1
|
Qualtieri A, De Benedittis S, Cerantonio A, Citrigno L, Di Palma G, Gallo O, Cavalcanti F, Spadafora P. Molecular Study of the Fukutin-Related Protein ( FKRP) Gene in Patients from Southern Italy with Duchenne/Becker-like Phenotype. Int J Mol Sci 2024; 25:10356. [PMID: 39408683 PMCID: PMC11476872 DOI: 10.3390/ijms251910356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Pathogenic variants localized in the gene coding for the Fukutin-Related Protein (FKRP) are responsible for Limb-Girdle Muscular Dystrophy type 9 (LGMDR9), Congenital Muscular Dystrophies type 1C (MDC1C), Walker-Warburg Syndrome (WWS), and Muscle-Eye-Brain diseases (MEBs). LGMDR9 is the fourth most common hereditary Limb Girdle Muscular Dystrophy in Italy. LGMDR9 patients with severe disease show an overlapping Duchenne/Becker phenotype and may have secondary dystrophin reduction on muscle biopsy. We conducted a molecular analysis of the FKRP gene by direct sequencing in 153 patients from Southern Italy (Calabria) with Duchenne/Becker-like phenotypes without confirmed genetic diagnosis. Mutational screening of the patients (112 men and 41 women, aged between 5 and 84 years), revealed pathogenic variants in 16 subjects. The most frequent variants identified were c.427C > A, p.R143S, and c.826C > A, p.L276I (NM_024301.5). The results obtained show that the Duchenne/Becker-like phenotype is frequently determined by mutations in the FKRP gene in our cohort and highlight the importance of considering LGMDR9 in the differential diagnosis of dystrophinopathies in Calabria. Finally, this study, which, to our knowledge, is the first conducted on Calabrian subjects, will contribute to the rapid identification and management of LGMDR9 patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Patrizia Spadafora
- Institute for Biomedical Research and Innovation, National Research Council, 87050 Mangone, Italy; (A.Q.); (S.D.B.); (A.C.); (L.C.); (G.D.P.); (O.G.); (F.C.)
| |
Collapse
|
2
|
Poudel BH, Fletcher S, Wilton SD, Aung-Htut M. Limb Girdle Muscular Dystrophy Type 2B (LGMD2B): Diagnosis and Therapeutic Possibilities. Int J Mol Sci 2024; 25:5572. [PMID: 38891760 PMCID: PMC11171558 DOI: 10.3390/ijms25115572] [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: 04/18/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Dysferlin is a large transmembrane protein involved in critical cellular processes including membrane repair and vesicle fusion. Mutations in the dysferlin gene (DYSF) can result in rare forms of muscular dystrophy; Miyoshi myopathy; limb girdle muscular dystrophy type 2B (LGMD2B); and distal myopathy. These conditions are collectively known as dysferlinopathies and are caused by more than 600 mutations that have been identified across the DYSF gene to date. In this review, we discuss the key molecular and clinical features of LGMD2B, the causative gene DYSF, and the associated dysferlin protein structure. We also provide an update on current approaches to LGMD2B diagnosis and advances in drug development, including splice switching antisense oligonucleotides. We give a brief update on clinical trials involving adeno-associated viral gene therapy and the current progress on CRISPR/Cas9 mediated therapy for LGMD2B, and then conclude by discussing the prospects of antisense oligomer-based intervention to treat selected mutations causing dysferlinopathies.
Collapse
Affiliation(s)
- Bal Hari Poudel
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
- Central Department of Biotechnology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| | - May Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
3
|
Xie X, Yuan Y, Huang Y, Hong X, Hong S, Chen G, Chen Y, Lin Y, Lu W, Fu W, Wang L. Effects of COL1A1 and SYTL2 on inflammatory cell infiltration and poor extracellular matrix remodeling of the vascular wall in thoracic aortic aneurysm. Chin Med J (Engl) 2024; 137:1105-1114. [PMID: 37640670 PMCID: PMC11062686 DOI: 10.1097/cm9.0000000000002808] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Thoracic aortic aneurysm (TAA) is a fatal cardiovascular disease, the pathogenesis of which has not yet been clarified. This study aimed to identify and validate the diagnostic markers of TAA to provide a strong theoretical basis for developing new methods to prevent and treat this disease. METHODS Gene expression profiles of the GSE9106, GSE26155, and GSE155468 datasets were acquired from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified using the "limma" package in R. Least absolute shrinkage and selection operator (LASSO), support vector machine-recursive feature elimination (SVM-RFE), random forest, and binary logistic regression analyses were used to screen the diagnostic marker genes. Single-sample gene set enrichment analysis (ssGSEA) was used to estimate immune cell infiltration in TAA. RESULTS A total of 16 DEGs were identified. The enrichment and functional correlation analyses showed that DEGs were mainly associated with inflammatory response pathways and collagen-related diseases. Collagen type I alpha 1 chain ( COL1A1 ) and synaptotagmin like 2 ( SYTL2 ) were identified as diagnostic marker genes with a high diagnostic value for TAA. The expression of COL1A1 and SYTL2 was considerably higher in TAA vascular wall tissues than in the corresponding normal tissues, and there were significant differences in the infiltration of immune cells between TAA and normal vascular wall tissues. Additionally, COL1A1 and SYTL2 expression were associated with the infiltration of immune cells in the vascular wall tissue. Single-cell analysis showed that COL1A1 in TAA was mainly derived from fibroblasts and SYTL2 mainly from cluster of differentiation (CD)8 + T cells. In addition, single-cell analysis indicated that fibroblasts and CD8 + T cells in TAA were significantly higher than those in normal arterial wall tissue. CONCLUSIONS COL1A1 and SYTL2 may serve as diagnostic marker genes for TAA. The upregulation of SYTL2 and COL1A1 may be involved in the inflammatory infiltration of the vessel wall and poor extracellular matrix remodeling, promoting the progression of TAA.
Collapse
Affiliation(s)
- Xinsheng Xie
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Ye Yuan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai 200032, China
| | - Yulong Huang
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Xiang Hong
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Shichai Hong
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Gang Chen
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Yihui Chen
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Yue Lin
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Weifeng Lu
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai 200032, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361015, China
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
5
|
Annexins and Membrane Repair Dysfunctions in Muscular Dystrophies. Int J Mol Sci 2021; 22:ijms22105276. [PMID: 34067866 PMCID: PMC8155887 DOI: 10.3390/ijms22105276] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Muscular dystrophies constitute a group of genetic disorders that cause weakness and progressive loss of skeletal muscle mass. Among them, Miyoshi muscular dystrophy 1 (MMD1), limb girdle muscular dystrophy type R2 (LGMDR2/2B), and LGMDR12 (2L) are characterized by mutation in gene encoding key membrane-repair protein, which leads to severe dysfunctions in sarcolemma repair. Cell membrane disruption is a physiological event induced by mechanical stress, such as muscle contraction and stretching. Like many eukaryotic cells, muscle fibers possess a protein machinery ensuring fast resealing of damaged plasma membrane. Members of the annexins A (ANXA) family belong to this protein machinery. ANXA are small soluble proteins, twelve in number in humans, which share the property of binding to membranes exposing negatively-charged phospholipids in the presence of calcium (Ca2+). Many ANXA have been reported to participate in membrane repair of varied cell types and species, including human skeletal muscle cells in which they may play a collective role in protection and repair of the sarcolemma. Here, we discuss the participation of ANXA in membrane repair of healthy skeletal muscle cells and how dysregulation of ANXA expression may impact the clinical severity of muscular dystrophies.
