151
|
Dominov JA, Uyan Ö, McKenna‐Yasek D, Nallamilli BRR, Kergourlay V, Bartoli M, Levy N, Hudson J, Evangelista T, Lochmuller H, Krahn M, Rufibach L, Hegde M, Brown RH. Correction of pseudoexon splicing caused by a novel intronic dysferlin mutation. Ann Clin Transl Neurol 2019; 6:642-654. [PMID: 31019989 PMCID: PMC6469257 DOI: 10.1002/acn3.738] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 01/12/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Dysferlin is a large transmembrane protein that functions in critical processes of membrane repair and vesicle fusion. Dysferlin-deficiency due to mutations in the dysferlin gene leads to muscular dystrophy (Miyoshi myopathy (MM), limb girdle muscular dystrophy type 2B (LGMD2B), distal myopathy with anterior tibial onset (DMAT)), typically with early adult onset. At least 416 pathogenic dysferlin mutations are known, but for approximately 17% of patients, one or both of their pathogenic variants remain undefined following standard exon sequencing methods that interrogate exons and nearby flanking intronic regions but not the majority of intronic regions. METHODS We sequenced RNA from myogenic cells to identify a novel dysferlin pathogenic variant in two affected siblings that previously had only one disease-causing variant identified. We designed antisense oligonucleotides (AONs) to bypass the effects of this mutation on RNA splicing. RESULTS We identified a new pathogenic point mutation deep within dysferlin intron 50i. This intronic variant causes aberrant mRNA splicing and inclusion of an additional pseudoexon (PE, we term PE50.1) within the mature dysferlin mRNA. PE50.1 inclusion alters the protein sequence, causing premature translation termination. We identified this mutation in 23 dysferlinopathy patients (seventeen families), revealing it to be one of the more prevalent dysferlin mutations. We used AON-mediated exon skipping to correct the aberrant PE50.1 splicing events in vitro, which increased normal mRNA production and significantly restored dysferlin protein expression. INTERPRETATION Deep intronic mutations can be a common underlying cause of dysferlinopathy, and importantly, could be treatable with AON-based exon-skipping strategies.
Collapse
Affiliation(s)
- Janice A. Dominov
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Özgün Uyan
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Diane McKenna‐Yasek
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Babi Ramesh Reddy Nallamilli
- Department of Human GeneticsEmory University School of MedicineAtlantaGeorgia
- Present address:
Perkin Elmer GenomicsWalthamMassachusetts
| | - Virginie Kergourlay
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
| | - Marc Bartoli
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
| | - Nicolas Levy
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
- Département de Génétique MédicaleAPHMHôpital Timone EnfantsMarseilleFrance
| | - Judith Hudson
- Northern Molecular Genetics ServiceNewcastle upon TyneUnited Kingdom
| | - Teresinha Evangelista
- Newcastle University John Walton Centre for Muscular Dystrophy ResearchMRC Centre for Neuromuscular DiseasesInstitute of Genetic MedicineNewcastle upon TyneUnited Kingdom
| | - Hanns Lochmuller
- Newcastle University John Walton Centre for Muscular Dystrophy ResearchMRC Centre for Neuromuscular DiseasesInstitute of Genetic MedicineNewcastle upon TyneUnited Kingdom
- Department of Neuropediatrics and Muscle DisordersFaculty of MedicineMedical Center–University of FreiburgFreiburgGermany
- Centro Nacional de Análisis Genómico (CNAG‐CRG)Center for Genomic RegulationBarcelona Institute of Science and Technology (BIST)BarcelonaCataloniaSpain
- Children's Hospital of Eastern Ontario Research InstituteUniversity of OttawaOttawaCanada
- Division of NeurologyDepartment of MedicineThe Ottawa HospitalOttawaCanada
| | - Martin Krahn
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
- Département de Génétique MédicaleAPHMHôpital Timone EnfantsMarseilleFrance
| | | | - Madhuri Hegde
- Department of Human GeneticsEmory University School of MedicineAtlantaGeorgia
| | - Robert H. Brown
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| |
Collapse
|
152
|
Nachun D, Gao F, Isaacs C, Strawser C, Yang Z, Dokuru D, Van Berlo V, Sears R, Farmer J, Perlman S, Lynch DR, Coppola G. Peripheral blood gene expression reveals an inflammatory transcriptomic signature in Friedreich's ataxia patients. Hum Mol Genet 2019; 27:2965-2977. [PMID: 29790959 PMCID: PMC6097013 DOI: 10.1093/hmg/ddy198] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Transcriptional changes in Friedreich's ataxia (FRDA), a rare and debilitating recessive Mendelian neurodegenerative disorder, have been studied in affected but inaccessible tissues-such as dorsal root ganglia, sensory neurons and cerebellum-in animal models or small patient series. However, transcriptional changes induced by FRDA in peripheral blood, a readily accessible tissue, have not been characterized in a large sample. We used differential expression, association with disability stage, network analysis and enrichment analysis to characterize the peripheral blood transcriptome and identify genes that were differentially expressed in FRDA patients (n = 418) compared with both heterozygous expansion carriers (n = 228) and controls (n = 93 739 individuals in total), or were associated with disease progression, resulting in a disease signature for FRDA. We identified a transcriptional signature strongly enriched for an inflammatory innate immune response. Future studies should seek to further characterize the role of peripheral inflammation in FRDA pathology and determine its relevance to overall disease progression.
Collapse
Affiliation(s)
- Daniel Nachun
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fuying Gao
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Charles Isaacs
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Zhongan Yang
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Deepika Dokuru
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Victoria Van Berlo
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Renee Sears
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Susan Perlman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - David R Lynch
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giovanni Coppola
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
153
|
Barzilai-Tutsch H, Dewulf M, Lamaze C, Butler Browne G, Pines M, Halevy O. A promotive effect for halofuginone on membrane repair and synaptotagmin-7 levels in muscle cells of dysferlin-null mice. Hum Mol Genet 2019; 27:2817-2829. [PMID: 29771357 DOI: 10.1093/hmg/ddy185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/09/2018] [Indexed: 11/14/2022] Open
Abstract
In the absence of dysferlin, skeletal muscle cells fail to reseal properly after injury, resulting in slow progress of the dysferlinopathy muscular dystrophy (MD). Halofuginone, a leading agent in preventing fibrosis in MDs, was tested for its effects on membrane resealing post-injury. A hypo-osmotic shock assay on myotubes derived from wild-type (Wt) and dysferlin-null (dysf-/-) mice revealed that pre-treatment with halofuginone reduces the percentage of membrane-ruptured myotubes only in dysf-/- myotubes. In laser-induced injury of isolated myofibers, halofuginone decreased the amount of FM1-43 at the injury site of dysf-/- myofibers while having no effect on Wt myofibers. These results implicate halofuginone in ameliorating muscle-cell membrane integrity in dysf-/- mice. Halofuginone increased lysosome scattering across the cytosol of dysf-/- primary myoblasts, in a protein kinase/extracellular signal-regulated protein kinase and phosphoinositide 3 kinase/Akt-dependent manner, in agreement with an elevation in lysosomal exocytotic activity in these cells. A spatial- and age-dependent synaptotagmin-7 (Syt-7) expression pattern was shown in dysf-/- versus Wt mice, suggesting that these pattern alterations are related to the disease progress and that sytnaptotagmin-7 may be compensating for the lack of dysferlin at least with regard to membrane resealing post-injury. While halofuginone did not affect patch-repair-complex key proteins, it further enhanced Syt-7 levels and its spread across the cytosol in dysf-/- myofibers and muscle tissue, and increased its co-localization with lysosomes. Together, the data imply a novel role for halofuginone in membrane-resealing events with Syt-7 possibly taking part in these events.
