7 transgenic mdx/utr-/- mice, enhanced alpha7beta1 integrin expression in the delta sgc-null mouse did not alleviate muscular dystrophy in these animals. Expression of the transgene in the delta sgc-null did not alleviate dystrophic histopathology, nor did it decrease cardiomyopathy or restore exercise tolerance. One hallmark of integrin-mediated alleviation of muscular dystrophy in the mdx/utr-/- background is the restoration of myotendinous junction integrity. As assessed by atomic force microscopy, myotendinous junctions from normal and delta sgc-null mice were indistinguishable, thus suggesting the important influence of myotendinous junction integrity on the severity of muscular dystrophy and providing a possible explanation for the inability of enhanced integrin expression to alleviate dystrophy in the delta sgc-null mouse. These results suggest that distinct mechanisms underlie the development of the diseases that arise from deficiencies in dystrophin and sarcoglycan.
Collapse
Affiliation(s)
- Derek J Milner
- Department of Cell and Developmental Biology, University of Illinois, B107 Chemical and Life Sciences Laboratory, 601 South Goodwin Ave., Urbana, IL 61801, USA
| | | |
Collapse
|
17
|
Delgado-Olguín P, Recillas-Targa F, Rosas-Vargas H, Salamanca F, Coral-Vázquez RM. Partial characterization of the mouse alpha-sarcoglycan promoter and its responsiveness to MyoD. ACTA ACUST UNITED AC 2006; 1759:240-6. [PMID: 16797743 DOI: 10.1016/j.bbaexp.2006.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Revised: 04/20/2006] [Accepted: 04/26/2006] [Indexed: 10/24/2022]
Abstract
The mouse alpha-sarcoglycan gene is expressed in muscle cells during differentiation, but its transcriptional regulation is not understood. We have characterized the promoter region of the mouse alpha-sarcoglycan gene. This region is composed of positive and negative regulatory elements that respond to the myogenic differentiation environment. Accordingly, MyoD transactivates the alpha-sarcoglycan full-length and the proximal promoter. Chromatin immunoprecipitation assays revealed that MyoD, TFIID, and TFIIB interact with the distal promoter in C2C12 myoblasts, a stage at which the alpha-SG promoter appears to drive basal activity. In myotubes, such factors are located concomitantly at the distal promoter and at a DNA region around the proximal promoter. In agreement with these results, TFIID and TFIIB co-immunoprecipitate with MyoD. We conclude that the alpha-SG promoter is activated by MyoD, which interacts with TFIID and TFIIB in a protein complex differentially located at the distal promoter and around the proximal promoter during myogenic cell differentiation.
Collapse
Affiliation(s)
- Paul Delgado-Olguín
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | | | | | | |
Collapse
|
18
|
Halayko AJ, Stelmack GL. The association of caveolae, actin, and the dystrophin-glycoprotein complex: a role in smooth muscle phenotype and function? Can J Physiol Pharmacol 2006; 83:877-91. [PMID: 16333360 DOI: 10.1139/y05-107] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Smooth muscle cells exhibit phenotypic and mechanical plasticity. During maturation, signalling pathways controlling actin dynamics modulate contractile apparatus-associated gene transcription and contractile apparatus remodelling resulting from length change. Differentiated myocytes accumulate abundant caveolae that evolve from the structural association of lipid rafts with caveolin-1, a protein with domains that confer unique functional properties. Caveolae and caveolin-1 modulate and participate in receptor-mediated signalling, and thus contribute to functional diversity of phenotypically similar myocytes. In mature smooth muscle, caveolae are partitioned into discrete linear domains aligned with structural proteins that tether actin to the extracellular matrix. Caveolin-1 binds with beta-dystroglycan, a subunit of the dystrophin glycoprotein complex (DGC), and with filamin, an actin binding protein that organizes cortical actin, to which integrins and focal adhesion complexes are anchored. The DGC is linked to the actin cytoskeleton by a dystrophin subunit and is a receptor for extracellular laminin. Thus, caveolae and caveolin-associated signalling proteins and receptors are linked via structural proteins to a dynamic filamentous actin network. Despite development of transgenic models to investigate caveolins and membrane-associated actin-linking proteins in skeletal and cardiac muscle function, only superficial understanding of this association in smooth muscle phenotype and function has emerged.