Collapse
|
6
|
Bittel DC, Chandra G, Tirunagri LMS, Deora AB, Medikayala S, Scheffer L, Defour A, Jaiswal JK. Annexin A2 Mediates Dysferlin Accumulation and Muscle Cell Membrane Repair. Cells 2020; 9:cells9091919. [PMID: 32824910 PMCID: PMC7565960 DOI: 10.3390/cells9091919] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023] Open
Abstract
Muscle cell plasma membrane is frequently damaged by mechanical activity, and its repair requires the membrane protein dysferlin. We previously identified that, similar to dysferlin deficit, lack of annexin A2 (AnxA2) also impairs repair of skeletal myofibers. Here, we have studied the mechanism of AnxA2-mediated muscle cell membrane repair in cultured muscle cells. We find that injury-triggered increase in cytosolic calcium causes AnxA2 to bind dysferlin and accumulate on dysferlin-containing vesicles as well as with dysferlin at the site of membrane injury. AnxA2 accumulates on the injured plasma membrane in cholesterol-rich lipid microdomains and requires Src kinase activity and the presence of cholesterol. Lack of AnxA2 and its failure to translocate to the plasma membrane, both prevent calcium-triggered dysferlin translocation to the plasma membrane and compromise repair of the injured plasma membrane. Our studies identify that Anx2 senses calcium increase and injury-triggered change in plasma membrane cholesterol to facilitate dysferlin delivery and repair of the injured plasma membrane.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Goutam Chandra
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Laxmi M. S. Tirunagri
- Department of Cellular Biophysics, The Rockefeller University, New York, NY 10065, USA;
| | - Arun B. Deora
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Sushma Medikayala
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Luana Scheffer
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Aurelia Defour
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
- Department of Genomics and Precision medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
- Correspondence: ; Tel.: +1-(202)476-6456; Fax: +1-(202)476-6014
| |
Collapse
|
7
|
Bittel AJ, Sreetama SC, Bittel DC, Horn A, Novak JS, Yokota T, Zhang A, Maruyama R, Rowel Q. Lim K, Jaiswal JK, Chen YW. Membrane Repair Deficit in Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2020; 21:E5575. [PMID: 32759720 PMCID: PMC7432481 DOI: 10.3390/ijms21155575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Deficits in plasma membrane repair have been identified in dysferlinopathy and Duchenne Muscular Dystrophy, and contribute to progressive myopathy. Although Facioscapulohumeral Muscular Dystrophy (FSHD) shares clinicopathological features with these muscular dystrophies, it is unknown if FSHD is characterized by plasma membrane repair deficits. Therefore, we exposed immortalized human FSHD myoblasts, immortalized myoblasts from unaffected siblings, and myofibers from a murine model of FSHD (FLExDUX4) to focal, pulsed laser ablation of the sarcolemma. Repair kinetics and success were determined from the accumulation of intracellular FM1-43 dye post-injury. We subsequently treated FSHD myoblasts with a DUX4-targeting antisense oligonucleotide (AON) to reduce DUX4 expression, and with the antioxidant Trolox to determine the role of DUX4 expression and oxidative stress in membrane repair. Compared to unaffected myoblasts, FSHD myoblasts demonstrate poor repair and a greater percentage of cells that failed to repair, which was mitigated by AON and Trolox treatments. Similar repair deficits were identified in FLExDUX4 myofibers. This is the first study to identify plasma membrane repair deficits in myoblasts from individuals with FSHD, and in myofibers from a murine model of FSHD. Our results suggest that DUX4 expression and oxidative stress may be important targets for future membrane-repair therapies.
Collapse
Affiliation(s)
- Adam J. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Sen Chandra Sreetama
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Daniel C. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Adam Horn
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - James S. Novak
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Genomics and Precision Medicine, The George Washington University School of Medicine and Health Science, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Aiping Zhang
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Kenji Rowel Q. Lim
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Jyoti K. Jaiswal
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Yi-Wen Chen
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| |
Collapse
|
8
|
Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun 2019; 10:2430. [PMID: 31160583 PMCID: PMC6547715 DOI: 10.1038/s41467-019-10438-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/10/2019] [Indexed: 12/30/2022] Open
Abstract
Muscle loss due to fibrotic or adipogenic replacement of myofibers is common in muscle diseases and muscle-resident fibro/adipogenic precursors (FAPs) are implicated in this process. While FAP-mediated muscle fibrosis is widely studied in muscle diseases, the role of FAPs in adipogenic muscle loss is not well understood. Adipogenic muscle loss is a feature of limb girdle muscular dystrophy 2B (LGMD2B) - a disease caused by mutations in dysferlin. Here we show that FAPs cause the adipogenic loss of dysferlin deficient muscle. Progressive accumulation of Annexin A2 (AnxA2) in the myofiber matrix causes FAP differentiation into adipocytes. Lack of AnxA2 prevents FAP adipogenesis, protecting against adipogenic loss of dysferlinopathic muscle while exogenous AnxA2 enhances muscle loss. Pharmacological inhibition of FAP adipogenesis arrests adipogenic replacement and degeneration of dysferlin-deficient muscle. These results demonstrate the pathogenic role of FAPs in LGMD2B and establish these cells as therapeutic targets to ameliorate muscle loss in patients.
Collapse
|
9
|
Begam M, Collier AF, Mueller AL, Roche R, Galen SS, Roche JA. Diltiazem improves contractile properties of skeletal muscle in dysferlin-deficient BLAJ mice, but does not reduce contraction-induced muscle damage. Physiol Rep 2018; 6:e13727. [PMID: 29890050 PMCID: PMC5995314 DOI: 10.14814/phy2.13727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/06/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
B6.A-Dysfprmd /GeneJ (BLAJ) mice model human limb-girdle muscular dystrophy 2B (LGMD2B), which is linked to mutations in the dysferlin (DYSF) gene. We tested the hypothesis that, the calcium ion (Ca2+ ) channel blocker diltiazem (DTZ), reduces contraction-induced skeletal muscle damage, in BLAJ mice. We randomly assigned mice (N = 12; 3-4 month old males) to one of two groups - DTZ (N = 6) or vehicle (VEH, distilled water, N = 6). We conditioned mice with either DTZ or VEH for 1 week, after which, their tibialis anterior (TA) muscles were tested for contractile torque and susceptibility to injury from forced eccentric contractions. We continued dosing with DTZ or VEH for 3 days following eccentric contractions, and then studied torque recovery and muscle damage. We analyzed contractile torque before eccentric contractions, immediately after eccentric contractions, and at 3 days after eccentric contractions; and counted damaged fibers in the injured and uninjured TA muscles. We found that DTZ improved contractile torque before and immediately after forced eccentric contractions, but did not reduce delayed-onset muscle damage that was observed at 3 days after eccentric contractions.
Collapse
Affiliation(s)
- Morium Begam
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| | - Alyssa F. Collier
- Program in Physical TherapyWashington University in St. Louis School of MedicineSt. LouisMissouri
| | - Amber L. Mueller
- Program in Molecular MedicineUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Renuka Roche
- Eastern Michigan University School of Health SciencesYpsilantiMichigan
| | - Sujay S. Galen
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| | - Joseph A. Roche
- Physical Therapy ProgramDepartment of Health Care SciencesEugene Applebaum College of Pharmacy and Health SciencesWayne State UniversityDetroitMichigan
| |
Collapse
|
10
|
Sellers SL, Milad N, White Z, Pascoe C, Chan R, Payne GW, Seow C, Rossi F, Seidman MA, Bernatchez P. Increased nonHDL cholesterol levels cause muscle wasting and ambulatory dysfunction in the mouse model of LGMD2B. J Lipid Res 2017; 59:261-272. [PMID: 29175948 DOI: 10.1194/jlr.m079459] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/19/2017] [Indexed: 12/22/2022] Open
Abstract
Progressive limb and girdle muscle atrophy leading to loss of ambulation is a hallmark of dysferlinopathies, which include limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. However, animal models fail to fully reproduce the disease severity observed in humans, with dysferlin-null (Dysf-/-) mice exhibiting minor muscle damage and weakness without dramatic ambulatory dysfunction. As we have previously reported significant Dysf expression in blood vessels, we investigated the role of vascular function in development of muscle pathology by generating a Dysf-deficient mouse model with vascular disease. This was achieved by crossing Dysf-/- mice with ApoE-/- mice, which have high levels of nonHDL-associated cholesterol. Double-knockout Dysf-/-ApoE-/- mice exhibited severe ambulatory dysfunction by 11 months of age. In limb-girdle muscles, histology confirmed dramatic muscle wasting, fibrofatty replacement, and myofiber damage in Dysf-/-ApoE-/- mice without affecting the ratio of centrally nucleated myofibers. Although there were no major changes in ex vivo diaphragm and soleus muscle function, histological analyses revealed these muscles to be untouched by damage and remodelling. In all, these data suggest that cholesterol may be deleterious to dysferlinopathic muscle and lead to ambulatory dysfunction. Moreover, differences in plasma lipid handling between mice and humans could be a key factor affecting dysferlinopathy severity.