Collapse
Affiliation(s)
- Hila Barzilai-Tutsch
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Melissa Dewulf
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Christophe Lamaze
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Gillian Butler Browne
- Center for Research in Myology, CNRS FRE 3617, UPMC Univ Paris 06, UM76, INSERM U974, Sorbonne Universités, Paris, France
| | - Mark Pines
- The Volcani Center, Institute of Animal Science, Bet Dagan 52505, Israel
| | - Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
154
|
Obrova K, Cyrklaff M, Frank R, Mair GR, Mueller AK. Transmission of the malaria parasite requires ferlin for gamete egress from the red blood cell. Cell Microbiol 2019; 21:e12999. [PMID: 30597708 DOI: 10.1111/cmi.12999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/15/2018] [Accepted: 12/09/2018] [Indexed: 02/02/2023]
Abstract
Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.
Collapse
Affiliation(s)
- Klara Obrova
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Roland Frank
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Gunnar R Mair
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Ann-Kristin Mueller
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infectious Diseases (DZIF), Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
155
|
Algahtani H, Shirah B, Alassiri AH, Habib BA, Almuhanna R, Ahamed MF. Limb-girdle muscular dystrophy type 2B: An unusual cause of proximal muscular weakness in Saudi Arabia. J Back Musculoskelet Rehabil 2019; 31:999-1004. [PMID: 29966189 DOI: 10.3233/bmr-181129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dysferlinopathies encompass a group of neuromuscular diseases characterized by the absence of dysferlin in skeletal muscle. It is a genetic disorder caused by a mutation in the dysferlin gene (DYSF) with an autosomal recessive mode of inheritance. In this article, we report a case of Limb-girdle muscular dystrophy type 2B with a rare homozygous duplication c.164dupA, p.(Ile57Hisfs*8) (rs863225020) in DYSF in a Saudi patient. To the best of our knowledge, this is the first case from Saudi Arabia with complete clinical data, pathology findings, radiology findings, and genetic analysis. Although there is no curative treatment for this disease, an accurate diagnosis is important to avoid using steroids and immunosuppressive medications, which are not effective and may have several side effects. Further studies are needed to explore potential therapies for this rare condition.
Collapse
Affiliation(s)
- Hussein Algahtani
- King Abdulaziz Medical City, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Bader Shirah
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Ali H Alassiri
- Department of Pathology and Lab Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Ben Attia Habib
- Department of Pathology and Lab Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Rakan Almuhanna
- Department of Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | | |
Collapse
|
156
|
Gómez-Andrés D, Díaz J, Munell F, Sánchez-Montáñez Á, Pulido-Valdeolivas I, Suazo L, Garrido C, Quijano-Roy S, Bevilacqua JA. Disease duration and disability in dysfeRlinopathy can be described by muscle imaging using heatmaps and random forests. Muscle Nerve 2019; 59:436-444. [PMID: 30578674 DOI: 10.1002/mus.26403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/25/2018] [Accepted: 12/16/2018] [Indexed: 11/09/2022]
Abstract
INTRODUCTION The manner in which imaging patterns change over the disease course and with increasing disability in dysferlinopathy is not fully understood. METHODS Fibroadipose infiltration of 61 muscles was scored based on whole-body MRI of 33 patients with dysferlinopathy and represented in a heatmap. We trained random forests to predict disease duration, Motor Function Measure dimension 1 (MFM-D1), and modified Rankin scale (MRS) score based on muscle scoring and selected the most important muscle for predictions. RESULTS The heatmap delineated positive and negative fingerprints in dysferlinopathy. Disease duration was related to infiltration of infraspinatus, teres major-minor, and supraspinatus muscles. MFM-D1 decreased with higher infiltration of teres major-minor, triceps, and sartorius. MRS related to infiltration of vastus medialis, gracilis, infraspinatus, and sartorius. DISCUSSION Dysferlinopathy shows a recognizable muscle MRI pattern. Fibroadipose infiltration in specific muscles of the thigh and the upper limb appears to be an important marker for disease progression. Muscle Nerve 59:436-444, 2019.
Collapse
Affiliation(s)
- David Gómez-Andrés
- Paediatric Neurology, Vall d'Hebron University Hospital and VHIR (Euro-NMD, ERN-RND), Barcelona, Spain
| | - Jorge Díaz
- Medical Imaging Center, University of Chile Clinical Hospital, Santiago, Chile
| | - Francina Munell
- Paediatric Neurology, Vall d'Hebron University Hospital and VHIR (Euro-NMD, ERN-RND), Barcelona, Spain
| | - Ángel Sánchez-Montáñez
- Paediatric Neuroradiology, Vall d'Hebron University Hospital (Euro-NMD, ERN-RND), Barcelona, Spain
| | - Irene Pulido-Valdeolivas
- Visual Pathway Laboratory, Neuroimmunology Center and Neurology Department, Biomedical Research Center August Pi i Sunyer (IDIBAPS), Hospital Clínic Barcelona, Spain
| | - Lionel Suazo
- Medical Imaging Center, University of Chile Clinical Hospital, Santiago, Chile
| | - Cristián Garrido
- Medical Imaging Center, University of Chile Clinical Hospital, Santiago, Chile
| | - Susana Quijano-Roy
- APHP-Neurology and Intensive Care Department. University Hospital Raymond Poincaré, Garches, U1179 Versailles University, Neuromuscular Disorders Reference Center of Nord-Est-Île de France, ERN Neuro-NMD, France
| | - Jorge A Bevilacqua
- Neuromuscular Unit, Department of Neurology and Neurosurgery, University of Chile Clinical Hospital.,Department of Anatomy and Legal Medicine, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
157
|
Takahashi K. Effects of Prednisone on a Patient with Dysferlinopathy Assessed by Maximal Voluntary Isometric Contraction: Alternate-Day Low-Dose Administration for a 17-Year Period. Case Rep Neurol 2019; 11:10-16. [PMID: 31043956 PMCID: PMC6477497 DOI: 10.1159/000495746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/26/2018] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids are candidates for the pharmacological treatment of dysferlinopathy. Deflazacort, however, showed a worse effect on muscle strength than placebo. Alternate-day low-dose prednisone may have beneficial effects with fewer adverse effects. The outcomes for a female patient with dysferlinopathy (limb-girdle muscular dystrophy type 2B) were assessed by maximal voluntary isometric contraction (MVIC) using a newly devised chair and arm table with push-pull type strain gauges. Grip strength was also measured isometrically. Prednisone 15 mg was started orally at the age of 24 years and was taken every other day in the morning until 41 years of age. The MVIC of flexion of the knees and elbows increased gradually and significantly. The MVIC of extension of the knees and elbows increased to a lesser extent. Isometric grip strength showed no remarkable increase, but strength was sustained over 10 years. Muscle fiber types account for these differences. The beneficial effects of alternate-day prednisone treatment on dysferlinopathy are reported.