Collapse
Affiliation(s)
- Andrew J Halayko
- Department of Physiology, University of Manitoba, Winnipeg, Canada.
| | | |
Collapse
|
19
|
Macarak EJ, Schulz J, Zderic SA, Sado Y, Ninomiya Y, Polyak E, Chacko S, Howard PS. Smooth muscle trans-membrane sarcoglycan complex in partial bladder outlet obstruction. Histochem Cell Biol 2006; 126:71-82. [PMID: 16435124 DOI: 10.1007/s00418-005-0135-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2005] [Indexed: 10/25/2022]
Abstract
The urinary bladder experiences both distension and contraction as a part of the normal filling and emptying cycle. To empty properly, tension generated intracellularly in a smooth muscle cell must be smoothly and efficiently transferred across its sarcolemma to the basement membrane, which mediates its binding to both the extracellular matrix and to other cells. As a consequence of urethral obstruction, the bladder cannot generate appropriate force to contract the organ, thereby leading to inefficient emptying and associated sequelae. In this study, an animal model of urethral obstruction was utilized to study the membrane-associated structures that transfer tension across the sarcolemma of bladder smooth muscle cells. Immunohistochemical localization of key components of the smooth muscle tension transfer apparatus (TTA) was performed utilizing specific antibodies against:(1) the alpha-chains of type IV collagen, a basement membrane component, and (2) beta-sarcoglycan, an integral membrane protein that is a participant in the physical linkage between the cytoskeleton and the basement membrane. We demonstrate, in obstructed animals, that there is a pronounced disruption of the TTA with a physical displacement of these two components that can be demonstrated at the level of the light microscope using scanning confocal microscopy. Electron microscopy further demonstrates significant increases in the size of the junctional plaques between smooth muscle cells.
Collapse
Affiliation(s)
- Edward J Macarak
- Department of Anatomy and Cell Biology School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, PA 19104-6030, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Regulation of coronary vascular tone is critical for proper perfusion and function of the myocardium. Many disease processes result in compromised regulation of coronary vascular tone and impaired myocardial perfusion. A common result of coronary vascular dysfunction is the development of areas of replacement fibrosis within the myocardium and surrounding the vasculature. Both intravascular processes, such as coronary atherosclerosis and endothelial dysfunction, and extravascular processes, including compromised myocardial metabolism, hormone excesses, and altered local signaling, may result in coronary vascular dysregulation. Coronary occlusion events, in turn, lead to myocardial damage and the activation of inflammatory cells and fibroblasts. The role of fibroblasts in regulating myocardial fibrosis and the contribution of myofibroblasts, cells that have limited contractile potential while retaining many of the extracellular matrix regulating processes of the fibroblast, may also contribute to the development of myocardial disease. In this review we examine the recent literature on myocardial fibrosis and myofibroblast activity, highlighting the effects of several classes of cardiovascular agents on the remodeling process.
Collapse
Affiliation(s)
- Matthew T Wheeler
- Section of Cardiology, Department of Medicine, The University of Chicago, 5841 South Maryland Avenue, MC 6088, Chicago, IL 60637, USA
| | | |
Collapse
|
21
|
Liton PB, Liu X, Stamer WD, Challa P, Epstein DL, Gonzalez P. Specific targeting of gene expression to a subset of human trabecular meshwork cells using the chitinase 3-like 1 promoter. Invest Ophthalmol Vis Sci 2005; 46:183-90. [PMID: 15623772 PMCID: PMC3152459 DOI: 10.1167/iovs.04-0330] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To compare the gene expression profile of trabecular meshwork (TM) and Schlemm's canal (SC) primary cultures and to identify promoters for targeting gene expression to specific cells in the outflow pathway. METHODS The differential gene expression profile of four human TM and three SC primary cultures was analyzed by gene microarrays (Affymetrix, Santa Clara, CA) and confirmed by quantitative real-time PCR. Based on the results, a recombinant adenovirus was constructed with the expression of the reporter gene LacZ driven by the 5' promoter region of the chitinase 3-like 1 (Ch3L1) gene (AdCh3L1-LacZ). The expression of the Ch3L1 promoter was analyzed in human TM and SC cells and in human perfused anterior segments infected with AdCh3L1-LacZ. RESULTS gamma-Sarcoglycan, fibulin-2, and collagen XV were identified as the genes more highly expressed in SC than in TM cells. Ch3L1 showed the highest levels of differential expression in TM versus SC cells. Expression analysis of the Ch3L1 promoter demonstrated specific expression in a subset of the TM cells in cell culture and in perfused anterior segments. CONCLUSIONS Comparative analysis of gene expression between SC and TM primary cultures identified several genes with promoters potentially capable of targeting gene expression to specific cells within the outflow pathway. Results with the Ch3L1 promoter indicated that two different cell subtypes may be present in the TM. This study provides a new potential tool to investigate the role of these different cell types in both normal and pathophysiological function of the outflow pathway, with implications for possible future glaucoma gene therapy.