Collapse
Affiliation(s)
- Stephanie L Sellers
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Chris Pascoe
- St. Paul's Hospital, University of British Columbia, Vancouver, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Rayleigh Chan
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Geoffrey W Payne
- Providence Health Care, University of Northern British Columbia, Prince George, Canada
| | - Chun Seow
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada.,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Fabio Rossi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,Biomedical Research Centre, University of British Columbia, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Michael A Seidman
- St. Paul's Hospital, University of British Columbia, Vancouver, Canada.,Department of Pathology, Prince George, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics and UBC Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada .,St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
11
|
Defour A, Medikayala S, Van der Meulen JH, Hogarth MW, Holdreith N, Malatras A, Duddy W, Boehler J, Nagaraju K, Jaiswal JK. Annexin A2 links poor myofiber repair with inflammation and adipogenic replacement of the injured muscle. Hum Mol Genet 2017; 26:1979-1991. [PMID: 28334824 DOI: 10.1093/hmg/ddx065] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/17/2017] [Indexed: 01/12/2023] Open
Abstract
Repair of skeletal muscle after sarcolemmal damage involves dysferlin and dysferlin-interacting proteins such as annexins. Mice and patient lacking dysferlin exhibit chronic muscle inflammation and adipogenic replacement of the myofibers. Here, we show that similar to dysferlin, lack of annexin A2 (AnxA2) also results in poor myofiber repair and progressive muscle weakening with age. By longitudinal analysis of AnxA2-deficient muscle we find that poor myofiber repair due to the lack of AnxA2 does not result in chronic inflammation or adipogenic replacement of the myofibers. Further, deletion of AnxA2 in dysferlin deficient mice reduced muscle inflammation, adipogenic replacement of myofibers, and improved muscle function. These results identify multiple roles of AnxA2 in muscle repair, which includes facilitating myofiber repair, chronic muscle inflammation and adipogenic replacement of dysferlinopathic muscle. It also identifies inhibition of AnxA2-mediated inflammation as a novel therapeutic avenue for treating muscle loss in dysferlinopathy.
Collapse
Affiliation(s)
- Aurelia Defour
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Sushma Medikayala
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Jack H Van der Meulen
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Nicholas Holdreith
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Apostolos Malatras
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
| | - William Duddy
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, BT52 1SJ UK
| | - Jessica Boehler
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| |
Collapse
|
12
|
Baek JH, Many GM, Evesson FJ, Kelley VR. Dysferlinopathy Promotes an Intramuscle Expansion of Macrophages with a Cyto-Destructive Phenotype. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1245-1257. [PMID: 28412297 DOI: 10.1016/j.ajpath.2017.02.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/14/2017] [Indexed: 01/05/2023]
Abstract
Dysferlinopathies are a group of muscular dystrophies resulting from a genetic deficiency in Dysf. Macrophages, highly plastic cells that mediate tissue repair and destruction, are prominent within dystrophic skeletal muscles of dysferlinopathy patients. We hypothesized that Dysf-deficient muscle promotes recruitment, proliferation, and skewing of macrophages toward a cyto-destructive phenotype in dysferlinopathy. To track macrophage dynamics in dysferlinopathy, we adoptively transferred enhanced green fluorescent protein-labeled monocytes into Dysf-deficient BLA/J mice with age-related (2 to 10 months) muscle disease and Dysf-intact (C57BL/6 [B6]) mice. We detected an age- and disease-related increase in monocyte recruitment into Dysf-deficient muscles. Moreover, macrophages recruited into muscle proliferated locally and were skewed toward a cyto-destructive phenotype. By comparing Dysf-deficient and -intact monocytes, our data showed that Dysf in muscle, but not in macrophages, mediate intramuscle macrophage recruitment and proliferation. To further elucidate macrophage mechanisms related to dysferlinopathy, we investigated in vitro macrophage-myogenic cell interactions and found that Dysf-deficient muscle i) promotes macrophage proliferation, ii) skews macrophages toward a cyto-destructive phenotype, and iii) is more vulnerable to macrophage-mediated apoptosis. Taken together, our data suggest that the loss of Dysf expression in muscle, not macrophages, promotes the intramuscle expansion of cyto-destructive macrophages likely to contribute to dysferlinopathy. Identifying pathways within the Dysf-deficient muscle milieu that regulate cyto-destructive macrophages will potentially uncover therapeutic strategies for dysferlinopathies.
Collapse
Affiliation(s)
- Jea-Hyun Baek
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Gina M Many
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Frances J Evesson
- Department of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Vicki R Kelley
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
13
|
Vila MC, Klimek MB, Novak JS, Rayavarapu S, Uaesoontrachoon K, Boehler JF, Fiorillo AA, Hogarth MW, Zhang A, Shaughnessy C, Gordish-Dressman H, Burki U, Straub V, Lu QL, Partridge TA, Brown KJ, Hathout Y, van den Anker J, Hoffman EP, Nagaraju K. Elusive sources of variability of dystrophin rescue by exon skipping. Skelet Muscle 2015; 5:44. [PMID: 26634117 PMCID: PMC4667482 DOI: 10.1186/s13395-015-0070-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/24/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Systemic delivery of anti-sense oligonucleotides to Duchenne muscular dystrophy (DMD) patients to induce de novo dystrophin protein expression in muscle (exon skipping) is a promising therapy. Treatment with Phosphorodiamidate morpholino oligomers (PMO) lead to shorter de novo dystrophin protein in both animal models and DMD boys who otherwise lack dystrophin; however, restoration of dystrophin has been observed to be highly variable. Understanding the factors causing highly variable induction of dystrophin expression in pre-clinical models would likely lead to more effective means of exon skipping in both pre-clinical studies and human clinical trials. METHODS In the present study, we investigated possible factors that might lead to the variable success of exon skipping using morpholino drugs in the mdx mouse model. We tested whether specific muscle groups or fiber types showed better success than others and also correlated residual PMO concentration in muscle with the amount of de novo dystrophin protein 1 month after a single high-dose morpholino injection (800 mg/kg). We compared the results from six muscle groups using three different methods of dystrophin quantification: immunostaining, immunoblotting, and mass spectrometry assays. RESULTS The triceps muscle showed the greatest degree of rescue (average 38±28 % by immunostaining). All three dystrophin detection methods were generally concordant for all muscles. We show that dystrophin rescue occurs in a sporadic patchy pattern with high geographic variability across muscle sections. We did not find a correlation between residual morpholino drug in muscle tissue and the degree of dystrophin expression. CONCLUSIONS While we found some evidence of muscle group enhancement and successful rescue, our data also suggest that other yet-undefined factors may underlie the observed variability in the success of exon skipping. Our study highlights the challenges associated with quantifying dystrophin in clinical trials where a single small muscle biopsy is taken from a DMD patient.