Collapse
Affiliation(s)
- Keiichi Takahashi
- Department of Neurology, Takahashi Clinic for Neurological Disorders, Sanda, Japan
| |
Collapse
|
158
|
|
159
|
Nallamilli BRR, Chakravorty S, Kesari A, Tanner A, Ankala A, Schneider T, da Silva C, Beadling R, Alexander JJ, Askree SH, Whitt Z, Bean L, Collins C, Khadilkar S, Gaitonde P, Dastur R, Wicklund M, Mozaffar T, Harms M, Rufibach L, Mittal P, Hegde M. Genetic landscape and novel disease mechanisms from a large LGMD cohort of 4656 patients. Ann Clin Transl Neurol 2018; 5:1574-1587. [PMID: 30564623 PMCID: PMC6292381 DOI: 10.1002/acn3.649] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
Objective Limb‐girdle muscular dystrophies (LGMDs), one of the most heterogeneous neuromuscular disorders (NMDs), involves predominantly proximal‐muscle weakness with >30 genes associated with different subtypes. The clinical‐genetic overlap among subtypes and with other NMDs complicate disease‐subtype identification lengthening diagnostic process, increases overall costs hindering treatment/clinical‐trial recruitment. Currently seven LGMD clinical trials are active but still no gene‐therapy‐related treatment is available. Till‐date no nation‐wide large‐scale LGMD sequencing program was performed. Our objectives were to understand LGMD genetic basis, different subtypes’ relative prevalence across US and investigate underlying disease mechanisms. Methods A total of 4656 patients with clinically suspected‐LGMD across US were recruited to conduct next‐generation sequencing (NGS)‐based gene‐panel testing during June‐2015 to June‐2017 in CLIA‐CAP‐certified Emory‐Genetics‐Laboratory. Thirty‐five LGMD‐subtypes‐associated or LGMD‐like other NMD‐associated genes were investigated. Main outcomes were diagnostic yield, gene‐variant spectrum, and LGMD subtypes’ prevalence in a large US LGMD‐suspected population. Results Molecular diagnosis was established in 27% (1259 cases; 95% CI, 26–29%) of the patients with major contributing genes to LGMD phenotypes being: CAPN3(17%), DYSF(16%), FKRP(9%) and ANO5(7%). We observed an increased prevalence of genetically confirmed late‐onset Pompe disease, DNAJB6‐associated LGMD subtype1E and CAPN3‐associated autosomal‐dominant LGMDs. Interestingly, we identified a high prevalence of patients with pathogenic variants in more than one LGMD gene suggesting possible synergistic heterozygosity/digenic/multigenic contribution to disease presentation/progression that needs consideration as a part of diagnostic modality. Interpretation Overall, this study has improved our understanding of the relative prevalence of different LGMD subtypes, their respective genetic etiology, and the changing paradigm of their inheritance modes and novel mechanisms that will allow for improved timely treatment, management, and enrolment of molecularly diagnosed individuals in clinical trials.
Collapse
Affiliation(s)
| | | | - Akanchha Kesari
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | - Alice Tanner
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | - Arunkanth Ankala
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | | | | | | | - John J Alexander
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | - Syed Hussain Askree
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | - Zachary Whitt
- Emory University Department of Human Genetics Atlanta Georgia 30322.,Augusta University Augusta Georgia 30912
| | - Lora Bean
- Emory University Department of Human Genetics Atlanta Georgia 30322.,EGL Genetics-Eurofins Tucker Atlanta Georgia 30084
| | - Christin Collins
- Emory University Department of Human Genetics Atlanta Georgia 30322
| | - Satish Khadilkar
- Department of Neurology Bombay Hospital Mumbai Maharashtra India.,Department of Neurology Sir J J Group of Hospitals Grant Medical College Mumbai Maharashtra India
| | - Pradnya Gaitonde
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders (CAMDND) 400022 Mumbai India
| | - Rashna Dastur
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders (CAMDND) 400022 Mumbai India
| | - Matthew Wicklund
- Neurology The University of Colorado at Denver - Anschutz Medical Campus Aurora Colorado 80045
| | - Tahseen Mozaffar
- Neurology University of California, Irvine Orange California 92868
| | - Matthew Harms
- Department of Neurology Columbia University New York New York 10032
| | | | | | - Madhuri Hegde
- Emory University Department of Human Genetics Atlanta Georgia 30322
| |
Collapse
|
160
|
Milone M, Liewluck T. The unfolding spectrum of inherited distal myopathies. Muscle Nerve 2018; 59:283-294. [PMID: 30171629 DOI: 10.1002/mus.26332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/26/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Abstract
Distal myopathies are a group of rare muscle diseases characterized by distal weakness at onset. Although acquired myopathies can occasionally present with distal weakness, the majority of distal myopathies have a genetic etiology. Their age of onset varies from early-childhood to late-adulthood while the predominant muscle weakness can affect calf, ankle dorsiflexor, or distal upper limb muscles. A spectrum of muscle pathological changes, varying from nonspecific myopathic changes to rimmed vacuoles to myofibrillar pathology to nuclei centralization, have been noted. Likewise, the underlying molecular defect is heterogeneous. In addition, there is emerging evidence that distal myopathies can result from defective proteins encoded by genes causative of neurogenic disorders, be manifestation of multisystem proteinopathies or the result of the altered interplay between different genes. In this review, we provide an overview on the clinical, electrophysiological, pathological, and molecular aspects of distal myopathies, focusing on the most recent developments in the field. Muscle Nerve 59:283-294, 2019.
