1
|
Doody A, Alfano L, Diaz-Manera J, Lowes L, Mozaffar T, Mathews KD, Weihl CC, Wicklund M, Hung M, Statland J, Johnson NE. Defining clinical endpoints in limb girdle muscular dystrophy: a GRASP-LGMD study. BMC Neurol 2024; 24:96. [PMID: 38491364 PMCID: PMC10941356 DOI: 10.1186/s12883-024-03588-1] [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: 09/19/2023] [Accepted: 02/26/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND The Limb Girdle Muscular Dystrophies (LGMDs) are characterized by progressive weakness of the shoulder and hip girdle muscles as a result of over 30 different genetic mutations. This study is designed to develop clinical outcome assessments across the group of disorders. METHODS/DESIGN The primary goal of this study is to evaluate the utility of a set of outcome measures on a wide range of LGMD phenotypes and ability levels to determine if it would be possible to use similar outcomes between individuals with different phenotypes. We will perform a multi-center, 12-month study of 188 LGMD patients within the established Genetic Resolution and Assessments Solving Phenotypes in LGMD (GRASP-LGMD) Research Consortium, which is comprised of 11 sites in the United States and 2 sites in Europe. Enrolled patients will be clinically affected and have mutations in CAPN3 (LGMDR1), ANO5 (LGMDR12), DYSF (LGMDR2), DNAJB6 (LGMDD1), SGCA (LGMDR3), SGCB (LGMDR4), SGCD (LGMDR6), or SGCG (LGMDR5, or FKRP-related (LGMDR9). DISCUSSION To the best of our knowledge, this will be the largest consortium organized to prospectively validate clinical outcome assessments (COAs) in LGMD at its completion. These assessments will help clinical trial readiness by identifying reliable, valid, and responsive outcome measures as well as providing data driven clinical trial decision making for future clinical trials on therapeutic agents for LGMD. The results of this study will permit more efficient clinical trial design. All relevant data will be made available for investigators or companies involved in LGMD therapeutic development upon conclusion of this study as applicable. TRIAL REGISTRATION Clinicaltrials.gov NCT03981289; Date of registration: 6/10/2019.
Collapse
Affiliation(s)
- Amy Doody
- Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Linda Lowes
- Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | | | | | - Man Hung
- Roseman University, Salt Lake City, UT, USA
| | | | | |
Collapse
|
2
|
Tamura JI, Tamura T, Hoshino S, Imae R, Kato R, Yokono M, Nagase M, Ohno S, Manabe N, Yamaguchi Y, Manya H, Endo T. Chemical and Chemo-Enzymatic Syntheses of Glycans Containing Ribitol Phosphate Scaffolding of Matriglycan. ACS Chem Biol 2022; 17:1513-1523. [PMID: 35670527 DOI: 10.1021/acschembio.2c00181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ribitol phosphate modifications to the core M3 O-mannosyl glycan are important for the functional maturation of α-dystroglycan. Three sequentially extended partial structures of the core M3 O-mannosyl glycan including a tandem ribitol phosphate were regio- and stereo-selectively synthesized: Rbo5P-3GalNAcβ, Rbo5P-1Rbo5P-3GalNAcβ, and Xylβ1-4Rbo5P-1Rbo5P-3GalNAcβ (Rbo5P, d-ribitol-5-phosphate; GalNAc, N-acetyl-d-galactosamine; Xyl, d-xylose). Rbo5P-3GalNAcβ with p-nitrophenyl at the aglycon part served as a substrate for ribitol phosphate transferase (FKRP, fukutin-related protein), and its product was glycosylated by the actions of a series of glycosyltransferases, namely, ribitol xylosyltransferase 1 (RXYLT1), β1,4-glucuronyltransferase 1 (B4GAT1), and like-acetyl-glucosaminyltransferase (LARGE). Rbo5P-3GalNAcβ equipped with an alkyne-type aglycon was also active for FKRP. The molecular information obtained on FKRP suggests that Rbo5P-3GalNAcβ derivatives are the minimal units required as the acceptor glycan for Rbo5P transfer and may serve as a precursor for the elongation of the core M3 O-mannosyl glycan.
Collapse
Affiliation(s)
- Jun-Ichi Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan
| | - Takahiro Tamura
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori 680-8553, Japan
| | - Shunsuke Hoshino
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Rieko Imae
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Ryuichi Kato
- Structural Biology Research Center, Institute of Material Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan
| | - Mizuki Yokono
- Technical Department, Tottori University, Tottori 680-8550, Japan
| | - Mao Nagase
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Noriyoshi Manabe
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
3
|
Yonekawa T, Rauckhorst AJ, El-Hattab S, Cuellar MA, Venzke D, Anderson ME, Okuma H, Pewa AD, Taylor EB, Campbell KP. Large1 gene transfer in older myd mice with severe muscular dystrophy restores muscle function and greatly improves survival. SCIENCE ADVANCES 2022; 8:eabn0379. [PMID: 35613260 PMCID: PMC9132445 DOI: 10.1126/sciadv.abn0379] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/07/2022] [Indexed: 06/15/2023]
Abstract
Muscular dystrophy is a progressive and ultimately lethal neuromuscular disease. Although gene editing and gene transfer hold great promise as therapies when administered before the onset of severe clinical symptoms, it is unclear whether these strategies can restore muscle function and improve survival in the late stages of muscular dystrophy. Largemyd/Largemyd (myd) mice lack expression of like-acetylglucosaminyltransferase-1 (Large1) and exhibit severe muscle pathophysiology, impaired mobility, and a markedly reduced life span. Here, we show that systemic delivery of AAV2/9 CMV Large1 (AAVLarge1) in >34-week-old myd mice with advanced disease restores matriglycan expression on dystroglycan, attenuates skeletal muscle pathophysiology, improves motor and respiratory function, and normalizes systemic metabolism, which collectively and markedly extends survival. Our results in a mouse model of muscular dystrophy demonstrate that skeletal muscle function can be restored, illustrating its remarkable plasticity, and that survival can be greatly improved even after the onset of severe muscle pathophysiology.