Collapse
Affiliation(s)
- Maria Candida Vila
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Margaret Benny Klimek
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - James S Novak
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Sree Rayavarapu
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Kitipong Uaesoontrachoon
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Jessica F Boehler
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Alyson A Fiorillo
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Marshall W Hogarth
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Aiping Zhang
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Conner Shaughnessy
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Heather Gordish-Dressman
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Umar Burki
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases at Newcastle, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases at Newcastle, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Neuromuscular/ALS Center, Department of Neurology, Carolinas Medical Center, Charlotte, NC USA
| | - Terence A Partridge
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Kristy J Brown
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Yetrib Hathout
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - John van den Anker
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Center for Translational Science, Children's National Health System, Washington, DC, USA
| | - Eric P Hoffman
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
14
|
Dillingham BC, Benny Klimek ME, Gernapudi R, Rayavarapu S, Gallardo E, Van der Meulen JH, Jordan S, Ampong B, Gordish-Dressman H, Spurney CF, Nagaraju K. Inhibition of inflammation with celastrol fails to improve muscle function in dysferlin-deficient A/J mice. J Neurol Sci 2015; 356:157-62. [PMID: 26119397 DOI: 10.1016/j.jns.2015.06.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 01/17/2023]
Abstract
The dysferlin-deficient A/J mouse strain represents a homologous model for limb-girdle muscular dystrophy 2B. We evaluated the disease phenotype in 10 month old A/J mice compared to two dysferlin-sufficient, C57BL/6 and A/JOlaHsd, mouse lines to determine which functional end-points are sufficiently sensitive to define the disease phenotype for use in preclinical studies in the A/J strain. A/J mice had significantly lower open field behavioral activity (horizontal activity, total distance, movement time and vertical activity) when compared to C57BL/6 and A/JoIaHsd mice. Both A/J and A/JOIaHsd mice showed decreases in latency to fall with rotarod compared to C57BL/6. No changes were detected in grip strength, force measurements or motor coordination between these three groups. Furthermore, we have found that A/J muscle shows significantly increased levels of the pro-inflammatory cytokine TNF-α when compared to C57BL/6 mice, indicating an activation of NF-κB signaling as part of the inflammatory response in dysferlin-deficient muscle. Therefore, we assessed the effect of celastrol (a potent NF-κB inhibitor) on the disease phenotype in female A/J mice. Celastrol treatment for four months significantly reduced the inflammation in A/J muscle; however, it had no beneficial effect in improving muscle function, as assessed by grip strength, open field activity, and in vitro force contraction. In fact, celastrol treated mice showed a decrease in body mass, hindlimb grip strength and maximal EDL force. These findings suggest that inhibition of inflammation alone may not be sufficient to improve the muscle disease phenotype in dysferlin-deficient mice and may require combination therapies that target membrane stability to achieve a functional improvement in skeletal muscle.
Collapse
Affiliation(s)
- Blythe C Dillingham
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Margaret E Benny Klimek
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Ramkishore Gernapudi
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Sree Rayavarapu
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Eduard Gallardo
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Jack H Van der Meulen
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Sarah Jordan
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Beryl Ampong
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Heather Gordish-Dressman
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Christopher F Spurney
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA; Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2300 Eye Street, N.W., Ross 605, Washington, D.C., USA.
| |
Collapse
|
15
|
Roche JA, Tulapurkar ME, Mueller AL, van Rooijen N, Hasday JD, Lovering RM, Bloch RJ. Myofiber damage precedes macrophage infiltration after in vivo injury in dysferlin-deficient A/J mouse skeletal muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1686-98. [PMID: 25920768 PMCID: PMC4450316 DOI: 10.1016/j.ajpath.2015.02.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 12/31/2014] [Accepted: 02/12/2015] [Indexed: 12/16/2022]
Abstract
Mutations in the dysferlin gene (DYSF) lead to human muscular dystrophies known as dysferlinopathies. The dysferlin-deficient A/J mouse develops a mild myopathy after 6 months of age, and when younger models the subclinical phase of the human disease. We subjected the tibialis anterior muscle of 3- to 4-month-old A/J mice to in vivo large-strain injury (LSI) from lengthening contractions and studied the progression of torque loss, myofiber damage, and inflammation afterward. We report that myofiber damage in A/J mice occurs before inflammatory cell infiltration. Peak edema and inflammation, monitored by magnetic resonance imaging and by immunofluorescence labeling of neutrophils and macrophages, respectively, develop 24 to 72 hours after LSI, well after the appearance of damaged myofibers. Cytokine profiles 72 hours after injury are consistent with extensive macrophage infiltration. Dysferlin-sufficient A/WySnJ mice show much less myofiber damage and inflammation and lesser cytokine levels after LSI than do A/J mice. Partial suppression of macrophage infiltration by systemic administration of clodronate-incorporated liposomes fails to suppress LSI-induced damage or to accelerate torque recovery in A/J mice. The findings from our studies suggest that, although macrophage infiltration is prominent in dysferlin-deficient A/J muscle after LSI, it is the consequence and not the cause of progressive myofiber damage.
Collapse
Affiliation(s)
- Joseph A Roche
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland.
| | - Mohan E Tulapurkar
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Amber L Mueller
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Nico van Rooijen
- Clodronateliposomes.com, Amsterdam, the Netherlands; Department of Molecular Cell Biology, Faculty of Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Jeffrey D Hasday
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Richard M Lovering
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Robert J Bloch
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| |
Collapse
|
16
|
Defour A, Van der Meulen JH, Bhat R, Bigot A, Bashir R, Nagaraju K, Jaiswal JK. Dysferlin regulates cell membrane repair by facilitating injury-triggered acid sphingomyelinase secretion. Cell Death Dis 2014; 5:e1306. [PMID: 24967968 PMCID: PMC4079937 DOI: 10.1038/cddis.2014.272] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/08/2014] [Accepted: 05/20/2014] [Indexed: 01/17/2023]
Abstract
Dysferlin deficiency compromises the repair of injured muscle, but the underlying cellular mechanism remains elusive. To study this phenomenon, we have developed mouse and human myoblast models for dysferlinopathy. These dysferlinopathic myoblasts undergo normal differentiation but have a deficit in their ability to repair focal injury to their cell membrane. Imaging cells undergoing repair showed that dysferlin-deficit decreased the number of lysosomes present at the cell membrane, resulting in a delay and reduction in injury-triggered lysosomal exocytosis. We find repair of injured cells does not involve formation of intracellular membrane patch through lysosome-lysosome fusion; instead, individual lysosomes fuse with the injured cell membrane, releasing acid sphingomyelinase (ASM). ASM secretion was reduced in injured dysferlinopathic cells, and acute treatment with sphingomyelinase restored the repair ability of dysferlinopathic myoblasts and myofibers. Our results provide the mechanism for dysferlin-mediated repair of skeletal muscle sarcolemma and identify ASM as a potential therapy for dysferlinopathy.
Collapse
Affiliation(s)
- A Defour
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - J H Van der Meulen
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - R Bhat
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - A Bigot
- Institut de Myologie, UM76 Université Pierre et Marie Curie, U974 INSERM, UMR7215 CNRS, GH Pitié-Salpétrière, 47 bd de l'Hôpital, Paris, France
| | - R Bashir
- School of Biological and Biochemical Sciences, University of Durham, Durham, UK
| | - K Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - J K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
17
|
Abstract
Small GTPases are key signal transducers from extracellular stimuli to the nucleus that regulate a variety of cellular responses, including changes in gene expression and cell adhesion and migration. Accumulating data have demonstrated that abnormal activation of these small GTPases plays a critical role in the atherosclerosis characterized by vascular abnormalities, especially endothelial dysfunction and inflammation. Here, we discuss the linkage between small GTPases, inflammation, and atherogenesis. First, small GTPases affect gene expression of inflammatory cytokines through proinflammatory signaling pathways, such as nuclear factor-κB, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, interlukin-8, and monocyte chemoattractant protein-1. Then, these molecules regulate the vascular inflammation through cell adhesion and migration. In turn, small GTPases are also regulated by extracellular stimuli, such as L-selectin, thrombin, oxidized phospholipids, and interleukins. Thus, these inflammatory cytokines generate a vicious cycle for small GTPases and inflammatory responses in the atherogenesis.