Collapse
Affiliation(s)
| | - Teerin Liewluck
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
161
|
Eidem HR, Steenwyk JL, Wisecaver JH, Capra JA, Abbot P, Rokas A. integRATE: a desirability-based data integration framework for the prioritization of candidate genes across heterogeneous omics and its application to preterm birth. BMC Med Genomics 2018; 11:107. [PMID: 30453955 PMCID: PMC6245874 DOI: 10.1186/s12920-018-0426-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/07/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The integration of high-quality, genome-wide analyses offers a robust approach to elucidating genetic factors involved in complex human diseases. Even though several methods exist to integrate heterogeneous omics data, most biologists still manually select candidate genes by examining the intersection of lists of candidates stemming from analyses of different types of omics data that have been generated by imposing hard (strict) thresholds on quantitative variables, such as P-values and fold changes, increasing the chance of missing potentially important candidates. METHODS To better facilitate the unbiased integration of heterogeneous omics data collected from diverse platforms and samples, we propose a desirability function framework for identifying candidate genes with strong evidence across data types as targets for follow-up functional analysis. Our approach is targeted towards disease systems with sparse, heterogeneous omics data, so we tested it on one such pathology: spontaneous preterm birth (sPTB). RESULTS We developed the software integRATE, which uses desirability functions to rank genes both within and across studies, identifying well-supported candidate genes according to the cumulative weight of biological evidence rather than based on imposition of hard thresholds of key variables. Integrating 10 sPTB omics studies identified both genes in pathways previously suspected to be involved in sPTB as well as novel genes never before linked to this syndrome. integRATE is available as an R package on GitHub ( https://github.com/haleyeidem/integRATE ). CONCLUSIONS Desirability-based data integration is a solution most applicable in biological research areas where omics data is especially heterogeneous and sparse, allowing for the prioritization of candidate genes that can be used to inform more targeted downstream functional analyses.
Collapse
Affiliation(s)
- Haley R. Eidem
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| | - Jacob L. Steenwyk
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| | - Jennifer H. Wisecaver
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
- Department of Biochemistry, Purdue University, West Lafayette, IN USA
| | - John A. Capra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| | - Patrick Abbot
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
| |
Collapse
|
162
|
Lee JJA, Maruyama R, Duddy W, Sakurai H, Yokota T. Identification of Novel Antisense-Mediated Exon Skipping Targets in DYSF for Therapeutic Treatment of Dysferlinopathy. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:596-604. [PMID: 30439648 PMCID: PMC6234522 DOI: 10.1016/j.omtn.2018.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Abstract
Dysferlinopathy is a progressive myopathy caused by mutations in the dysferlin (DYSF) gene. Dysferlin protein plays a major role in plasma-membrane resealing. Some patients with DYSF deletion mutations exhibit mild symptoms, suggesting some regions of DYSF can be removed without significantly impacting protein function. Antisense-mediated exon-skipping therapy uses synthetic molecules called antisense oligonucleotides to modulate splicing, allowing exons harboring or near genetic mutations to be removed and the open reading frame corrected. Previous studies have focused on DYSF exon 32 skipping as a potential therapeutic approach, based on the association of a mild phenotype with the in-frame deletion of exon 32. To date, no other DYSF exon-skipping targets have been identified, and the relationship between DYSF exon deletion pattern and protein function remains largely uncharacterized. In this study, we utilized a membrane-wounding assay to evaluate the ability of plasmid constructs carrying mutant DYSF, as well as antisense oligonucleotides, to rescue membrane resealing in patient cells. We report that multi-exon skipping of DYSF exons 26–27 and 28–29 rescues plasma-membrane resealing. Successful translation of these findings into the development of clinical antisense drugs would establish new therapeutic approaches that would be applicable to ∼5%–7% (exons 26–27 skipping) and ∼8% (exons 28–29 skipping) of dysferlinopathy patients worldwide.
Collapse
Affiliation(s)
- Joshua J A Lee
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, United Kingdom
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada; The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
163
|
Nakamura M, Dominguez ANM, Decker JR, Hull AJ, Verboon JM, Parkhurst SM. Into the breach: how cells cope with wounds. Open Biol 2018; 8:rsob.180135. [PMID: 30282661 PMCID: PMC6223217 DOI: 10.1098/rsob.180135] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022] Open
Abstract
Repair of wounds to individual cells is crucial for organisms to survive daily physiological or environmental stresses, as well as pathogen assaults, which disrupt the plasma membrane. Sensing wounds, resealing membranes, closing wounds and remodelling plasma membrane/cortical cytoskeleton are four major steps that are essential to return cells to their pre-wounded states. This process relies on dynamic changes of the membrane/cytoskeleton that are indispensable for carrying out the repairs within tens of minutes. Studies from different cell wound repair models over the last two decades have revealed that the molecular mechanisms of single cell wound repair are very diverse and dependent on wound type, size, and/or species. Interestingly, different repair models have been shown to use similar proteins to achieve the same end result, albeit sometimes by distinctive mechanisms. Recent studies using cutting edge microscopy and molecular techniques are shedding new light on the molecular mechanisms during cellular wound repair. Here, we describe what is currently known about the mechanisms underlying this repair process. In addition, we discuss how the study of cellular wound repair—a powerful and inducible model—can contribute to our understanding of other fundamental biological processes such as cytokinesis, cell migration, cancer metastasis and human diseases.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Andrew N M Dominguez
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jacob R Decker
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Alexander J Hull
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey M Verboon
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
164
|
Diaz-Manera J, Fernandez-Torron R, LLauger J, James MK, Mayhew A, Smith FE, Moore UR, Blamire AM, Carlier PG, Rufibach L, Mittal P, Eagle M, Jacobs M, Hodgson T, Wallace D, Ward L, Smith M, Stramare R, Rampado A, Sato N, Tamaru T, Harwick B, Rico Gala S, Turk S, Coppenrath EM, Foster G, Bendahan D, Le Fur Y, Fricke ST, Otero H, Foster SL, Peduto A, Sawyer AM, Hilsden H, Lochmuller H, Grieben U, Spuler S, Tesi Rocha C, Day JW, Jones KJ, Bharucha-Goebel DX, Salort-Campana E, Harms M, Pestronk A, Krause S, Schreiber-Katz O, Walter MC, Paradas C, Hogrel JY, Stojkovic T, Takeda S, Mori-Yoshimura M, Bravver E, Sparks S, Bello L, Semplicini C, Pegoraro E, Mendell JR, Bushby K, Straub V. Muscle MRI in patients with dysferlinopathy: pattern recognition and implications for clinical trials. J Neurol Neurosurg Psychiatry 2018; 89:1071-1081. [PMID: 29735511 PMCID: PMC6166612 DOI: 10.1136/jnnp-2017-317488] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/02/2018] [Accepted: 03/26/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND OBJECTIVE Dysferlinopathies are a group of muscle disorders caused by mutations in the DYSF gene. Previous muscle imaging studies describe a selective pattern of muscle involvement in smaller patient cohorts, but a large imaging study across the entire spectrum of the dysferlinopathies had not been performed and previous imaging findings were not correlated with functional tests. METHODS We present cross-sectional T1-weighted muscle MRI data from 182 patients with genetically confirmed dysferlinopathies. We have analysed the pattern of muscles involved in the disease using hierarchical analysis and presented it as heatmaps. Results of the MRI scans have been correlated with relevant functional tests for each region of the body analysed. RESULTS In 181 of the 182 patients scanned, we observed muscle pathology on T1-weighted images, with the gastrocnemius medialis and the soleus being the most commonly affected muscles. A similar pattern of involvement was identified in most patients regardless of their clinical presentation. Increased muscle pathology on MRI correlated positively with disease duration and functional impairment. CONCLUSIONS The information generated by this study is of high diagnostic value and important for clinical trial development. We have been able to describe a pattern that can be considered as characteristic of dysferlinopathy. We have defined the natural history of the disease from a radiological point of view. These results enabled the identification of the most relevant regions of interest for quantitative MRI in longitudinal studies, such as clinical trials. CLINICAL TRIAL REGISTRATION NCT01676077.