Collapse
Affiliation(s)
- Paloma B. Liton
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - Xialin Liu
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - W. Daniel Stamer
- Department of Ophthalmology, University of Arizona, Tucson, Arizona
| | - Pratap Challa
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - David L. Epstein
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | - Pedro Gonzalez
- Department of Ophthalmology, Duke University, Durham, North Carolina
| |
Collapse
|
22
|
Ramírez-Sánchez I, Rosas-Vargas H, Ceballos-Reyes G, Salamanca F, Coral-Vázquez RM. Expression Analysis of the SG-SSPN Complex in Smooth Muscle and Endothelial Cells of Human Umbilical Cord Vessels. J Vasc Res 2005; 42:1-7. [PMID: 15583476 DOI: 10.1159/000082528] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Accepted: 09/27/2004] [Indexed: 11/19/2022] Open
Abstract
Recently, participation of the sarcoglycan (SG)-sarcospan (SSPN) complex in the development of cardiomyopathy in patients with limb-girdle muscular dystrophy has been shown, and presence of the complex in smooth muscle may be important for the contraction/dilation process of vessels. However, there are few studies determining the SG-SSPN complex in vascular smooth muscle and endothelial cells of vessels. In this study, we analyzed by reverse transcriptase-polymerase chain reaction and immunofluorescence the expression of different components of the complex in vein/artery smooth muscle and endothelial cells of the human umbilical cord. By RNA analysis, we observed expression of alpha-, beta-, gamma-, delta-, epsilon-SG, and SSPN in smooth muscle cells. In endothelial cells, RNA expression was restricted to beta-, delta-, epsilon-SG, and SSPN. At protein level, we observed in smooth muscle the presence of beta-, delta-, epsilon-SG, and SSPN. In endothelial cells, immunostaining only evidenced the presence of epsilon-SG and SSPN. However, colocalization of SGs and SSPN with dystrophin and utrophin was noted. These results, interestingly, suggest that the SG-SSPN complex may either form with dystrophin or utrophin in smooth muscle cells, and with utrophin in endothelial cells. Additionally, we also observed in some smooth muscle regions the colocalization of the SG-SSPN complex with caveolin, with colocalization being more pronounced between epsilon-SG-SSPN and caveolin in endothelial cells.
Collapse
Affiliation(s)
- I Ramírez-Sánchez
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI-IMSS, Mexico, D.F., Mexico
| | | | | | | | | |
Collapse
|
23
|
Abstract
In this review, we draw attention to the multiple mechanisms responsible for the pathogenesis of cardiomyopathies in patients with muscular dystrophies. More than one single mechanism is likely to be involved in the development of skeletal and cardiac muscle pathology even when there is a single protein defect. The best example is dystrophin deficiency, in which increased sarcolemmal permeability following eccentric exercise, reduced force generation, and abnormal signaling are all likely to contribute to the progressive muscle damage observed. In other conditions, such as the sarcoglycanopathies, a protein deficiency both in the striated cardiomyocte and the vascular smooth muscle appears to play a significant role. An entirely different mechanism of disease is likely in defects of nuclear envelope proteins, although the precise pathogenesis of this group of conditions is still not clear. Differences between the organization of skeletal and cardiac muscle protein complex are also only starting to emerge and will very likely be the focus of future research.