Collapse
Affiliation(s)
- Takahiro Yonekawa
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - Adam J. Rauckhorst
- Department of Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center (FOEDRC), and FOEDRC Metabolomics Core Facility, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Sara El-Hattab
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - Marco A. Cuellar
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - David Venzke
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - Mary E. Anderson
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - Hidehiko Okuma
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| | - Alvin D. Pewa
- Department of Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center (FOEDRC), and FOEDRC Metabolomics Core Facility, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Eric B. Taylor
- Department of Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center (FOEDRC), and FOEDRC Metabolomics Core Facility, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Kevin P. Campbell
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, Howard Hughes Medical Institute, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
4
|
Boyd A, Montandon M, Wood AJ, Currie PD. FKRP directed fibronectin glycosylation: A novel mechanism giving insights into muscular dystrophies? Bioessays 2022; 44:e2100270. [PMID: 35229908 DOI: 10.1002/bies.202100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
Abstract
The recently uncovered role of Fukutin-related protein (FKRP) in fibronectin glycosylation has challenged our understanding of the basis of disease pathogenesis in the muscular dystrophies. FKRP is a Golgi-resident glycosyltransferase implicated in a broad spectrum of muscular dystrophy (MD) pathologies that are not fully attributable to the well-described α-Dystroglycan hypoglycosylation. By revealing a new role for FKRP in the glycosylation of fibronectin, a modification critical for the development of the muscle basement membrane (MBM) and its associated muscle linkages, new possibilities for understanding clinical phenotype arise. This modification involves an interaction between FKRP and myosin-10, a protein involved in the Golgi organization and function. These observations suggest a FKRP nexus exists that controls two critical aspects to muscle fibre integrity, both fibre stability at the MBM and its elastic properties. This review explores the new potential disease axis in the context of our current knowledge of muscular dystrophies.
Collapse
Affiliation(s)
- Andrew Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Margo Montandon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
5
|
Kanagawa M. Dystroglycanopathy: From Elucidation of Molecular and Pathological Mechanisms to Development of Treatment Methods. Int J Mol Sci 2021; 22:ijms222313162. [PMID: 34884967 PMCID: PMC8658603 DOI: 10.3390/ijms222313162] [Citation(s) in RCA: 22] [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: 11/28/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 01/13/2023] Open
Abstract
Dystroglycanopathy is a collective term referring to muscular dystrophies with abnormal glycosylation of dystroglycan. At least 18 causative genes of dystroglycanopathy have been identified, and its clinical symptoms are diverse, ranging from severe congenital to adult-onset limb-girdle types. Moreover, some cases are associated with symptoms involving the central nervous system. In the 2010s, the structure of sugar chains involved in the onset of dystroglycanopathy and the functions of its causative gene products began to be identified as if they were filling the missing pieces of a jigsaw puzzle. In parallel with these discoveries, various dystroglycanopathy model mice had been created, which led to the elucidation of its pathological mechanisms. Then, treatment strategies based on the molecular basis of glycosylation began to be proposed after the latter half of the 2010s. This review briefly explains the sugar chain structure of dystroglycan and the functions of the causative gene products of dystroglycanopathy, followed by introducing the pathological mechanisms involved as revealed from analyses of dystroglycanopathy model mice. Finally, potential therapeutic approaches based on the pathological mechanisms involved are discussed.
Collapse
Affiliation(s)
- Motoi Kanagawa
- Department of Cell Biology and Molecular Medicine, Graduate School of Medicine, Ehime University, 454 Shitsukawa, Toon 791-0295, Ehime, Japan
| |
Collapse
|
6
|
Dhoke NR, Kim H, Selvaraj S, Azzag K, Zhou H, Oliveira NAJ, Tungtur S, Ortiz-Cordero C, Kiley J, Lu QL, Bang AG, Perlingeiro RCR. A universal gene correction approach for FKRP-associated dystroglycanopathies to enable autologous cell therapy. Cell Rep 2021; 36:109360. [PMID: 34260922 PMCID: PMC8327854 DOI: 10.1016/j.celrep.2021.109360] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 01/24/2023] Open
Abstract
Mutations in the fukutin-related protein (FKRP) gene result in a broad spectrum of muscular dystrophy (MD) phenotypes, including the severe Walker-Warburg syndrome (WWS). Here, we develop a gene-editing approach that replaces the entire mutant open reading frame with the wild-type sequence to universally correct all FKRP mutations. We apply this approach to correct FKRP mutations in induced pluripotent stem (iPS) cells derived from patients displaying broad clinical severity. Our findings show rescue of functional α-dystroglycan (α-DG) glycosylation in gene-edited WWS iPS cell-derived myotubes. Transplantation of gene-corrected myogenic progenitors in the FKRPP448L-NSG mouse model gives rise to myofiber and satellite cell engraftment and, importantly, restoration of α-DG functional glycosylation in vivo. These findings suggest the potential feasibility of using CRISPR-Cas9 technology in combination with patient-specific iPS cells for the future development of autologous cell transplantation for FKRP-associated MDs.
Collapse
Affiliation(s)
- Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nelio A J Oliveira
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sudheer Tungtur
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Carolina Ortiz-Cordero
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
7
|
Ortiz-Cordero C, Azzag K, Perlingeiro RCR. Fukutin-Related Protein: From Pathology to Treatments. Trends Cell Biol 2020; 31:197-210. [PMID: 33272829 DOI: 10.1016/j.tcb.2020.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/27/2022]
Abstract
Fukutin-related protein (FKRP) is a glycosyltransferase involved in the functional glycosylation of α-dystroglycan (DG), a key component in the link between the cytoskeleton and the extracellular matrix (ECM). Mutations in FKRP lead to dystroglycanopathies with broad severity, including limb-girdle and congenital muscular dystrophy. Studies over the past 5 years have elucidated the function of FKRP, which has expanded the number of therapeutic opportunities for patients carrying FKRP mutations. These include small molecules, gene delivery, and cell therapy. Here we summarize recent findings on the function of FKRP and describe available models for studying diseases and testing therapeutics. Lastly, we highlight preclinical studies that hold potential for the treatment of FKRP-associated dystroglycanopathies.