Collapse
Affiliation(s)
- Jordi Diaz-Manera
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain.,Neuromuscular Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Roberto Fernandez-Torron
- Neuromuscular Area, Biodonostia Health Research Institute, Neurology Service, Donostia University Hospital, Donostia-San Sebastian, Spain.,The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Jaume LLauger
- Radiology Department, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Fiona E Smith
- Magnetic Resonance Centre, Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ursula R Moore
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Andrew M Blamire
- Magnetic Resonance Centre, Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Pierre G Carlier
- AIM & CEA NMR Laboratory, Institute of Myology, Pitié-Salpêtrière University Hospital, Paris, France
| | | | | | - Michelle Eagle
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Marni Jacobs
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, District of Columbia, USA.,Department of Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, District of Columbia, USA
| | - Tim Hodgson
- Magnetic Resonance Centre, Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Dorothy Wallace
- Magnetic Resonance Centre, Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Louise Ward
- Magnetic Resonance Centre, Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Smith
- Department of Radiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Roberto Stramare
- Radiology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Alessandro Rampado
- Radiology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Noriko Sato
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takeshi Tamaru
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Bruce Harwick
- Department of Radiology, CMC Mercy Charlotte, Carolinas Healthcare System Neurosciences Institute, Charlotte, North Carolina, USA
| | - Susana Rico Gala
- Department of Radiology, Hospital U. Virgen de Valme, Sevilla, Spain
| | - Suna Turk
- AIM & CEA NMR Laboratory, Institute of Myology, Pitié-Salpêtrière University Hospital, Paris, France
| | - Eva M Coppenrath
- Department of Clinical Radiology, Ludwig-Maximilians-University, Munich, Germany
| | - Glenn Foster
- Center for Clinical Imaging Research CCIR, Washington University, St. Louis, Missouri, USA
| | - David Bendahan
- Centre de Résonance, Magnétique Biologique et Médicale, Marseille, France.,Aix-Marseille Université, Marseille, France
| | | | - Stanley T Fricke
- Department of Diagnostic Imaging and Radiology, Children's National Health System, Washington, District of Columbia, USA
| | - Hansel Otero
- Department of Diagnostic Imaging and Radiology, Children's National Health System, Washington, District of Columbia, USA
| | - Sheryl L Foster
- Department of Radiology, Westmead Hospital, Westmead, New South Wales, Australia.,Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Anthony Peduto
- Department of Radiology, Westmead Hospital, Westmead, New South Wales, Australia.,Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Anne Marie Sawyer
- Lucas Center for Imaging, Stanford University School of Medicine, Stanford, California, USA
| | - Heather Hilsden
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Hanns Lochmuller
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Ulrike Grieben
- Charite Muscle Research Unit, Experimental and Clinical Research Center, A Joint Co-operation of the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Charite Muscle Research Unit, Experimental and Clinical Research Center, A Joint Co-operation of the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Carolina Tesi Rocha
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - John W Day
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Kristi J Jones
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Sydney, New South Wales, Australia
| | - Diana X Bharucha-Goebel
- Department of Neurology, Children's National Health System, Washington, District of Columbia, USA.,National Institutes of Health (NINDS), Bethesda, Maryland, USA
| | | | - Matthew Harms
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alan Pestronk
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sabine Krause
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Olivia Schreiber-Katz
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Hospital U. Virgen del Rocío/Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Jean-Yves Hogrel
- Institut de Myologie, AP-HP, G.H. Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Tanya Stojkovic
- Institut de Myologie, AP-HP, G.H. Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Shin'ichi Takeda
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Elena Bravver
- Neurosciences Institute, Carolinas Healthcare System, Charlotte, North Carolina, USA
| | - Susan Sparks
- Neurosciences Institute, Carolinas Healthcare System, Charlotte, North Carolina, USA
| | - Luca Bello
- Department of Neurosciences, University of Padova, Padova, Italy
| | | | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova, Italy
| | | | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle upon Tyne, UK
| | | |
Collapse
|
165
|
Quinn C, Moore SA, Bardakjian TM, Karam C. Clinical Reasoning: A 30-year-old man with progressive weakness and atrophy. Neurology 2018; 87:e227-e230. [PMID: 27821570 DOI: 10.1212/wnl.0000000000003320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Colin Quinn
- From the Department of Neurology (C.Q., T.M.B.), University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA; Department of Pathology (S.A.M.), The University of Iowa, Carver College of Medicine, Iowa City, IA; and Department of Neurology (C.K.), Oregon Health and Science University, Portland, OR.