Collapse
Affiliation(s)
- Fiona C Goodwin
- Dubowitz Neuromuscular Centre, Imperial College London, Hammersmith Hospital Campus, UK
| | | |
Collapse
|
24
|
Ozawa E, Mizuno Y, Hagiwara Y, Sasaoka T, Yoshida M. Molecular and cell biology of the sarcoglycan complex. Muscle Nerve 2005; 32:563-76. [PMID: 15937871 DOI: 10.1002/mus.20349] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The original sarcoglycan (SG) complex has four subunits and comprises a subcomplex of the dystrophin-dystrophin-associated protein complex. Each SG gene has been shown to be responsible for limb-girdle muscular dystrophy, called sarcoglycanopathy (SGP). In this review, we detail the characteristics of the SG subunits, and the mechanism of the formation of the SG complex and various molecules associated with this complex. We discuss the molecular mechanisms of SGP based on studies mostly using SGP animal models. In addition, we describe other SG molecules, epsilon- and zeta-SGs, with special reference to their expression and roles in vascular smooth muscle, which are currently in dispute. We further consider the maternally imprinted nature of the epsilon-SG gene. Finally, we stress that the SG complex cannot work by itself and works in a larger complex system, called the transverse fixation system, which forms an array of molecules responsible for various muscular dystrophies.
Collapse
Affiliation(s)
- Eijiro Ozawa
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Ogawahigashi-cho, Kodaira, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
25
|
Wheeler MT, Korcarz CE, Collins KA, Lapidos KA, Hack AA, Lyons MR, Zarnegar S, Earley JU, Lang RM, McNally EM. Secondary coronary artery vasospasm promotes cardiomyopathy progression. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1063-71. [PMID: 14982859 PMCID: PMC1614719 DOI: 10.1016/s0002-9440(10)63193-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Genetic defects in the plasma membrane-associated sarcoglycan complex produce cardiomyopathy characterized by focal degeneration. The infarct-like pattern of cardiac degeneration has led to the hypothesis that coronary artery vasospasm underlies cardiomyopathy in this disorder. We evaluated the coronary vasculature of gamma-sarcoglycan mutant mice and found microvascular filling defects consistent with arterial vasospasm. However, the vascular smooth muscle sarcoglycan complex was intact in the coronary arteries of gamma-sarcoglycan hearts with perturbation of the sarcoglycan complex only within the adjacent myocytes. Thus, in this model, coronary artery vasospasm derives from a vascular smooth muscle-cell extrinsic process. To reduce this secondary vasospasm, we treated gamma-sarcoglycan-deficient mice with the calcium channel antagonist verapamil. Verapamil treatment eliminated evidence of vasospasm and ameliorated histological and functional evidence of cardiomyopathic progression. Echocardiography of verapamil-treated, gamma-sarcoglycan-null mice showed an improvement in left ventricular fractional shortening (44.3 +/- 13.3% treated versus 37.4 +/- 15.3% untreated), maximal velocity at the aortic outflow tract (114.9 +/- 27.9 cm/second versus 92.8 +/- 22.7 cm/second), and cardiac index (1.06 +/- 0.30 ml/minute/g versus 0.67 +/- 0.16 ml/minute/g, P < 0.05). These data indicate that secondary vasospasm contributes to the development of cardiomyopathy and is an important therapeutic target to limit cardiomyopathy progression.
Collapse
Affiliation(s)
- Matthew T Wheeler
- Department of Molecular Genetics and Cell Biology, Section of Cardiology, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Laval SH, Bushby KMD. Limb-girdle muscular dystrophies - from genetics to molecular pathology. Neuropathol Appl Neurobiol 2004; 30:91-105. [PMID: 15043707 DOI: 10.1111/j.1365-2990.2004.00555.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The limb-girdle muscular dystrophies are a diverse group of muscle-wasting disorders characteristically affecting the large muscles of the pelvic and shoulder girdles. Molecular genetic analyses have demonstrated causative mutations in the genes encoding a disparate collection of proteins involved in all aspects of muscle cell biology. Muscular dystrophy includes a spectrum of disorders caused by loss of the linkage between the extracellular matrix and the actin cytoskeleton. Within this are the forms of limb-girdle muscular dystrophy caused by deficiencies of the sarcoglycan complex and by aberrant glycosylation of alpha-dystroglycan caused by mutations in the fukutin-related protein gene. However, other forms of this disease have distinct pathophysiological mechanisms. For example, deficiency of dysferlin disrupts sarcolemmal membrane repair, whilst loss of calpain-3 may exert its pathological influence either by perturbation of the IkappaBalpha/NF-kappaB pathway, or through calpain-dependent cytoskeletal remodelling. Caveolin-3 is implicated in numerous cell-signalling pathways and involved in the biogenesis of the T-tubule system. Alterations in the nuclear lamina caused by mutations in laminA/C, sarcomeric changes in titin, telethonin or myotilin at the Z-disc, and subtle changes in the extracellular matrix proteins laminin-alpha2 or collagen VI can all lead to a limb-girdle muscular dystrophy phenotype, although the specific pathological mechanisms remain obscure. Differential diagnosis of these disorders requires the careful application of a broad range of disciplines: clinical assessment, immunohistochemistry and immunoblotting using a panel of antibodies and extensive molecular genetic analyses.