Collapse
Affiliation(s)
- Carolina Ortiz-Cordero
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Nickolls AR, Lee MM, Zukosky K, Mallon BS, Bönnemann CG. Human embryoid bodies as a 3D tissue model of the extracellular matrix and α-dystroglycanopathies. Dis Model Mech 2020; 13:dmm042986. [PMID: 32423971 PMCID: PMC7328151 DOI: 10.1242/dmm.042986] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
The basal lamina is a specialized sheet of dense extracellular matrix (ECM) linked to the plasma membrane of specific cell types in their tissue context, which serves as a structural scaffold for organ genesis and maintenance. Disruption of the basal lamina and its functions is central to many disease processes, including cancer metastasis, kidney disease, eye disease, muscular dystrophies and specific types of brain malformation. The latter three pathologies occur in the α-dystroglycanopathies, which are caused by dysfunction of the ECM receptor α-dystroglycan. However, opportunities to study the basal lamina in various human disease tissues are restricted owing to its limited accessibility. Here, we report the generation of embryoid bodies from human induced pluripotent stem cells that model the basal lamina. Embryoid bodies cultured via this protocol mimic pre-gastrulation embryonic development, consisting of an epithelial core surrounded by a basal lamina and a peripheral layer of ECM-secreting endoderm. In α-dystroglycanopathy patient embryoid bodies, electron and fluorescence microscopy reveal ultrastructural basal lamina defects and reduced ECM accumulation. By starting from patient-derived cells, these results establish a method for the in vitro synthesis of patient-specific basal lamina and recapitulate disease-relevant ECM defects seen in the α-dystroglycanopathies. Finally, we apply this system to evaluate an experimental ribitol supplement therapy on genetically diverse α-dystroglycanopathy patient samples.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Michelle M Lee
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristen Zukosky
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Barbara S Mallon
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
9
|
Ribitol enhances matriglycan of α-dystroglycan in breast cancer cells without affecting cell growth. Sci Rep 2020; 10:4935. [PMID: 32188898 PMCID: PMC7080755 DOI: 10.1038/s41598-020-61747-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/25/2020] [Indexed: 11/24/2022] Open
Abstract
The laminin-binding glycan (matriglycan) on α-dystroglycan (α-DG) enables diverse roles, from neuronal development to muscle integrity. Reduction or loss of matriglycan has also been implicated in cancer development and metastasis, and specifically associated with high-grade tumors and poor prognoses in breast cancers. Hyperglycosylation of α-DG with LARGE overexpression is shown to inhibit cancer cell growth and tumorigenicity. We recently demonstrated that ribitol, considered to be a metabolic end-product, enhances matriglycan expression in dystrophic muscles in vivo. In the current study, we tested the hypothesis that ribitol could also enhance matriglycan expression in cancer cells. Our results showed for the first time that ribitol is able to significantly enhance the expression of matriglycan on α-DG in breast cancer cells. The ribitol effect is associated with an increase in levels of CDP-ribitol, the substrate for the ribitol-5-phosphate transferases FKRP and FKTN. Direct use of CDP-ribitol is also effective for matriglycan expression. Ribitol treatment does not alter the expression of FKRP, FKTN as well as LARGEs and ISPD which are critical for the synthesis of matriglycan. The results suggest that alteration in substrates could also be involved in regulation of matriglycan expression. Interestingly, expression of matriglycan is related to cell cycle progression with highest levels in S and G2 phases and ribitol treatment does not alter the pattern. Although matriglycan up-regulation does not affect cell cycle progression and proliferation of the cancer cells tested, the novel substrate-mediated treatment opens a new approach easily applicable to experimental systems in vivo for further exploitation of matriglycan expression in cancer progression and for therapeutic potential.
Collapse
|
10
|
Thuriot F, Gravel E, Buote C, Doyon M, Lapointe E, Marcoux L, Larue S, Nadeau A, Chénier S, Waters PJ, Jacques PÉ, Gravel S, Lévesque S. Molecular diagnosis of muscular diseases in outpatient clinics: A Canadian perspective. NEUROLOGY-GENETICS 2020; 6:e408. [PMID: 32337335 PMCID: PMC7164974 DOI: 10.1212/nxg.0000000000000408] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/24/2020] [Indexed: 12/18/2022]
Abstract
Objective To evaluate the diagnostic yield of an 89-gene panel in a large cohort of patients with suspected muscle disorders and to compare the diagnostic yield of gene panel and exome sequencing approaches. Methods We tested 1,236 patients from outpatient clinics across Canada using a gene panel and performed exome sequencing for 46 other patients with sequential analysis of 89 genes followed by all mendelian genes. Sequencing and analysis were performed in patients with muscle weakness or symptoms suggestive of a muscle disorder and showing at least 1 supporting clinical laboratory. Results We identified a molecular diagnosis in 187 (15.1%) of the 1,236 patients tested with the 89-gene panel. Diagnoses were distributed across 40 different genes, but 6 (DMD, RYR1, CAPN3, PYGM, DYSF, and FKRP) explained about half of all cases. Cardiac anomalies, positive family history, age <60 years, and creatine kinase >1,000 IU/L were all associated with increased diagnostic yield. Exome sequencing identified a diagnosis in 10 (21.7%) of the 46 patients tested. Among these, 3 were attributed to genes not included in the 89-gene panel. Despite differences in median coverage, only 1 of the 187 diagnoses that were identified on gene panel in the 1,236 patients could have been potentially missed if exome sequencing had been performed instead. Conclusions Our study supports the use of gene panel testing in patients with suspected muscle disorders from outpatient clinics. It also shows that exome sequencing has a low risk of missing diagnoses compared with gene panel, while potentially increasing the diagnostic yield of patients with muscle disorders.