Collapse
|
18
|
Sula A, Cole AR, Yeats C, Orengo C, Keep NH. Crystal structures of the human Dysferlin inner DysF domain. BMC STRUCTURAL BIOLOGY 2014; 14:3. [PMID: 24438169 PMCID: PMC3898210 DOI: 10.1186/1472-6807-14-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/15/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Mutations in dysferlin, the first protein linked with the cell membrane repair mechanism, causes a group of muscular dystrophies called dysferlinopathies. Dysferlin is a type two-anchored membrane protein, with a single C terminal trans-membrane helix, and most of the protein lying in cytoplasm. Dysferlin contains several C2 domains and two DysF domains which are nested one inside the other. Many pathogenic point mutations fall in the DysF domain region. RESULTS We describe the crystal structure of the human dysferlin inner DysF domain with a resolution of 1.9 Ångstroms. Most of the pathogenic mutations are part of aromatic/arginine stacks that hold the domain in a folded conformation. The high resolution of the structure show that these interactions are a mixture of parallel ring/guanadinium stacking, perpendicular H bond stacking and aliphatic chain packing. CONCLUSIONS The high resolution structure of the Dysferlin DysF domain gives a template on which to interpret in detail the pathogenic mutations that lead to disease.
Collapse
Affiliation(s)
| | | | | | | | - Nicholas H Keep
- Crystallography, Biological Sciences, Institute for Structural and Molecular Biology, Birkbeck University of London, Malet Street, London WC1E 7HX, UK.
| |
Collapse
|
19
|
Fukuda M. Rab27 effectors, pleiotropic regulators in secretory pathways. Traffic 2013; 14:949-63. [PMID: 23678941 DOI: 10.1111/tra.12083] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/13/2013] [Accepted: 05/16/2013] [Indexed: 12/18/2022]
Abstract
Rab27, a member of the small GTPase Rab family, is widely conserved in metazoan, and two Rab27 isoforms, Rab27A and Rab27B, are present in vertebrates. Rab27A was the first Rab protein whose dysfunction was found to cause a human hereditary disease, type 2 Griscelli syndrome, which is characterized by silvery hair and immunodeficiency. The discovery in the 21st century of three distinct types of mammalian Rab27A effectors [synaptotagmin-like protein (Slp), Slp homologue lacking C2 domains (Slac2), and Munc13-4] that specifically bind active Rab27A has greatly accelerated our understanding not only of the molecular mechanisms of Rab27A-mediated membrane traffic (e.g. melanosome transport and regulated secretion) but of the symptoms of Griscelli syndrome patients at the molecular level. Because Rab27B is widely expressed in various tissues together with Rab27A and has been found to have the ability to bind all of the Rab27A effectors that have been tested, Rab27A and Rab27B were initially thought to function redundantly by sharing common Rab27 effectors. However, recent evidence has indicated that by interacting with different Rab27 effectors Rab27A and Rab27B play different roles in special types of secretion (e.g. exosome secretion and mast cell secretion) even within the same cell type. In this review article, I describe the current state of our understanding of the functions of Rab27 effectors in secretory pathways.
Collapse
Affiliation(s)
- Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
20
|
Upregulation of the Rab27a-dependent trafficking and secretory mechanisms improves lysosomal transport, alleviates endoplasmic reticulum stress, and reduces lysosome overload in cystinosis. Mol Cell Biol 2013; 33:2950-62. [PMID: 23716592 DOI: 10.1128/mcb.00417-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cystinosis is a lysosomal storage disorder caused by the accumulation of the amino acid cystine due to genetic defects in the CTNS gene, which encodes cystinosin, the lysosomal cystine transporter. Although many cellular dysfunctions have been described in cystinosis, the mechanisms leading to these defects are not well understood. Here, we show that increased lysosomal overload induced by accumulated cystine leads to cellular abnormalities, including vesicular transport defects and increased endoplasmic reticulum (ER) stress, and that correction of lysosomal transport improves cellular function in cystinosis. We found that Rab27a was expressed in proximal tubular cells (PTCs) and partially colocalized with the lysosomal marker LAMP-1. The expression of Rab27a but not other small GTPases, including Rab3 and Rab7, was downregulated in kidneys from Ctns-/- mice and in human PTCs from cystinotic patients. Using total internal reflection fluorescence microscopy, we found that lysosomal transport is impaired in Ctns-/- cells. Ctns-/- cells showed significant ER expansion and a marked increase in the unfolded protein response-induced chaperones Grp78 and Grp94. Upregulation of the Rab27a-dependent vesicular trafficking mechanisms rescued the defective lysosomal transport phenotype and reduced ER stress in cystinotic cells. Importantly, reconstitution of lysosomal transport mediated by Rab27a led to decreased lysosomal overload, manifested as reduced cystine cellular content. Our data suggest that upregulation of the Rab27a-dependent lysosomal trafficking and secretory pathways contributes to the correction of some of the cellular defects induced by lysosomal overload in cystinosis, including ER stress.
Collapse
|
21
|
Nilsson MI, Laureano ML, Saeed M, Tarnopolsky MA. Dysferlin aggregation in limb-girdle muscular dystrophy type 2B/myoshi myopathy necessitates mutational screen for diagnosis. Muscle Nerve 2013; 47:740-7. [PMID: 23519732 DOI: 10.1002/mus.23666] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2012] [Indexed: 12/24/2022]
Affiliation(s)
- Mats I. Nilsson
- Department of Pediatrics and Medicine; Neuromuscular Clinic; McMaster University Hospital; 1200 Main Street West; Hamilton; Ontario L8N 3Z5; Canada
| | - Marissa L. Laureano
- Department of Pediatrics and Medicine; Neuromuscular Clinic; McMaster University Hospital; 1200 Main Street West; Hamilton; Ontario L8N 3Z5; Canada
| | - Munim Saeed
- Department of Pediatrics and Medicine; Neuromuscular Clinic; McMaster University Hospital; 1200 Main Street West; Hamilton; Ontario L8N 3Z5; Canada
| | - Mark A. Tarnopolsky
- Department of Pediatrics and Medicine; Neuromuscular Clinic; McMaster University Hospital; 1200 Main Street West; Hamilton; Ontario L8N 3Z5; Canada
| |
Collapse
|
22
|
Expression of myoferlin in human and murine carcinoma tumors: role in membrane repair, cell proliferation, and tumorigenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1900-9. [PMID: 23499551 DOI: 10.1016/j.ajpath.2013.01.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 12/19/2022]
Abstract
Cancer cells are often characterized by high proliferation rates, a consequence of increased mitotic signaling coupled with unchecked cellular growth. We recently demonstrated that vascular endothelial cells unexpectedly express ferlins, a family of muscle-specific proteins capable of regulating the fusion of lipid patches to the plasma membrane, and that these highly regulated membrane fusion events are essential to endothelial cell proliferation and homeostasis. Here, we show that human and mouse breast cancer cell lines also express myoferlin at various levels, and that the processes of transformation, epithelial-mesenchymal transition, and metastasis do not appear to have any effect on myoferlin expression in vitro. In vivo, we observed that solid mouse and human carcinoma tissues also express high levels of myoferlin protein. Loss-of-function studies performed in mice revealed that myoferlin gene knockdown can attenuate cancer cell proliferation in vitro and decrease tumor burden, and that accelerated tumor cell growth appears to rely on intact myoferlin-dependent membrane repair and signaling under exponential growth conditions. To our knowledge, these data provide the first evidence of myoferlin expression in solid human and mouse tumors. We have thus identified a novel membrane repair process that likely helps sustain the high growth rates characteristic of tumors, and we suggest that interfering with normal myoferlin expression and/or membrane repair and remodeling may provide therapeutically relevant antiproliferative effects.
Collapse
|
23
|
Bernardini C, Censi F, Lattanzi W, Calcagnini G, Giuliani A. Gene regulation networks in early phase of Duchenne muscular dystrophy. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2013; 10:393-400. [PMID: 23929863 DOI: 10.1109/tcbb.2013.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The aim of this study was to analyze previously published gene expression data of skeletal muscle biopsies of Duchenne muscular dystrophy (DMD) patients and controls (gene expression omnibus database, accession #GSE6011) using systems biology approaches. We applied an unsupervised method to discriminate patient and control populations, based on principal component analysis, using the gene expressions as units and patients as variables. The genes having the highest absolute scores in the discrimination between the groups, were then analyzed in terms of gene expression networks, on the basis of their mutual correlation in the two groups. The correlation network structures suggest two different modes of gene regulation in the two groups, reminiscent of important aspects of DMD pathogenesis.