Collapse
Affiliation(s)
- S H Laval
- Institute of Human Genetics, International Centre for Life, Newcastle-upon-Tyne, UK
| | | |
Collapse
|
27
|
Wheeler MT, Allikian MJ, Heydemann A, McNally EM. The sarcoglycan complex in striated and vascular smooth muscle. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:389-97. [PMID: 12858564 DOI: 10.1101/sqb.2002.67.389] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- M T Wheeler
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
28
|
Wicklund MP, Mendell JR. The limb girdle muscular dystrophies: our ever-expanding knowledge. J Clin Neuromuscul Dis 2003; 5:12-28. [PMID: 19078718 DOI: 10.1097/00131402-200309000-00003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The limb girdle muscular dystrophies (LGMDs) represent a genetically diverse group of disorders. Currently, chromosomal loci are known for at least 5 autosomal-dominant and 10 autosomal-recessive subgroups. In 13 of these, recognized genes and protein products generate an assortment of phenotypes, some unique and many overlapping. In some disorders, novel clinical features are sufficiently distinct so as to proffer clues to the diagnosis of a specific LGMD subtype. An armamentarium of laboratory tools is required to confirm specific subtypes of LGMD. These might only be available in neuromuscular centers specializing in this form of dystrophy. Currently, supportive therapy is the predominant means of treatment, but further understanding of unique pathogenic mechanisms holds promise for the future.
Collapse
Affiliation(s)
- Matthew P Wicklund
- From the Department of Neurology, Wilford Hall Medical Center, Lackland Air Force Base, Texas (Dr Wicklund); and the Department of Neurology, The Ohio State University, Columbus, Ohio (Dr Mendell)
| | | |
Collapse
|
29
|
Durbeej M, Sawatzki SM, Barresi R, Schmainda KM, Allamand V, Michele DE, Campbell KP. Gene transfer establishes primacy of striated vs. smooth muscle sarcoglycan complex in limb-girdle muscular dystrophy. Proc Natl Acad Sci U S A 2003; 100:8910-5. [PMID: 12851463 PMCID: PMC166412 DOI: 10.1073/pnas.1537554100] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Limb-girdle muscular dystrophy types 2E and F are characterized by skeletal muscle weakness and often cardiomyopathy and are due to mutations in the genes encoding beta- and delta-sarcoglycan. We previously demonstrated that loss of sarcoglycans in smooth muscle leads to constrictions of the microvasculature that contributes to the cardiac phenotype. It is unclear how vasculature abnormalities affect skeletal muscle. We injected recombinant beta- or delta-sarcoglycan adenoviruses into skeletal muscles of corresponding null mice. We hypothesized that the adenoviruses would not transduce vascular smooth muscle, and we would only target skeletal muscle. Indeed, sustained expression of intact sarcoglycan-sarcospan complex was noted at the sarcolemma, neuromuscular junction, myotendinous junction, and in peripheral nerve, but not in vascular smooth muscle. Gene transfer of the corresponding deleted sarcoglycan gene preserved sarcolemmal integrity, prevented pathological dystrophy and hypertrophy, and protected against exercised-induced damage. We conclude that vascular dysfunction is not a primary cause of beta- and delta-sarcoglycan-deficient muscular dystrophy. In addition, we show successful functional rescue of entire muscles after adenovirus-mediated gene delivery. Thus, virus-mediated gene transfer of sarcoglycans to skeletal muscle in combination with pharmacological prevention of cardiomyopathy constitute promising therapeutic strategies for limb-girdle muscular dystrophies.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cytoskeletal Proteins/chemistry
- Cytoskeletal Proteins/deficiency
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Dystroglycans
- Gene Transfer Techniques
- Genetic Therapy
- Macromolecular Substances
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/injuries
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Smooth/metabolism
- Muscle, Smooth/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscular Dystrophies/genetics