Collapse
Affiliation(s)
- Fanny Thuriot
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Elaine Gravel
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Caroline Buote
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Marianne Doyon
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Elvy Lapointe
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Lydia Marcoux
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Sandrine Larue
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Amélie Nadeau
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Sébastien Chénier
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Paula J Waters
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Pierre-Étienne Jacques
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Serge Gravel
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| | - Sébastien Lévesque
- Department of Pediatrics (F.T., E.G., C.B., M.D., L.M., A.N., S.C., P.J.W., S.G., S. Lévesque), Université de Sherbrooke; Sherbrooke Genomic Medicine (F.T., E.G., C.B., S.G., S. Lévesque); RNomic's Platform (E.L.), Université de Sherbrooke; Department of Neurology (S. Larue), Notre-Dame Hospital, Université de Montréal; Department of Biology (P.-É.J.), Université de Sherbrooke; and Department of Computer Sciences (P.-É.J.), Université de Sherbrooke, Quebec, Canada
| |
Collapse
|
11
|
biAb Mediated Restoration of the Linkage between Dystroglycan and Laminin-211 as a Therapeutic Approach for α-Dystroglycanopathies. Mol Ther 2019; 28:664-676. [PMID: 31843448 PMCID: PMC7001080 DOI: 10.1016/j.ymthe.2019.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/15/2019] [Accepted: 11/23/2019] [Indexed: 11/20/2022] Open
Abstract
Patients with α-dystroglycanopathies, a subgroup of rare congenital muscular dystrophies, present with a spectrum of clinical manifestations that includes muscular dystrophy as well as CNS and ocular abnormalities. Although patients with α-dystroglycanopathies are genetically heterogeneous, they share a common defect of aberrant post-translational glycosylation modification of the dystroglycan alpha-subunit, which renders it defective in binding to several extracellular ligands such as laminin-211 in skeletal muscles, agrin in neuromuscular junctions, neurexin in the CNS, and pikachurin in the eye, leading to various symptoms. The genetic heterogeneity associated with the development of α-dystroglycanopathies poses significant challenges to developing a generalized treatment to address the spectrum of genetic defects. Here, we propose the development of a bispecific antibody (biAb) that functions as a surrogate molecular linker to reconnect laminin-211 and the dystroglycan beta-subunit to ameliorate sarcolemmal fragility, a primary pathology in patients with α-dystroglycan-related muscular dystrophies. We show that the treatment of LARGEmyd-3J mice, an α-dystroglycanopathy model, with the biAb improved muscle function and protected muscles from exercise-induced damage. These results demonstrate the viability of a biAb that binds to laminin-211 and dystroglycan simultaneously as a potential treatment for patients with α-dystroglycanopathy.
Collapse
|
12
|
Nickolls AR, Bönnemann CG. The roles of dystroglycan in the nervous system: insights from animal models of muscular dystrophy. Dis Model Mech 2018; 11:11/12/dmm035931. [PMID: 30578246 PMCID: PMC6307911 DOI: 10.1242/dmm.035931] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dystroglycan is a cell membrane protein that binds to the extracellular matrix in a variety of mammalian tissues. The α-subunit of dystroglycan (αDG) is heavily glycosylated, including a special O-mannosyl glycoepitope, relying upon this unique glycosylation to bind its matrix ligands. A distinct group of muscular dystrophies results from specific hypoglycosylation of αDG, and they are frequently associated with central nervous system involvement, ranging from profound brain malformation to intellectual disability without evident morphological defects. There is an expanding literature addressing the function of αDG in the nervous system, with recent reports demonstrating important roles in brain development and in the maintenance of neuronal synapses. Much of these data are derived from an increasingly rich array of experimental animal models. This Review aims to synthesize the information from such diverse models, formulating an up-to-date understanding about the various functions of αDG in neurons and glia of the central and peripheral nervous systems. Where possible, we integrate these data with our knowledge of the human disorders to promote translation from basic mechanistic findings to clinical therapies that take the neural phenotypes into account. Summary: Dystroglycan is a ubiquitous matrix receptor linked to brain and muscle disease. Unraveling the functions of this protein will inform basic and translational research on neural development and muscular dystrophies.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Balci-Hayta B, Talim B, Kale G, Dincer P. LARGE expression in different types of muscular dystrophies other than dystroglycanopathy. BMC Neurol 2018; 18:207. [PMID: 30553274 PMCID: PMC6295086 DOI: 10.1186/s12883-018-1207-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/27/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Alpha-dystroglycan (αDG) is an extracellular peripheral glycoprotein that acts as a receptor for both extracellular matrix proteins containing laminin globular domains and certain arenaviruses. An important enzyme, known as Like-acetylglucosaminyltransferase (LARGE), has been shown to transfer repeating units of -glucuronic acid-β1,3-xylose-α1,3- (matriglycan) to αDG that is required for functional receptor as an extracellular matrix protein scaffold. The reduction in the amount of LARGE-dependent matriglycan result in heterogeneous forms of dystroglycanopathy that is associated with hypoglycosylation of αDG and a consequent lack of ligand-binding activity. Our aim was to investigate whether LARGE expression showed correlation with glycosylation of αDG and histopathological parameters in different types of muscular dystrophies, except for dystroglycanopathies. METHODS The expression level of LARGE and glycosylation status of αDG were examined in skeletal muscle biopsies from 26 patients with various forms of muscular dystrophy [Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), sarcoglycanopathy, dysferlinopathy, calpainopathy, and merosin and collagen VI deficient congenital muscular dystrophies (CMDs)] and correlation of results with different histopathological features was investigated. RESULTS Despite the fact that these diseases are not caused by defects of glycosyltransferases, decreased expression of LARGE was detected in many patient samples, partly correlating with the type of muscular dystrophy. Although immunolabelling of fully glycosylated αDG with VIA4-1 was reduced in dystrophinopathy patients, no significant relationship between reduction of LARGE expression and αDG hypoglycosylation was detected. Also, Merosin deficient CMD patients showed normal immunostaining with αDG despite severe reduction of LARGE expression. CONCLUSIONS Our data shows that it is not always possible to correlate LARGE expression and αDG glycosylation in different types of muscular dystrophies and suggests that there might be differences in αDG processing by LARGE which could be regulated under different pathological conditions.
Collapse
Affiliation(s)
- Burcu Balci-Hayta
- Department of Medical Biology, Hacettepe University Faculty of Medicine, 06100 Sihhiye, Ankara, Turkey
| | - Beril Talim
- Department of Pediatrics, Pathology Unit, Hacettepe University Faculty of Medicine, 06100 Sihhiye, Ankara, Turkey
| | - Gulsev Kale
- Department of Pediatrics, Pathology Unit, Hacettepe University Faculty of Medicine, 06100 Sihhiye, Ankara, Turkey
| | - Pervin Dincer
- Department of Medical Biology, Hacettepe University Faculty of Medicine, 06100 Sihhiye, Ankara, Turkey
| |
Collapse
|
14
|
Vannoy CH, Leroy V, Lu QL. Dose-Dependent Effects of FKRP Gene-Replacement Therapy on Functional Rescue and Longevity in Dystrophic Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:106-120. [PMID: 30417025 PMCID: PMC6222079 DOI: 10.1016/j.omtm.2018.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/08/2018] [Indexed: 12/31/2022]
Abstract
Muscular dystrophy-dystroglycanopathies (MDDGs) resulting from fukutin-related protein (FKRP) gene mutations are rare disorders that result in a wide spectrum of clinical severity based on the age of onset, the degree of myogenic atrophy, and/or neurologic involvement. There is no cure for any of the FKRP-related disorders, and few options are available for symptom management. Herein, we examine the longitudinal effects of a dose-escalation study to evaluate the safety and therapeutic potential of FKRP gene-replacement therapy in a p.P448L (FKRPP448L) mouse model of MDDG. A recombinant adeno-associated virus (AAV) serotype 9 vector expressing human FKRP (AAV9-FKRP) was systemically administered to FKRPP448L mice at 5 weeks of age, when early onset of the disease is evidenced. A comprehensive analysis of protein and gene expression, histopathology, skeletal muscle function, and cardiorespiratory function was performed over short (9-week) and/or long-term (52-week) study periods. Additional studies assessed the impact of FKRP gene-replacement therapy on lifespan at an advanced stage of disease progression. Results indicate that treatment intervention can restore the biochemical defects in a dose-dependent manner, with potential for improvement in the trajectory of disease progression and extension of the expected lifespan. This study supports the initiation of early-stage clinical trials for FKRP-related disorders.