| | - Steven A Moore
- From the Department of Neurology (C.Q., T.M.B.), University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA; Department of Pathology (S.A.M.), The University of Iowa, Carver College of Medicine, Iowa City, IA; and Department of Neurology (C.K.), Oregon Health and Science University, Portland, OR
| | - Tanya M Bardakjian
- From the Department of Neurology (C.Q., T.M.B.), University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA; Department of Pathology (S.A.M.), The University of Iowa, Carver College of Medicine, Iowa City, IA; and Department of Neurology (C.K.), Oregon Health and Science University, Portland, OR
| | - Chafic Karam
- From the Department of Neurology (C.Q., T.M.B.), University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA; Department of Pathology (S.A.M.), The University of Iowa, Carver College of Medicine, Iowa City, IA; and Department of Neurology (C.K.), Oregon Health and Science University, Portland, OR
| |
Collapse
|
166
|
Abstract
The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
167
|
De Luna N, Suarez-Calvet X, Garicano M, Fernandez-Simon E, Rojas-García R, Diaz-Manera J, Querol L, Illa I, Gallardo E. Effect of MAPK Inhibition on the Differentiation of a Rhabdomyosarcoma Cell Line Combined With CRISPR/Cas9 Technology: An In Vitro Model of Human Muscle Diseases. J Neuropathol Exp Neurol 2018; 77:964-972. [DOI: 10.1093/jnen/nly078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Noemí De Luna
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Xavier Suarez-Calvet
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Maialen Garicano
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Esther Fernandez-Simon
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Ricardo Rojas-García
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Jordi Diaz-Manera
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Luis Querol
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Isabel Illa
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| | - Eduard Gallardo
- Department of Neuromuscular Diseases Laboratory, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Raras
| |
Collapse
|
168
|
Sreetama SC, Chandra G, Van der Meulen JH, Ahmad MM, Suzuki P, Bhuvanendran S, Nagaraju K, Hoffman EP, Jaiswal JK. Membrane Stabilization by Modified Steroid Offers a Potential Therapy for Muscular Dystrophy Due to Dysferlin Deficit. Mol Ther 2018; 26:2231-2242. [PMID: 30166241 PMCID: PMC6127637 DOI: 10.1016/j.ymthe.2018.07.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 07/15/2018] [Accepted: 07/24/2018] [Indexed: 11/16/2022] Open
Abstract
Mutations of the DYSF gene leading to reduced dysferlin protein level causes limb girdle muscular dystrophy type 2B (LGMD2B). Dysferlin facilitates sarcolemmal membrane repair in healthy myofibers, thus its deficit compromises myofiber repair and leads to chronic muscle inflammation. An experimental therapeutic approach for LGMD2B is to protect damage or improve repair of myofiber sarcolemma. Here, we compared the effects of prednisolone and vamorolone (a dissociative steroid; VBP15) on dysferlin-deficient myofiber repair. Vamorolone, but not prednisolone, stabilized dysferlin-deficient muscle cell membrane and improved repair of dysferlin-deficient mouse (B6A/J) myofibers injured by focal sarcolemmal damage, eccentric contraction-induced injury or injury due to spontaneous in vivo activity. Vamorolone decreased sarcolemmal lipid mobility, increased muscle strength, and decreased late-stage myofiber loss due to adipogenic infiltration. In contrast, the conventional glucocorticoid prednisolone failed to stabilize dysferlin deficient muscle cell membrane or improve repair of dysferlinopathic patient myoblasts and mouse myofibers. Instead, prednisolone treatment increased muscle weakness and myofiber atrophy in B6A/J mice—findings that correlate with reports of prednisolone worsening symptoms of LGMD2B patients. Our findings showing improved cellular and pre-clinical efficacy of vamorolone compared to prednisolone and better safety profile of vamorolone indicates the suitability of vamorolone for clinical trials in LGMD2B.
Collapse
Affiliation(s)
- Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Jack H Van der Meulen
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Mohammad Mahad Ahmad
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Peter Suzuki
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Shivaprasad Bhuvanendran
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA.
| |
Collapse
|
169
|
Zhou L, Middel V, Reischl M, Strähle U, Nienhaus GU. Distinct amino acid motifs carrying multiple positive charges regulate membrane targeting of dysferlin and MG53. PLoS One 2018; 13:e0202052. [PMID: 30092031 PMCID: PMC6084962 DOI: 10.1371/journal.pone.0202052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/26/2018] [Indexed: 11/17/2022] Open
Abstract
Dysferlin (Dysf) and mitsugumin53 (MG53) are two key proteins involved in membrane repair of muscle cells which are efficiently recruited to the sarcolemma upon lesioning. Plasma membrane localization and recruitment of a Dysf fragment to membrane lesions in zebrafish myofibers relies on the presence of a short, polybasic amino acid motif, WRRFK. Here we show that the positive charges carried by this motif are responsible for this function. In mouse MG53, we have identified a similar motif with multiple basic residues, WKKMFR. A single amino acid replacement, K279A, leads to severe aggregation of MG53 in inclusion bodies in HeLa cells. This result is due to the loss of positive charge, as shown by studying the effects of other neutral amino acids at position 279. Consequently, our data suggest that positively charged amino acid stretches play an essential role in the localization and function of Dysf and MG53.