- Muscular Dystrophies/metabolism
- Muscular Dystrophies/pathology
- Muscular Dystrophies/therapy
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/therapy
- Neuromuscular Junction/metabolism
- Physical Exertion
- Sarcoglycans
- Sarcolemma/metabolism
- Schwann Cells/metabolism
Collapse
Affiliation(s)
- Madeleine Durbeej
- Howard Hughes Medical Institute, Department of Physiology and Biophysics, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242-1101, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Durbeej M, Campbell KP. Muscular dystrophies involving the dystrophin-glycoprotein complex: an overview of current mouse models. Curr Opin Genet Dev 2002; 12:349-61. [PMID: 12076680 DOI: 10.1016/s0959-437x(02)00309-x] [Citation(s) in RCA: 326] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The dystrophin-glycoprotein complex (DGC) is a multisubunit complex that connects the cytoskeleton of a muscle fiber to its surrounding extracellular matrix. Mutations in the DGC disrupt the complex and lead to muscular dystrophy. There are a few naturally occurring animal models of DGC-associated muscular dystrophy (e.g. the dystrophin-deficient mdx mouse, dystrophic golden retriever dog, HFMD cat and the delta-sarcoglycan-deficient BIO 14.6 cardiomyopathic hamster) that share common genetic protein abnormalities similar to those of the human disease. However, the naturally occurring animal models only partially resemble human disease. In addition, no naturally occurring mouse models associated with loss of other DGC components are available. This has encouraged the generation of genetically engineered mouse models for DGC-linked muscular dystrophy. Not only have analyses of these mice led to a significant improvement in our understanding of the pathogenetic mechanisms for the development of muscular dystrophy, but they will also be immensely valuable tools for the development of novel therapeutic approaches for these incapacitating diseases.
Collapse
Affiliation(s)
- Madeleine Durbeej
- Howard Hughes Medical Institute, Department of Physiology and Biophysics, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
31
|
Bönnemann CG, Finkel RS. Sarcolemmal proteins and the spectrum of limb-girdle muscular dystrophies. Semin Pediatr Neurol 2002; 9:81-99. [PMID: 12139001 DOI: 10.1053/spen.2002.33795] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Proteins of the sarcolemma are of crucial importance for the pathogenesis of muscular dystrophies. This update focuses on the dystrophin-associated proteins including the dystroglycan and sarcoglycan complexes, caveolin-3, dysferlin, and the extracellular matrix component collagen type VI. The molecular findings are correlated with some of the clinical phenotypes that are part of the limb-girdle muscular dystrophy spectrum, including fukutin-related proteinopathy (LGMD 21), the sarcoglycanopathies (LGMD 2C-F), caveolinopathy (LGMD 1C), dysferlinopathy (LGMD 2B), and finally Bethlem myopathy. Although recent progress has been tremendous, much remains to be learned about the pathophysiological consequences caused by a deficiency of any one of these components.
Collapse
Affiliation(s)
- Carsten G Bönnemann
- Division of Neurology, The Children's Hospital of Philadelphia, PA 19104, USA
| | | |
Collapse
|
32
|
Conrad K, Deppe A, Neumann S, Breen M, Quignon P, André C, Brenig B, Leeb T. Characterization and chromosome assignment of the canine gamma-sarcoglycan gene (SGCG) to CFA 25q21-->q23. Cytogenet Genome Res 2002; 94:186-9. [PMID: 11856878 DOI: 10.1159/000048813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mutations in the gene for gamma-sarcoglycan (SGCG) located on HSA 13q12 are responsible for limb girdle muscular dystrophy (LGMD2C) in human. Here we report the cloning of the canine SGCG gene together with its genomic structure and several intragenic polymorphisms. The coding part of the canine SGCG contains seven exons spanning at least 70 kb of genomic DNA. The chromosome assignment of the canine SGCG gene to CFA 25q21-->q23 confirms that the canine syntenic group 10 corresponds to CFA 25 and also supports the findings of human-canine reciprocal chromosome painting.