Collapse
|
24
|
Roostalu U, Strähle U. In vivo imaging of molecular interactions at damaged sarcolemma. Dev Cell 2012; 22:515-29. [PMID: 22421042 DOI: 10.1016/j.devcel.2011.12.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 10/25/2011] [Accepted: 12/14/2011] [Indexed: 12/30/2022]
Abstract
Muscle cells have a remarkable capability to repair plasma membrane lesions. Mutations in dysferlin (dysf) are known to elicit a progressive myopathy in humans, probably due to impaired sarcolemmal repair. We show here that loss of Dysf and annexin A6 (Anxa6) function lead to myopathy in zebrafish. By use of high-resolution imaging of myofibers in intact animals, we reveal sequential phases in sarcolemmal repair. Initially, membrane vesicles enriched in Dysf together with cytoplasmic Anxa6 form a tight patch at the lesion independently of one another. In the subsequent steps, annexin A2a (Anxa2a) followed by annexin A1a (Anxa1a) accumulate at the patch; the recruitment of these annexins depends on Dysf and Anxa6. Thus, sarcolemmal repair relies on the ordered assembly of a protein-membrane scaffold. Moreover, we provide several lines of evidence that the membrane for sarcolemmal repair is derived from a specialized plasma membrane compartment.
Collapse
Affiliation(s)
- Urmas Roostalu
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology and University of Heidelberg, Eggenstein-Leopoldshafen, Germany.
| | | |
Collapse
|
25
|
Non-invasive optical imaging of muscle pathology in mdx mice using cathepsin caged near-infrared imaging. Mol Imaging Biol 2011; 13:462-470. [PMID: 20661652 PMCID: PMC3087873 DOI: 10.1007/s11307-010-0376-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE To develop a reliable live-animal imaging method for monitoring muscle pathology in mouse models of myopathy. PROCEDURES A caged near-infrared Cathepsin B (CTSB) substrate, ProSense 680, is evaluated in the dystrophin deficient mdx mice, a genetic homologue of Duchenne muscular dystrophy via optical imaging. RESULTS We show high levels of infrared signal in dystrophic muscle relative to healthy muscle at 24 h post-injection. Imaging for CTSB presence revealed localization to inflammatory infiltrates and regenerating muscle fibers. A time series myotoxin-induced muscle injury experiment showed that CTSB activity and its mRNA levels peaked at the interface between inflammation and myoblast fusion stage of recovery. Prednisone treatment in mdx mice resulted in decreased CTSB activity and increased grip strength in forelimbs and hindlimbs. CONCLUSIONS Optical imaging of CTSB activity is an ideal method to sensitively monitor inflammation, regeneration, and response to therapy in myopathic skeletal muscle.
Collapse
|
26
|
Guryanova OA, Drazba JA, Frolova EI, Chumakov PM. Actin cytoskeleton remodeling by the alternatively spliced isoform of PDLIM4/RIL protein. J Biol Chem 2011; 286:26849-59. [PMID: 21636573 DOI: 10.1074/jbc.m111.241554] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RIL (product of PDLIM4 gene) is an actin-associated protein that has previously been shown to stimulate actin bundling by interacting with actin-cross-linking protein α-actinin-1 and increasing its affinity to filamentous actin. Here, we report that the alternatively spliced isoform of RIL, denoted here as RILaltCterm, functions as a dominant-negative modulator of RIL-mediated actin reorganization. RILaltCterm is regulated at the level of protein stability, and this protein isoform accumulates particularly in response to oxidative stress. We show that the alternative C-terminal segment of RILaltCterm has a disordered structure that directs the protein to rapid degradation in the core 20 S proteasomes. Such degradation is ubiquitin-independent and can be blocked by binding to NAD(P)H quinone oxidoreductase NQO1, a detoxifying enzyme induced by prolonged exposure to oxidative stress. We show that either overexpression of RILaltCterm or its stabilization by stresses counteracts the effects produced by full-length RIL on organization of actin cytoskeleton and cell motility. Taken together, the data suggest a mechanism for fine-tuning actin cytoskeleton rearrangement in response to stresses.
Collapse
Affiliation(s)
- Olga A Guryanova
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
27
|
172nd ENMC International Workshop: dysferlinopathies 29-31 January 2010, Naarden, The Netherlands. Neuromuscul Disord 2011; 21:503-12. [PMID: 21602046 DOI: 10.1016/j.nmd.2011.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 04/06/2011] [Accepted: 04/15/2011] [Indexed: 11/24/2022]
|
28
|
Barthélémy F, Wein N, Krahn M, Lévy N, Bartoli M. Translational research and therapeutic perspectives in dysferlinopathies. Mol Med 2011; 17:875-82. [PMID: 21556485 DOI: 10.2119/molmed.2011.00084] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/05/2011] [Indexed: 12/13/2022] Open
Abstract
Dysferlinopathies are autosomal recessive disorders caused by mutations in the dysferlin (DYSF) gene, encoding the dysferlin protein. DYSF mutations lead to a wide range of muscular phenotypes, with the most prominent being Miyoshi myopathy (MM) and limb girdle muscular dystrophy type 2B (LGMD2B) and the second most common being LGMD. Symptoms generally appear at the end of childhood and, although disease progression is typically slow, walking impairments eventually result. Dysferlin is a modular type II transmembrane protein for which numerous binding partners have been identified. Although dysferlin function is only partially elucidated, this large protein contains seven calcium sensor C2 domains, shown to play a key role in muscle membrane repair. On the basis of this major function, along with detailed clinical observations, it has been possible to design various therapeutic approaches for dysferlin-deficient patients. Among them, exon-skipping and minigene transfer strategies have been evaluated at the preclinical level and, to date, represent promising approaches for clinical trials. This review aims to summarize the pathophysiology of dysferlinopathies and to evaluate the therapeutic potential for treatments currently under development.
Collapse
Affiliation(s)
- Florian Barthélémy
- University of the Mediterranean, Marseille Medical School, Marseille, France Inserm UMR_S 910 Medical Genetics and Functional Genomics Marseille, France
| | | | | | | | | |
Collapse
|
29
|
Martini P, Risso D, Sales G, Romualdi C, Lanfranchi G, Cagnin S. Statistical Test of Expression Pattern (STEPath): a new strategy to integrate gene expression data with genomic information in individual and meta-analysis studies. BMC Bioinformatics 2011; 12:92. [PMID: 21481242 PMCID: PMC3094239 DOI: 10.1186/1471-2105-12-92] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 04/11/2011] [Indexed: 01/21/2023] Open
Abstract
Background In the last decades, microarray technology has spread, leading to a dramatic increase of publicly available datasets. The first statistical tools developed were focused on the identification of significant differentially expressed genes. Later, researchers moved toward the systematic integration of gene expression profiles with additional biological information, such as chromosomal location, ontological annotations or sequence features. The analysis of gene expression linked to physical location of genes on chromosomes allows the identification of transcriptionally imbalanced regions, while, Gene Set Analysis focuses on the detection of coordinated changes in transcriptional levels among sets of biologically related genes. In this field, meta-analysis offers the possibility to compare different studies, addressing the same biological question to fully exploit public gene expression datasets. Results We describe STEPath, a method that starts from gene expression profiles and integrates the analysis of imbalanced region as an a priori step before performing gene set analysis. The application of STEPath in individual studies produced gene set scores weighted by chromosomal activation. As a final step, we propose a way to compare these scores across different studies (meta-analysis) on related biological issues. One complication with meta-analysis is batch effects, which occur because molecular measurements are affected by laboratory conditions, reagent lots and personnel differences. Major problems occur when batch effects are correlated with an outcome of interest and lead to incorrect conclusions. We evaluated the power of combining chromosome mapping and gene set enrichment analysis, performing the analysis on a dataset of leukaemia (example of individual study) and on a dataset of skeletal muscle diseases (meta-analysis approach). In leukaemia, we identified the Hox gene set, a gene set closely related to the pathology that other algorithms of gene set analysis do not identify, while the meta-analysis approach on muscular disease discriminates between related pathologies and correlates similar ones from different studies. Conclusions STEPath is a new method that integrates gene expression profiles, genomic co-expressed regions and the information about the biological function of genes. The usage of the STEPath-computed gene set scores overcomes batch effects in the meta-analysis approaches allowing the direct comparison of different pathologies and different studies on a gene set activation level.