Collapse
Affiliation(s)
- Lu Zhou
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Volker Middel
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Markus Reischl
- Institute for Applied Computer Science, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - G Ulrich Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
170
|
Croissant C, Bouvet F, Tan S, Bouter A. Imaging Membrane Repair in Single Cells Using Correlative Light and Electron Microscopy. ACTA ACUST UNITED AC 2018; 81:e55. [PMID: 30085404 DOI: 10.1002/cpcb.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many cells possess the ability to repair plasma membrane disruption in physiological conditions. Growing evidence indicates a correlation between membrane repair and many human diseases. For example, a negative correlation is observed in muscle where failure to reseal sarcolemma may contribute to the development of muscular dystrophies. Instead, a positive correlation is observed in cancer cells where membrane repair may be exacerbated during metastasis. Here we describe a protocol that combines laser technology for membrane damage, immunostaining with gold nanoparticles and imaging by fluorescence microscopy and transmission electron microscopy (TEM), which allows the characterization of the molecular machinery involved in membrane repair. Fluorescence microscopy enables to determine the subcellular localization of candidate proteins in damaged cells while TEM offers high-resolution ultrastructural analysis of the µm²-disruption site, which enables to decipher the membrane repair mechanism. Here we focus on the study of human skeletal muscle cells, for obvious clinical interest, but this protocol is also suitable for other cell types. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Coralie Croissant
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, Pessac, France
| | - Flora Bouvet
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, Pessac, France
| | - Sisareuth Tan
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, Pessac, France
| | - Anthony Bouter
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, Pessac, France
| |
Collapse
|
171
|
Urao N, Mirza RE, Corbiere TF, Hollander Z, Borchers CH, Koh TJ. Thrombospondin-1 and disease progression in dysferlinopathy. Hum Mol Genet 2018; 26:4951-4960. [PMID: 29206970 DOI: 10.1093/hmg/ddx378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/05/2017] [Indexed: 01/30/2023] Open
Abstract
The purpose of this study was to determine whether thrombospondin (TSP)-1 promotes macrophage activity and disease progression in dysferlinopathy. First, we found that levels of TSP-1 are elevated in blood of non-ambulant dysferlinopathy patients compared with ambulant patients and healthy controls, supporting the idea that TSP-1 levels are correlated with disease progression. We then crossed dysferlinopathic BlaJ mice with TSP-1 knockout mice and assessed disease progression longitudinally with magnetic resonance imaging (MRI). In these mice, deletion of TSP-1 ameliorated loss in volume and mass of the moderately affected gluteal muscle but not of the severely affected psoas muscle. T2 MRI parameters revealed that loss of TSP-1 modestly inhibited inflammation only in gluteal muscle of male mice. Histological assessment indicated that deletion of TSP-1 reduced inflammatory cell infiltration of muscle fibers, but only early in disease progression. In addition, flow cytometry analysis revealed that, in males, TSP-1 knockout reduced macrophage infiltration and phagocytic activity, which is consistent with TSP-1-enhanced phagocytosis and pro-inflammatory cytokine induction in cultured macrophages. In summary, TSP-1 appears to play an accessory role in modulating Mp activity in BlaJ mice in a gender, age and muscle-dependent manner, but is unlikely a primary driver of disease progression of dysferlinopathy.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rita E Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Thomas F Corbiere
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zsuzsanna Hollander
- PROOF Center of Excellence, Vancouver, BC, Canada.,UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Christoph H Borchers
- University of Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Victoria, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.,Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
172
|
Potter RA, Griffin DA, Sondergaard PC, Johnson RW, Pozsgai ER, Heller KN, Peterson EL, Lehtimäki KK, Windish HP, Mittal PJ, Albrecht DE, Mendell JR, Rodino-Klapac LR. Systemic Delivery of Dysferlin Overlap Vectors Provides Long-Term Gene Expression and Functional Improvement for Dysferlinopathy. Hum Gene Ther 2018; 29:749-762. [PMID: 28707952 PMCID: PMC6066196 DOI: 10.1089/hum.2017.062] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
Dysferlinopathies comprise a family of disorders caused by mutations in the dysferlin (DYSF) gene, leading to a progressive dystrophy characterized by chronic muscle fiber loss, fat replacement, and fibrosis. To correct the underlying histopathology and function, expression of full-length DYSF is required. Dual adeno-associated virus vectors have been developed, defined by a region of homology, to serve as a substrate for reconstitution of the full 6.5 kb dysferlin cDNA. Previous work studied the efficacy of this treatment through intramuscular and regional delivery routes. To maximize clinical efficacy, dysferlin-deficient mice were treated systemically to target all muscles through the vasculature for efficacy and safety studies. Mice were evaluated at multiple time points between 4 and 13 months post treatment for dysferlin expression and functional improvement using magnetic resonance imaging and magnetic resonance spectroscopy and membrane repair. A systemic dose of 6 × 1012 vector genomes resulted in widespread gene expression in the muscles. Treated muscles showed a significant decrease in central nucleation, collagen deposition, and improvement of membrane repair to wild-type levels. Treated gluteus muscles were significantly improved compared to placebo-treated muscles and were equivalent to wild type in volume, intra- and extramyocellular lipid accumulation, and fat percentage using magnetic resonance imaging and magnetic resonance spectroscopy. Dual-vector treatment allows for production of full-length functional dysferlin with no toxicity. This confirms previous safety data and validates translation of systemic gene delivery for dysferlinopathy patients.
Collapse
Affiliation(s)
- Rachael A. Potter
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Danielle A. Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Patricia C. Sondergaard
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ryan W. Johnson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Eric R. Pozsgai
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| | - Kristin N. Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ellyn L. Peterson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | | | | | | | | | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| | - Louise R. Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
- Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| |
Collapse
|
173
|
Zhang Y, Long C, Bassel-Duby R, Olson EN. Myoediting: Toward Prevention of Muscular Dystrophy by Therapeutic Genome Editing. Physiol Rev 2018; 98:1205-1240. [PMID: 29717930 PMCID: PMC6335101 DOI: 10.1152/physrev.00046.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022] Open
Abstract
Muscular dystrophies represent a large group of genetic disorders that significantly impair quality of life and often progress to premature death. There is no effective treatment for these debilitating diseases. Most therapies, developed to date, focus on alleviating the symptoms or targeting the secondary effects, while the underlying gene mutation is still present in the human genome. The discovery and application of programmable nucleases for site-specific DNA double-stranded breaks provides a powerful tool for precise genome engineering. In particular, the CRISPR/Cas system has revolutionized the genome editing field and is providing a new path for disease treatment by targeting the disease-causing genetic mutations. In this review, we provide a historical overview of genome-editing technologies, summarize the most recent advances, and discuss potential strategies and challenges for permanently correcting genetic mutations that cause muscular dystrophies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Molecular Biology, Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Chengzu Long
- Department of Molecular Biology, Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Eric N Olson
- Department of Molecular Biology, Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| |
Collapse
|
174
|
Jalali-Sefid-Dashti M, Nel M, Heckmann JM, Gamieldien J. Exome sequencing identifies novel dysferlin mutation in a family with pauci-symptomatic heterozygous carriers. BMC MEDICAL GENETICS 2018; 19:95. [PMID: 29879922 PMCID: PMC5992709 DOI: 10.1186/s12881-018-0613-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND We investigated a South African family of admixed ancestry in which the first generation (G1) developed insidious progressive distal to proximal weakness in their twenties, while their offspring (G2) experienced severe unexpected symptoms of myalgia and cramps since adolescence. Our aim was to identify deleterious mutations that segregate with the affected individuals in this family. METHODS Exome sequencing was performed on five cases, which included three affected G1 siblings and two pauci-symptomatic G2 offspring. As controls we included an unaffected G1 sibling and a spouse of one of the G1 affected individuals. Homozygous or potentially compound heterozygous variants that were predicted to be functional and segregated with the affected G1 siblings, were further evaluated. Additionally, we considered variants in all genes segregating exclusively with the affected (G1) and pauci-symptomatic (G2) individuals to address the possibility of a pseudo-autosomal dominant inheritance pattern in this family. RESULTS All affected G1 individuals were homozygous for a novel truncating p.Tyr1433Ter DYSF (dysferlin) mutation, with their asymptomatic sibling and both pauci-symptomatic G2 offspring carrying only a single mutant allele. Sanger sequencing confirmed segregation of the variant. No additional potentially contributing variant was found in the DYSF or any other relevant gene in the pauci-symptomatic carriers. CONCLUSION Our finding of a truncating dysferlin mutation confirmed dysferlinopathy in this family and we propose that the single mutant allele is the primary contributor to the neuromuscular symptoms seen in the second-generation pauci-symptomatic carriers.