Collapse
Affiliation(s)
- Charles Harvey Vannoy
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
- Corresponding author: Charles Harvey Vannoy, McColl-Lockwood Laboratory for Muscular Dystrophy Research, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA.
| | - Victoria Leroy
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
- Corresponding author: Qi Long Lu, McColl-Lockwood Laboratory for Muscular Dystrophy Research, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA.
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW To construct a framework to understand the different molecular interventions for muscular dystrophy. RECENT FINDINGS The recent approval of antisense oligonucleotides treatment for Duchenne muscular dystrophy and spinal muscular atrophy and current clinical trials using recombinant adeno-associated virus for the treatment of those diseases suggests that we are at a tipping point where we are able to treat and potentially cure muscular dystrophies. Understanding the basic molecular pathogenesis of muscular dystrophies and the molecular biology of the treatment allows for critical evaluation of the proposed therapies.
Collapse
Affiliation(s)
- Ava Y Lin
- Department of Neurology, University of Washington, Box 356465, 1959 NE Pacific Street, Seattle, WA, 98195-6465, USA
| | - Leo H Wang
- Department of Neurology, University of Washington, Box 356465, 1959 NE Pacific Street, Seattle, WA, 98195-6465, USA.
| |
Collapse
|
16
|
Tucker JD, Lu PJ, Xiao X, Lu QL. Overexpression of Mutant FKRP Restores Functional Glycosylation and Improves Dystrophic Phenotype in FKRP Mutant Mice. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 11:216-227. [PMID: 29858056 PMCID: PMC5992437 DOI: 10.1016/j.omtn.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/20/2018] [Accepted: 02/23/2018] [Indexed: 11/17/2022]
Abstract
Autosomal recessive homozygous or compound heterozygous mutations in FKRP result in forms of muscular dystrophy-dystroglycanopathy varying in age of onset, clinical presentation, and disease progression, ranging from the severe Walker-Warburg, type A,5 (MDDGA5), muscle-eye-brain (MDDGB5) with or without cognitive deficit, to limb-girdle type 2I (MDDGC5). Phenotypic variation indicates degrees of functionality of individual FKRP mutation, which has been supported by the presence of residual expression of functionally glycosylated α-dystroglycan (DG) in muscles of both animal models and patients. However, direct evidence showing enhancement in glycosylation of α-DG by mutant FKRP is lacking. Using AAV9-mediated overexpression of mutant human FKRP bearing the P448L mutation (mhFKRP-P448L) associated with severe congenital muscular dystrophy (CMD), we demonstrate the restoration of functional glycosylation of α-DG and reduction in markers of disease progression. Expression of mhFKRP-P448L also corrects dystrophic phenotypes in the models of L276I mutation with mild disease phenotype and causes no obvious histological or biomarker alteration in C57BL/6 normal mice. Our results confirm the existing function of mutant FKRP. The results also suggest that mutant FKRP could be an alternative approach for potential gene therapy should normal FKRP gene products be immunogenic.
Collapse
Affiliation(s)
- Jason D Tucker
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Pei J Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Qi L Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, James G. Cannon Research Center, Carolinas Medical Center, Charlotte, NC 28203, USA.
| |
Collapse
|
17
|
Frattini P, Villa C, De Santis F, Meregalli M, Belicchi M, Erratico S, Bella P, Raimondi MT, Lu Q, Torrente Y. Autologous intramuscular transplantation of engineered satellite cells induces exosome-mediated systemic expression of Fukutin-related protein and rescues disease phenotype in a murine model of limb-girdle muscular dystrophy type 2I. Hum Mol Genet 2018; 26:3682-3698. [PMID: 28666318 PMCID: PMC5886111 DOI: 10.1093/hmg/ddx252] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022] Open
Abstract
α-Dystroglycanopathies are a group of muscular dystrophies characterized by α-DG hypoglycosylation and reduced extracellular ligand-binding affinity. Among other genes involved in the α-DG glycosylation process, fukutin related protein (FKRP) gene mutations generate a wide range of pathologies from mild limb girdle muscular dystrophy 2I (LGMD2I), severe congenital muscular dystrophy 1C (MDC1C), to Walker-Warburg Syndrome and Muscle-Eye-Brain disease. FKRP gene encodes for a glycosyltransferase that in vivo transfers a ribitol phosphate group from a CDP –ribitol present in muscles to α-DG, while in vitro it can be secreted as monomer of 60kDa. Consistently, new evidences reported glycosyltransferases in the blood, freely circulating or wrapped within vesicles. Although the physiological function of blood stream glycosyltransferases remains unclear, they are likely released from blood borne or distant cells. Thus, we hypothesized that freely or wrapped FKRP might circulate as an extracellular glycosyltransferase, able to exert a “glycan remodelling” process, even at distal compartments. Interestingly, we firstly demonstrated a successful transduction of MDC1C blood-derived CD133+ cells and FKRP L276IKI mouse derived satellite cells by a lentiviral vector expressing the wild-type of human FKRP gene. Moreover, we showed that LV-FKRP cells were driven to release exosomes carrying FKRP. Similarly, we observed the presence of FKRP positive exosomes in the plasma of FKRP L276IKI mice intramuscularly injected with engineered satellite cells. The distribution of FKRP protein boosted by exosomes determined its restoration within muscle tissues, an overall recovery of α-DG glycosylation and improved muscle strength, suggesting a systemic supply of FKRP protein acting as glycosyltransferase.