Collapse
Affiliation(s)
- Paolo Martini
- CRIBI Biotechnology Centre, Department of Biology, University of Padova, via U, Bassi 58/B, 35121 Padova, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Klinge L, Aboumousa A, Eagle M, Hudson J, Sarkozy A, Vita G, Charlton R, Roberts M, Straub V, Barresi R, Lochmüller H, Bushby K. New aspects on patients affected by dysferlin deficient muscular dystrophy. J Neurol Neurosurg Psychiatry 2010; 81:946-53. [PMID: 19528035 PMCID: PMC2975994 DOI: 10.1136/jnnp.2009.178038] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations in the dysferlin gene lead to limb girdle muscular dystrophy 2B, Miyoshi myopathy and distal anterior compartment myopathy. A cohort of 36 patients affected by dysferlinopathy is described, in the first UK study of clinical, genetic, pathological and biochemical data. The diagnosis was established by reduction of dysferlin in the muscle biopsy and subsequent mutational analysis of the dysferlin gene. Seventeen mutations were novel; the majority of mutations were small deletions/insertions, and no mutational hotspots were identified. Sixty-one per cent of patients (22 patients) initially presented with limb girdle muscular dystrophy 2B, 31% (11 patients) with a Miyoshi phenotype, one patient with proximodistal mode of onset, one patient with muscle stiffness after exercise and one patient as a symptomatic carrier. A wider range of age of onset was noted than previously reported, with 25% of patients having first symptoms before the age of 13 years. Independent of the initial mode of presentation, in our cohort of patients the gastrocnemius muscle was the most severely affected muscle leading to an inability to stand on tiptoes, and lower limbs were affected more severely than upper limbs. As previous anecdotal evidence on patients affected by dysferlinopathy suggests good muscle prowess before onset of symptoms, we also investigated pre-symptomatic fitness levels of the patients. Fifty-three per cent of the patients were very active and sporty before the onset of symptoms which makes the clinical course of dysferlinopathy unusual within the different forms of muscular dystrophy and provides a challenge to understanding the underlying pathomechanisms in this disease.
Collapse
Affiliation(s)
- Lars Klinge
- University of Newcastle, Institute of Human Genetics, The Institute of Human Genetics, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rocha CT, Hoffman EP. Limb-girdle and congenital muscular dystrophies: current diagnostics, management, and emerging technologies. Curr Neurol Neurosci Rep 2010; 10:267-76. [PMID: 20467841 DOI: 10.1007/s11910-010-0119-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The muscular dystrophies show muscle degeneration and regeneration (necrotizing myopathy) on muscle biopsy, typically associated with elevated serum creatine kinase, and muscle weakness. In 1986, the first causative gene was identified for the most prevalent and best-characterized form of muscular dystrophy, Duchenne muscular dystrophy. Over the past 25 years, the number of other genes determined to cause different subtypes has grown rapidly. This review gives a synopsis of the 45 genetically defined types of muscular dystrophies and describes the clinical, pathologic, and molecular aspects of each disease. DNA diagnosis remains the most sensitive and specific method for differential diagnosis, but molecular diagnostics can be expensive and complex (because of multiple genes at multiple testing facilities) and reimbursement may be challenging to obtain. However, emerging DNA sequencing technologies (eg, single-molecule third-generation sequencing units) promise to dramatically reduce the complexity and costs of DNA diagnostics. Treatment for nearly all forms remains supportive and is aimed at preventing complications. However, several promising approaches have entered clinical trials, providing tangible hope that quality of life will improve for many patients in the near future.
Collapse
Affiliation(s)
- Carolina Tesi Rocha
- Research Center for Genetic Medicine, Division of Neurology, Department of Pediatrics, Children's National Medical Center, 111 Michigan Avenue Northwest, Washington, DC 20010, USA.
| | | |
Collapse
|
32
|
Role of thrombospondin 1 in macrophage inflammation in dysferlin myopathy. J Neuropathol Exp Neurol 2010; 69:643-53. [PMID: 20467328 DOI: 10.1097/nen.0b013e3181e0d01c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Muscle inflammation can be a prominent feature in several muscular dystrophies. In dysferlin myopathy, it is mainly composed of macrophages. To understand the origin of inflammation in dysferlin-deficient muscle, we analyzed soluble factors involved in monocyte chemotaxis released by myoblasts and myotubes from control and dysferlinopathy patients using a transwell system. Dysferlin-deficient myotubes released more soluble factors involved in monocyte chemotaxis compared with controls (p < 0.001). Messenger RNA microarray analysis showed a 3.2-fold increase of thrombospondin 1 (TSP-1) expression in dysferlin-deficient myotubes. Retrotranscriptasepolymerase chain reaction analysis, ELISA, and immunohistochemistry confirmed these results. Dysferlin mRNA knockdown with short-interfering RNA in normal myogenic cells resulted in TSP-1 mRNA upregulation and increased chemotaxis. Furthermore, monocyte chemotaxis was decreased when TSP-1 was blocked by specific antibodies. In muscle biopsies from dysferlinopathy patients, TSP-1 expression was increased in muscle fibers but not in biopsies of patientswith other myopathies with inflammation; TSP-1 was seen in some macrophages in all samples analyzed. Taken together, the data demonstrate that dysferlin-deficient muscle upregulates TSP-1 in vivoand in vitro and indicate that endogenous chemotactic factors arecrucial to the sustained inflammatory process observed in dysferlinopathies.
Collapse
|
33
|
Rawat R, Cohen TV, Ampong B, Francia D, Henriques-Pons A, Hoffman EP, Nagaraju K. Inflammasome up-regulation and activation in dysferlin-deficient skeletal muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2891-900. [PMID: 20413686 DOI: 10.2353/ajpath.2010.090058] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A deficiency of the dysferlin protein results in limb girdle muscular dystrophy type 2B and Miyoshi myopathy, with resulting plasma membrane abnormalities in myofibers. Many patients show muscle inflammation, but the molecular mechanisms that initiate and perpetuate this inflammation are not well understood. We previously showed abnormal activation of macrophages and hypothesized that activation of the inflammasome pathway may play a role in disease progression. To test this, we studied the inflammasome molecular platform in dysferlin-deficient human and mouse muscle. Consistent with our model, components of the NACHT, LRR and PYD-containing proteins (NALP)-3 inflammasome pathway were specifically up-regulated and activated in dysferlin-deficient but not in dystrophin-deficient and normal muscle. We demonstrate for the first time that normal primary skeletal muscle cells are capable of secreting IL-1beta in response to combined treatment with lipopolysaccharide and the P2X7 receptor agonist, benzylated ATP, suggesting that not only immune cells but also muscle cells can actively participate in inflammasome formation. In addition, we show that dysferlin-deficient primary muscle cells express toll-like receptors (TLRs; TLR-2 and TLR-4) and can efficiently produce IL-1beta in response to lipopolysaccharide and benzylated ATP. These data indicate that skeletal muscle is an active contributor of IL-1beta and strategies that interfere with this pathway may be therapeutically useful for patients with limb girdle muscular dystrophy type 2B.