Collapse
Affiliation(s)
- K Conrad
- Institute of Veterinary Medicine, University of Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Blake DJ, Weir A, Newey SE, Davies KE. Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev 2002; 82:291-329. [PMID: 11917091 DOI: 10.1152/physrev.00028.2001] [Citation(s) in RCA: 842] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The X-linked muscle-wasting disease Duchenne muscular dystrophy is caused by mutations in the gene encoding dystrophin. There is currently no effective treatment for the disease; however, the complex molecular pathology of this disorder is now being unravelled. Dystrophin is located at the muscle sarcolemma in a membrane-spanning protein complex that connects the cytoskeleton to the basal lamina. Mutations in many components of the dystrophin protein complex cause other forms of autosomally inherited muscular dystrophy, indicating the importance of this complex in normal muscle function. Although the precise function of dystrophin is unknown, the lack of protein causes membrane destabilization and the activation of multiple pathophysiological processes, many of which converge on alterations in intracellular calcium handling. Dystrophin is also the prototype of a family of dystrophin-related proteins, many of which are found in muscle. This family includes utrophin and alpha-dystrobrevin, which are involved in the maintenance of the neuromuscular junction architecture and in muscle homeostasis. New insights into the pathophysiology of dystrophic muscle, the identification of compensating proteins, and the discovery of new binding partners are paving the way for novel therapeutic strategies to treat this fatal muscle disease. This review discusses the role of the dystrophin complex and protein family in muscle and describes the physiological processes that are affected in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Derek J Blake
- Medical Research Council, Functional Genetics Unit, Department of Human Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
34
|
Rando TA. The dystrophin-glycoprotein complex, cellular signaling, and the regulation of cell survival in the muscular dystrophies. Muscle Nerve 2001; 24:1575-94. [PMID: 11745966 DOI: 10.1002/mus.1192] [Citation(s) in RCA: 277] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mutations of different components of the dystrophin-glycoprotein complex (DGC) cause muscular dystrophies that vary in terms of severity, age of onset, and selective involvement of muscle groups. Although the primary pathogenetic processes in the muscular dystrophies have clearly been identified as apoptotic and necrotic muscle cell death, the pathogenetic mechanisms that lead to cell death remain to be determined. Studies of components of the DGC in muscle and in nonmuscle tissues have revealed that the DGC is undoubtedly a multifunctional complex and a highly dynamic structure, in contrast to the unidimensional concept of the DGC as a mechanical component in the cell. Analysis of the DGC reveals compelling analogies to two other membrane-associated protein complexes, namely integrins and caveolins. Each of these complexes mediates signal transduction cascades in the cell, and disruption of each complex causes muscular dystrophies. The signal transduction cascades associated with the DGC, like those associated with integrins and caveolins, play important roles in cell survival signaling, cellular defense mechanisms, and regulation of the balance between cell survival and cell death. This review focuses on the functional components of the DGC, highlighting the evidence of their participation in cellular signaling processes important for cell survival. Elucidating the link between these functional components and the pathogenetic processes leading to cell death is the foremost challenge to understanding the mechanisms of disease expression in the muscular dystrophies due to defects in the DGC.
Collapse
Affiliation(s)
- T A Rando
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Room A-343, Stanford, California 94305-5235, USA.
| |
Collapse
|
35
|
Cohn RD, Durbeej M, Moore SA, Coral-Vazquez R, Prouty S, Campbell KP. Prevention of cardiomyopathy in mouse models lacking the smooth muscle sarcoglycan-sarcospan complex. J Clin Invest 2001; 107:R1-7. [PMID: 11160141 PMCID: PMC199179 DOI: 10.1172/jci11642] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cardiomyopathy is a multifactorial disease, and the dystrophin-glycoprotein complex has been implicated in the pathogenesis of both hereditary and acquired forms of the disease. Using mouse models of cardiomyopathy made by ablating genes for components of the sarcoglycan complex, we show that long-term treatment with verapamil, a calcium channel blocker with vasodilator properties, can alleviate the severe cardiomyopathic phenotype, restoring normal serum levels for cardiac troponin I and normal cardiac muscle morphology. Interruption of verapamil treatment leads again to vascular dysfunction and acute myocardial necrosis, indicating that predilection for cardiomyopathy is a continuing process. In contrast, verapamil did not prevent cardiac muscle pathology in dystrophin-deficient mdx mice, which neither show a disruption of the sarcoglycan complex in vascular smooth muscle nor vascular dysfunction. Hence, our data strongly suggest that pharmacological intervention with verapamil merits investigation as a potential therapeutic option not only for patients with sarcoglycan mutations, but also for patients with idiopathic cardiomyopathy associated with myocardial ischemia not related to atherosclerotic coronary artery disease.
Collapse
Affiliation(s)
- R D Cohn
- Howard Hughes Medical Institute, Department of Physiology and Biophysics, Department of Neurology, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | |
Collapse
|