Collapse
Affiliation(s)
- Paola Frattini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Francesca De Santis
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Mirella Meregalli
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.,Novystem S.r.l., Milan, Italy
| | - Marzia Belicchi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.,Novystem S.r.l., Milan, Italy
| | | | - Pamela Bella
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Qilong Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Neuromuscular/ALS Center, Department of Neurology, Carolinas Medical Center, Charlotte, North Carolina, NC, USA
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.,Novystem S.r.l., Milan, Italy.,Ystem S.r.l., Milan, Italy
| |
Collapse
|
18
|
Gicquel E, Maizonnier N, Foltz SJ, Martin WJ, Bourg N, Svinartchouk F, Charton K, Beedle AM, Richard I. AAV-mediated transfer of FKRP shows therapeutic efficacy in a murine model but requires control of gene expression. Hum Mol Genet 2017; 26:1952-1965. [PMID: 28334834 DOI: 10.1093/hmg/ddx066] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/17/2017] [Indexed: 02/06/2023] Open
Abstract
Limb Girdle Muscular Dystrophies type 2I (LGMD2I), a recessive autosomal muscular dystrophy, is caused by mutations in the Fukutin Related Protein (FKRP) gene. It has been proposed that FKRP, a ribitol-5-phosphate transferase, is a participant in α-dystroglycan (αDG) glycosylation, which is important to ensure the cell/matrix anchor of muscle fibers. A LGMD2I knock-in mouse model was generated to express the most frequent mutation (L276I) encountered in patients. The expression of FKRP was not altered neither at transcriptional nor at translational levels, but its function was impacted since abnormal glycosylation of αDG was observed. Skeletal muscles were functionally impaired from 2 months of age and a moderate dystrophic pattern was evident starting from 6 months of age. Gene transfer with a rAAV2/9 vector expressing Fkrp restored biochemical defects, corrected the histological abnormalities and improved the resistance to eccentric stress in the mouse model. However, injection of high doses of the vector induced a decrease of αDG glycosylation and laminin binding, even in WT animals. Finally, intravenous injection of the rAAV-Fkrp vector into a dystroglycanopathy mouse model due to Fukutin (Fktn) knock-out indicated a dose-dependent toxicity. These data suggest requirement for a control of FKRP expression in muscles.
Collapse
Affiliation(s)
- Evelyne Gicquel
- INSERM, U951, INTEGRARE Research Unit, Généthon, Evry, F-91002, France
| | | | - Steven J Foltz
- Pharmaceutical & Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, USA
| | - William J Martin
- Animal Health Research Center, University of Georgia, Athens, GA 30602, USA
| | - Nathalie Bourg
- INSERM, U951, INTEGRARE Research Unit, Généthon, Evry, F-91002, France
| | | | - Karine Charton
- INSERM, U951, INTEGRARE Research Unit, Généthon, Evry, F-91002, France
| | - Aaron M Beedle
- Pharmaceutical & Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, USA.,Pharmaceutical Sciences, Binghamton University SUNY, Binghamton, NY 13902, USA
| | - Isabelle Richard
- INSERM, U951, INTEGRARE Research Unit, Généthon, Evry, F-91002, France
| |
Collapse
|
19
|
Nelson CE, Robinson-Hamm JN, Gersbach CA. Genome engineering: a new approach to gene therapy for neuromuscular disorders. Nat Rev Neurol 2017; 13:647-661. [DOI: 10.1038/nrneurol.2017.126] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Vannoy CH, Xiao W, Lu P, Xiao X, Lu QL. Efficacy of Gene Therapy Is Dependent on Disease Progression in Dystrophic Mice with Mutations in the FKRP Gene. Mol Ther Methods Clin Dev 2017; 5:31-42. [PMID: 28480302 PMCID: PMC5415313 DOI: 10.1016/j.omtm.2017.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/20/2017] [Indexed: 01/17/2023]
Abstract
Loss-of-function mutations in the Fukutin-related protein (FKRP) gene cause limb-girdle muscular dystrophy type 2I (LGMD2I) and other forms of congenital muscular dystrophy-dystroglycanopathy that are associated with glycosylation defects in the α-dystroglycan (α-DG) protein. Systemic administration of a single dose of recombinant adeno-associated virus serotype 9 (AAV9) vector expressing human FKRP to a mouse model of LGMD2I at various stages of disease progression was evaluated. The results demonstrate rescue of functional glycosylation of α-DG and muscle function, along with improvements in muscle structure at all disease stages versus age-matched untreated cohorts. Nevertheless, mice treated in the latter stages of disease progression revealed a decrease in beneficial effects of the treatment. The results provide a proof of concept for future clinical trials in patients with FKRP-related muscular dystrophy and demonstrate that AAV-mediated gene therapy can potentially benefit patients at all stages of disease progression, but earlier intervention would be highly preferred.
Collapse
Affiliation(s)
- Charles Harvey Vannoy
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, NC 28203, USA
| | - Will Xiao
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, NC 28203, USA
| | - Peijuan Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, NC 28203, USA
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, NC 28203, USA
| |
Collapse
|
21
|
Vannoy CH, Zhou H, Qiao C, Xiao X, Bang AG, Lu QL. Adeno-Associated Virus-Mediated Mini-Agrin Delivery Is Unable to Rescue Disease Phenotype in a Mouse Model of Limb Girdle Muscular Dystrophy Type 2I. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:431-440. [PMID: 28107841 DOI: 10.1016/j.ajpath.2016.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 11/19/2022]
Abstract
Agrin is a basement membrane-specific proteoglycan that can regulate orientation of cytoskeleton proteins and improve function of dystrophic skeletal muscle. In skeletal muscle, agrin binds with high affinity to laminin(s) and α-dystroglycan (α-DG), an integral part of the dystrophin-glycoprotein complex. Miniaturized forms of agrin (mAgrin) have been shown to ameliorate disease pathology in a laminin-α2 knockout mouse model of muscular dystrophy, acting as a link between α-DG and laminin(s). Here, we test whether mAgrin might also improve pathologies associated with FKRP-related dystroglycanopathies, another form of muscular dystrophy characterized by weak interactions between muscle and basement membranes. We demonstrate in vitro that mAgrin enhances laminin binding to primary myoblasts and fibroblasts from an FKRP mutant mouse model and that this enhancement is abrogated when mAgrin is in molar excess relative to laminin. However, in vivo delivery of mAgrin via adeno-associated virus (AAV) into FKRP mutant mice was unable to improve dystrophic phenotypes, both histologically and functionally. These results likely reflect insufficient binding of mAgrin to hypoglycosylated α-DG on muscle fibers and possibly abrogation of binding from molar excess of overexpressed AAV-delivered mAgrin. Further exploration of mAgrin modification is necessary to strengthen its binding to other membrane components, including hypoglycosylated α-DG, for potential therapeutic applications.