Collapse
Affiliation(s)
- Rashmi Rawat
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Lostal W, Bartoli M, Bourg N, Roudaut C, Bentaïb A, Miyake K, Guerchet N, Fougerousse F, McNeil P, Richard I. Efficient recovery of dysferlin deficiency by dual adeno-associated vector-mediated gene transfer. Hum Mol Genet 2010; 19:1897-907. [PMID: 20154340 DOI: 10.1093/hmg/ddq065] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Deficiency of the dysferlin protein presents as two major clinical phenotypes: limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. Dysferlin is known to participate in membrane repair, providing a potential hypothesis to the underlying pathophysiology of these diseases. The size of the dysferlin cDNA prevents its direct incorporation into an adeno-associated virus (AAV) vector for therapeutic gene transfer into muscle. To bypass this limitation, we split the dysferlin cDNA at the exon 28/29 junction and cloned it into two independent AAV vectors carrying the appropriate splicing sequences. Intramuscular injection of the corresponding vectors into a dysferlin-deficient mouse model led to the expression of full-length dysferlin for at least 1 year. Importantly, systemic injection in the tail vein of the two vectors led to a widespread although weak expression of the full-length protein. Injections were associated with an improvement of the histological aspect of the muscle, a reduction in the number of necrotic fibers, restoration of membrane repair capacity and a global improvement in locomotor activity. Altogether, these data support the use of such a strategy for the treatment of dysferlin deficiency.
Collapse
Affiliation(s)
- William Lostal
- Généthon, CNRS UMR8587 LAMBE, 1, rue de l'Internationale, 91000 Evry, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Olsen NJ, Prather H, Li QZ, Burns DK. Autoantibody profiles in two patients with non-autoimmune muscle disease implicate a role for gliadin autoreactivity. Neuromuscul Disord 2010; 20:188-91. [PMID: 20110170 DOI: 10.1016/j.nmd.2009.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 11/15/2009] [Accepted: 12/22/2009] [Indexed: 12/25/2022]
Abstract
The objective of this case study was to characterize autoreactivity in two patients with non-autoimmune forms of muscle disease who had positivity for antinuclear antibodies (ANA) and Ro (SSA) autoantibodies. Serum samples from these two patients were applied to an autoantigen protein array with more than 70 specificities and were compared to samples from healthy controls and patients with systemic lupus erythematosus. Both myopathy patients had high levels of gliadin autoreactivity in serum and one patient had an overall autoantibody profile with lupus-like features. The findings suggest that some disorders of muscle that are considered non-autoimmune, may in fact have autoimmune features. Further examination of the role of subclinical gluten autoreactivity in the pathogenesis of myopathy syndromes has the potential to suggest improved approaches to diagnosis and treatment of these conditions.
Collapse
Affiliation(s)
- Nancy J Olsen
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, USA.
| | | | | | | |
Collapse
|
36
|
Roche JA, Lovering RM, Roche R, Ru LW, Reed PW, Bloch RJ. Extensive mononuclear infiltration and myogenesis characterize recovery of dysferlin-null skeletal muscle from contraction-induced injuries. Am J Physiol Cell Physiol 2009; 298:C298-312. [PMID: 19923419 DOI: 10.1152/ajpcell.00122.2009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We studied the response of dysferlin-null and control skeletal muscle to large- and small-strain injuries to the ankle dorsiflexors in mice. We measured contractile torque and counted fibers retaining 10-kDa fluorescein dextran, necrotic fibers, macrophages, and fibers with central nuclei and expressing developmental myosin heavy chain to assess contractile function, membrane resealing, necrosis, inflammation, and myogenesis. We also studied recovery after blunting myogenesis with X-irradiation. We report that dysferlin-null myofibers retain 10-kDa dextran for 3 days after large-strain injury but are lost thereafter, following necrosis and inflammation. Recovery of dysferlin-null muscle requires myogenesis, which delays the return of contractile function compared with controls, which recover from large-strain injury by repairing damaged myofibers without significant inflammation, necrosis, or myogenesis. Recovery of control and dysferlin-null muscles from small-strain injury involved inflammation and necrosis followed by myogenesis, all of which were more pronounced in the dysferlin-null muscles, which recovered more slowly. Both control and dysferlin-null muscles also retained 10-kDa dextran for 3 days after small-strain injury. We conclude that dysferlin-null myofibers can survive contraction-induced injury for at least 3 days but are subsequently eliminated by necrosis and inflammation. Myogenesis to replace lost fibers does not appear to be significantly compromised in dysferlin-null mice.
Collapse
Affiliation(s)
- Joseph A Roche
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
37
|
Millay DP, Maillet M, Roche JA, Sargent MA, McNally EM, Bloch RJ, Molkentin JD. Genetic manipulation of dysferlin expression in skeletal muscle: novel insights into muscular dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1817-23. [PMID: 19834057 DOI: 10.2353/ajpath.2009.090107] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mutations in the gene DYSF, which codes for the protein dysferlin, underlie Miyoshi myopathy and limb-girdle muscular dystrophy 2B in humans and produce a slowly progressing skeletal muscle degenerative disease in mice. Dysferlin is a Ca(2+)-sensing, regulatory protein that is involved in membrane repair after injury. To assess the function of dysferlin in healthy and dystrophic skeletal muscle, we generated skeletal muscle-specific transgenic mice with threefold overexpression of this protein. These mice were phenotypically indistinguishable from wild-type, and more importantly, the transgene completely rescued the muscular dystrophy (MD) disease in Dysf-null A/J mice. The dysferlin transgene rescued all histopathology and macrophage infiltration in skeletal muscle of Dysf(-/-) A/J mice, as well as promoted the rapid recovery of muscle function after forced lengthening contractions. These results indicate that MD in A/J mice is autonomous to skeletal muscle and not initiated by any other cell type. However, overexpression of dysferlin did not improve dystrophic symptoms or membrane instability in the dystrophin-glycoprotein complex-lacking Scgd (delta-sarcoglycan) null mouse, indicating that dysferlin functionality is not a limiting factor underlying membrane repair in other models of MD. In summary, the restoration of dysferlin in skeletal muscle fibers is sufficient to rescue the MD in Dysf-deficient mice, although its mild overexpression does not appear to functionally enhance membrane repair in other models of MD.
Collapse
Affiliation(s)
- Douglas P Millay
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Lang CT, Markham KB, Behrendt NJ, Suarez AA, Samuels P, Vandre DD, Robinson JM, Ackerman WE. Placental dysferlin expression is reduced in severe preeclampsia. Placenta 2009; 30:711-8. [PMID: 19545895 DOI: 10.1016/j.placenta.2009.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 05/11/2009] [Accepted: 05/11/2009] [Indexed: 12/13/2022]
Abstract
Dysferlin (DYSF) and myoferlin (MYOF), members of the ferlin family of membrane proteins, are co-expressed in human placental syncytiotrophoblast (STB). Although the role of these ferlin proteins in the placenta has yet to be established, it has been suggested that DYSF and MYOF may contribute to the stability of the apical STB plasma membrane. The release of STB-derived cellular debris increases in the setting of preeclampsia (PE), suggesting relative destabilization of the hemochorial interface. To test whether PE was associated with alterations in placental expression of DYSF and/or MYOF, a cross-sectional study was performed using specimens of villous placenta collected form women with severe PE (n=10) and normotensive controls (n=10). DYSF and MYOF expression were examined using quantitative real-time RT-PCR, immunoblotting, and immunofluorescence labeling of tissue specimens. Placental DYSF expression was 57% lower at the mRNA level (p=0.03) and 38% lower at the protein level (p=0.026) in severe PE as compared to normotensive subjects. There were no differences in placental MYOF protein or mRNA expression between these groups. No appreciable changes in the distribution of DYSF or MYOF within placental villi was observed in PE relative to control specimens. We conclude that DYSF expression is reduced in severe PE relative to gestational age-matched controls. As DYSF has a role in membrane repair, these data suggest a role for DYSF in the stability of the apical STB plasma membrane and may account, at least in part, for the increased shedding of microparticles from this membrane in PE.
Collapse
Affiliation(s)
- C T Lang
- Department of Obstetrics & Gynecology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|