Collapse
Affiliation(s)
- Charles H Vannoy
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, North Carolina
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Chunping Qiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| | - Qi L Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Carolinas Healthcare System, Charlotte, North Carolina.
| |
Collapse
|
22
|
Keramaris E, Lu PJ, Tucker J, Lu QL. Expression of glycosylated α-dystroglycan in newborn skeletal and cardiac muscles of fukutin related protein (FKRP) mutant mice. Muscle Nerve 2016; 55:582-590. [DOI: 10.1002/mus.25378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Elizabeth Keramaris
- McColl-Lockwood Laboratory for Muscular Dystrophy Research; Cannon Research Center, Carolinas Medical Center; 1542 Garden Terrace Charlotte North Carolina 28203 USA
| | - Pei J. Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research; Cannon Research Center, Carolinas Medical Center; 1542 Garden Terrace Charlotte North Carolina 28203 USA
| | - Jason Tucker
- McColl-Lockwood Laboratory for Muscular Dystrophy Research; Cannon Research Center, Carolinas Medical Center; 1542 Garden Terrace Charlotte North Carolina 28203 USA
| | - Qi L. Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research; Cannon Research Center, Carolinas Medical Center; 1542 Garden Terrace Charlotte North Carolina 28203 USA
| |
Collapse
|
23
|
Postnatal Gene Therapy Improves Spatial Learning Despite the Presence of Neuronal Ectopia in a Model of Neuronal Migration Disorder. Genes (Basel) 2016; 7:genes7120105. [PMID: 27916859 PMCID: PMC5192481 DOI: 10.3390/genes7120105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/17/2016] [Accepted: 11/19/2016] [Indexed: 11/25/2022] Open
Abstract
Patients with type II lissencephaly, a neuronal migration disorder with ectopic neurons, suffer from severe mental retardation, including learning deficits. There is no effective therapy to prevent or correct the formation of neuronal ectopia, which is presumed to cause cognitive deficits. We hypothesized that learning deficits were not solely caused by neuronal ectopia and that postnatal gene therapy could improve learning without correcting the neuronal ectopia formed during fetal development. To test this hypothesis, we evaluated spatial learning of cerebral cortex-specific protein O-mannosyltransferase 2 (POMT2, an enzyme required for O-mannosyl glycosylation) knockout mice and compared to the knockout mice that were injected with an adeno-associated viral vector (AAV) encoding POMT2 into the postnatal brains with Barnes maze. The data showed that the knockout mice exhibited reduced glycosylation in the cerebral cortex, reduced dendritic spine density on CA1 neurons, and increased latency to the target hole in the Barnes maze, indicating learning deficits. Postnatal gene therapy restored functional glycosylation, rescued dendritic spine defects, and improved performance on the Barnes maze by the knockout mice even though neuronal ectopia was not corrected. These results indicate that postnatal gene therapy improves spatial learning despite the presence of neuronal ectopia.
Collapse
|
24
|
Mechanistic aspects of the formation of α-dystroglycan and therapeutic research for the treatment of α-dystroglycanopathy: A review. Mol Aspects Med 2016; 51:115-24. [PMID: 27421908 DOI: 10.1016/j.mam.2016.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 02/08/2023]
Abstract
α-Dystroglycanopathy, an autosomal recessive disease, is associated with the development of a variety of diseases, including muscular dystrophy. In humans, α-dystroglycanopathy includes various types of congenital muscular dystrophy such as Fukuyama type congenital muscular dystrophy (FCMD), muscle eye brain disease (MEB), and the Walker Warburg syndrome (WWS), and types of limb girdle muscular dystrophy 2I (LGMD2I). α-Dystroglycanopathy share a common etiology, since it is invariably caused by gene mutations that are associated with the O-mannose glycosylation pathway of α-dystroglycan (α-DG). α-DG is a central member of the dystrophin glycoprotein complex (DGC) family in peripheral membranes, and the proper glycosylation of α-DG is essential for it to bind to extracellular matrix proteins, such as laminin, to cell components. The disruption of this ligand-binding is thought to result in damage to cell membrane integration, leading to the development of muscular dystrophy. Clinical manifestations of α-dystroglycanopathy frequently include mild to severe alterations in the central nervous system and optical manifestations in addition to muscular dystrophy. Eighteen causative genes for α-dystroglycanopathy have been identified to date, and it is likely that more will be reported in the near future. These findings have stimulated extensive and energetic investigations in this research field, and novel glycosylation pathways have been implicated in the process. At the same time, the use of gene therapy, antisense therapy, and enzymatic supplementation have been evaluated as therapeutic possibilities for some types of α-dystroglycanopathy. Here we review the molecular and clinical findings associated with α-dystroglycanopathy and the development of therapeutic approaches, by comparing the approaches with the development of Duchenne muscular dystrophy.
Collapse
|
25
|
Yue Y, Binalsheikh IM, Leach SB, Domeier TL, Duan D. Prospect of gene therapy for cardiomyopathy in hereditary muscular dystrophy. Expert Opin Orphan Drugs 2015; 4:169-183. [PMID: 27340611 DOI: 10.1517/21678707.2016.1124039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cardiac involvement is a common feature in muscular dystrophies. It presents as heart failure and/or arrhythmia. Traditionally, dystrophic cardiomyopathy is treated with symptom-relieving medications. Identification of disease-causing genes and investigation on pathogenic mechanisms have opened new opportunities to treat dystrophic cardiomyopathy with gene therapy. Replacing/repairing the mutated gene and/or targeting the pathogenic process/mechanisms using alternative genes may attenuate heart disease in muscular dystrophies. AREAS COVERED Duchenne muscular dystrophy is the most common muscular dystrophy. Duchenne cardiomyopathy has been the primary focus of ongoing dystrophic cardiomyopathy gene therapy studies. Here, we use Duchenne cardiomyopathy gene therapy to showcase recent developments and to outline the path forward. We also discuss gene therapy status for cardiomyopathy associated with limb-girdle and congenital muscular dystrophies, and myotonic dystrophy. EXPERT OPINION Gene therapy for dystrophic cardiomyopathy has taken a slow but steady path forward. Preclinical studies over the last decades have addressed many fundamental questions. Adeno-associated virus-mediated gene therapy has significantly improved the outcomes in rodent models of Duchenne and limb girdle muscular dystrophies. Validation of these encouraging results in large animal models will pave the way to future human trials.
Collapse
Affiliation(s)
- Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri
| | | | - Stacey B Leach
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri
| | - Timothy L Domeier
- Department of Medical Physiology and Pharmacology, School of Medicine, University of Missouri
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri; Department of Neurology, School of Medicine, University of Missouri
| |
Collapse
|
26
|
Genetic Engineering of Dystroglycan in Animal Models of Muscular Dystrophy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:635792. [PMID: 26380289 PMCID: PMC4561298 DOI: 10.1155/2015/635792] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/11/2015] [Indexed: 01/24/2023]
Abstract
In skeletal muscle, dystroglycan (DG) is the central component of the dystrophin-glycoprotein complex (DGC), a multimeric protein complex that ensures a strong mechanical link between the extracellular matrix and the cytoskeleton. Several muscular dystrophies arise from mutations hitting most of the components of the DGC. Mutations within the DG gene (DAG1) have been recently associated with two forms of muscular dystrophy, one displaying a milder and one a more severe phenotype. This review focuses specifically on the animal (murine and others) model systems that have been developed with the aim of directly engineering DAG1 in order to study the DG function in skeletal muscle as well as in other tissues. In the last years, conditional animal models overcoming the embryonic lethality of the DG knock-out in mouse have been generated and helped clarifying the crucial role of DG in skeletal muscle, while an increasing number of studies on knock-in mice are aimed at understanding the contribution of single amino acids to the stability of DG and to the possible development of muscular dystrophy.
Collapse
|
27
|
Yoshida-Moriguchi T, Campbell KP. Matriglycan: a novel polysaccharide that links dystroglycan to the basement membrane. Glycobiology 2015; 25:702-13. [PMID: 25882296 PMCID: PMC4453867 DOI: 10.1093/glycob/cwv021] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 04/08/2015] [Indexed: 01/01/2023] Open
Abstract
Associations between cells and the basement membrane are critical for a variety of biological events including cell proliferation, cell migration, cell differentiation and the maintenance of tissue integrity. Dystroglycan is a highly glycosylated basement membrane receptor, and is involved in physiological processes that maintain integrity of the skeletal muscle, as well as development and function of the central nervous system. Aberrant O-glycosylation of the α subunit of this protein, and a concomitant loss of dystroglycan's ability to function as a receptor for extracellular matrix (ECM) ligands that bear laminin globular (LG) domains, occurs in several congenital/limb-girdle muscular dystrophies (also referred to as dystroglycanopathies). Recent genetic studies revealed that mutations in DAG1 (which encodes dystroglycan) and at least 17 other genes disrupt the ECM receptor function of dystroglycan and cause disease. Here, we summarize recent advances in our understanding of the enzymatic functions of two of these disease genes: the like-glycosyltransferase (LARGE) and protein O-mannose kinase (POMK, previously referred to as SGK196). In addition, we discuss the structure of the glycan that directly binds the ECM ligands and the mechanisms by which this functional motif is linked to dystroglycan. In light of the fact that dystroglycan functions as a matrix receptor and the polysaccharide synthesized by LARGE is the binding motif for matrix proteins, we propose to name this novel polysaccharide structure matriglycan.
Collapse
Affiliation(s)
- Takako Yoshida-Moriguchi
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, IA 52242-1101, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Department of Molecular Physiology and Biophysics, Department of Neurology, Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, 4283 Carver Biomedical Research Building, 285 Newton Road, Iowa City, IA 52242-1101, USA
| |
Collapse
|
28
|
Ohtsuka Y, Kanagawa M, Yu CC, Ito C, Chiyo T, Kobayashi K, Okada T, Takeda S, Toda T. Fukutin is prerequisite to ameliorate muscular dystrophic phenotype by myofiber-selective LARGE expression. Sci Rep 2015; 5:8316. [PMID: 25661440 PMCID: PMC4321163 DOI: 10.1038/srep08316] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/13/2015] [Indexed: 12/22/2022] Open
Abstract
α-Dystroglycanopathy (α-DGP) is a group of muscular dystrophy characterized by abnormal glycosylation of α-dystroglycan (α-DG), including Fukuyama congenital muscular dystrophy (FCMD), muscle-eye-brain disease, Walker-Warburg syndrome, and congenital muscular dystrophy type 1D (MDC1D), etc. LARGE, the causative gene for MDC1D, encodes a glycosyltransferase to form [-3Xyl-α1,3GlcAβ1-] polymer in the terminal end of the post-phosphoryl moiety, which is essential for α-DG function. It has been proposed that LARGE possesses the great potential to rescue glycosylation defects in α-DGPs regardless of causative genes. However, the in vivo therapeutic benefit of using LARGE activity is controversial. To explore the conditions needed for successful LARGE gene therapy, here we used Large-deficient and fukutin-deficient mouse models for MDC1D and FCMD, respectively. Myofibre-selective LARGE expression via systemic adeno-associated viral gene transfer ameliorated dystrophic pathology of Large-deficient mice even when intervention occurred after disease manifestation. However, the same strategy failed to ameliorate the dystrophic phenotype of fukutin-conditional knockout mice. Furthermore, forced expression of Large in fukutin-deficient embryonic stem cells also failed to recover α-DG glycosylation, however coexpression with fukutin strongly enhanced α-DG glycosylation. Together, our data demonstrated that fukutin is required for LARGE-dependent rescue of α-DG glycosylation, and thus suggesting new directions for LARGE-utilizing therapy targeted to myofibres.
Collapse
Affiliation(s)
- Yoshihisa Ohtsuka
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Motoi Kanagawa
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Chih-Chieh Yu
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Chiyomi Ito
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Tomoko Chiyo
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, 187-8502, Japan
| | - Kazuhiro Kobayashi
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Takashi Okada
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, 187-8502, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, 187-8502, Japan
| | - Tatsushi Toda
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| |
Collapse
|