Collapse
Affiliation(s)
- Mahjoubeh Jalali-Sefid-Dashti
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, 7535, South Africa
| | - Melissa Nel
- Division of Neurology, Department of Medicine, University of Cape Town, Observatory, 7925, South Africa
| | - Jeannine M Heckmann
- E8-74, Neurology, New Groote Schuur Hospital Observatory, Cape Town, 7925, South Africa
| | - Junaid Gamieldien
- South African National Bioinformatics Institute, University of the Western Cape, Private Bag X17, Bellville, 7535, South Africa.
| |
Collapse
|
175
|
Alexander JJ, Quigg RJ. Muscle, myeloid cells, and complement: a complex interaction. Cell Mol Immunol 2018; 15:992-993. [PMID: 29872116 DOI: 10.1038/s41423-018-0049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 05/13/2018] [Indexed: 11/09/2022] Open
Affiliation(s)
| | - Richard J Quigg
- Department of Medicine, SUNY at Buffalo, Buffalo, NY, 14086, USA
| |
Collapse
|
176
|
Barthélémy F, Defour A, Lévy N, Krahn M, Bartoli M. Muscle Cells Fix Breaches by Orchestrating a Membrane Repair Ballet. J Neuromuscul Dis 2018; 5:21-28. [PMID: 29480214 PMCID: PMC5836414 DOI: 10.3233/jnd-170251] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Skeletal muscle undergoes many micro-membrane lesions at physiological state. Based on their sizes and magnitude these lesions are repaired via different complexes on a specific spatio-temporal manner. One of the major repair complex is a dysferlin-dependent mechanism. Accordingly, mutations in the DYSF gene encoding dysferlin results in the development of several muscle pathologies called dysferlinopathies, where abnormalities of the membrane repair process have been characterized in patients and animal models. Recent efforts have been deployed to decipher the function of dysferlin, they shed light on its direct implication in sarcolemma resealing after injuries. These discoveries served as a strong ground to design therapeutic approaches for dysferlin-deficient patients. This review detailed the different partners and function of dysferlin and positions the sarcolemma repair in normal and pathological conditions.
Collapse
Affiliation(s)
- Florian Barthélémy
- Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA.,Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
| | - Aurélia Defour
- Aix Marseille University, MMG, INSERM, Marseille, France
| | - Nicolas Lévy
- Aix Marseille University, MMG, INSERM, Marseille, France
| | - Martin Krahn
- Aix Marseille University, MMG, INSERM, Marseille, France
| | - Marc Bartoli
- Aix Marseille University, MMG, INSERM, Marseille, France
| |
Collapse
|
177
|
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: 0.7] [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
|
178
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
179
|
Collier AF, Gumerson J, Lehtimäki K, Puoliväli J, Jones JW, Kane MA, Manne S, O'Neill A, Windish HP, Ahtoniemi T, Williams BA, Albrecht DE, Bloch RJ. Effect of Ibuprofen on Skeletal Muscle of Dysferlin-Null Mice. J Pharmacol Exp Ther 2018; 364:409-419. [PMID: 29284661 PMCID: PMC5801553 DOI: 10.1124/jpet.117.244244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/21/2017] [Indexed: 12/19/2022] Open
Abstract
Ibuprofen, a nonsteroidal anti-inflammatory drug, and nitric oxide (NO) donors have been reported to reduce the severity of muscular dystrophies in mice associated with the absence of dystrophin or α-sarcoglycan, but their effects on mice that are dystrophic due to the absence of dysferlin have not been examined. We have tested ibuprofen, as well as isosorbide dinitrate (ISDN), a NO donor, to learn whether used alone or together they protect dysferlin-null muscle in A/J mice from large strain injury (LSI) induced by a series of high strain lengthening contractions. Mice were maintained on chow containing ibuprofen and ISDN for 4 weeks. They were then subjected to LSI and maintained on the drugs for 3 additional days. We measured loss of torque immediately following injury and at day 3 postinjury, fiber necrosis, and macrophage infiltration at day 3 postinjury, and serum levels of the drugs at the time of euthanasia. Loss of torque immediately after injury was not altered by the drugs. However, the torque on day 3 postinjury significantly decreased as a function of ibuprofen concentration in the serum (range, 0.67-8.2 µg/ml), independent of ISDN. The effects of ISDN on torque loss at day 3 postinjury were not significant. In long-term studies of dysferlinopathic BlAJ mice, lower doses of ibuprofen had no effects on muscle morphology, but reduced treadmill running by 40%. Our results indicate that ibuprofen can have deleterious effects on dysferlin-null muscle and suggest that its use at pharmacological doses should be avoided by individuals with dysferlinopathies.
Collapse
Affiliation(s)
- Alyssa F Collier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jessica Gumerson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Kimmo Lehtimäki
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jukka Puoliväli
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jace W Jones
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Maureen A Kane
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Sankeerth Manne
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Andrea O'Neill
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Hillarie P Windish
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Toni Ahtoniemi
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Bradley A Williams
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Douglas E Albrecht
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| |
Collapse
|
180
|
Dubey R, Malhotra SS, Gupta SK. Forskolin-mediated BeWo cell fusion involves down-regulation of miR-92a-1-5p that targets dysferlin and protein kinase cAMP-activated catalytic subunit alpha. Am J Reprod Immunol 2018; 79:e12834. [PMID: 29484758 DOI: 10.1111/aji.12834] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/04/2018] [Indexed: 12/16/2022] Open
Abstract
PROBLEM To study the role of miRNA(s) during trophoblastic BeWo cell fusion. METHOD OF STUDY Changes in miRNA(s) profile of BeWo cells treated with forskolin were analyzed using Affymetrix miRNA microarray platform. Down-regulated miRNA, miR-92a-1-5p, was overexpressed in BeWo cells followed by forskolin treatment to understand its relevance in the process of BeWo cell fusion by desmoplakin I+II staining and hCG secretion by ELISA. Predicted targets of miR-92a-1-5p were also confirmed by qRT-PCR/Western blotting. RESULTS The miRNA profiling of BeWo cells after forskolin (25 μmol/L) treatment identified miR-92a-1-5p as the most significantly down-regulated miRNA both at 24 and 48 hours time points. Overexpression of miR-92a-1-5p in these cells led to a significant decrease in forskolin-mediated cell fusion and hCG secretion. miRNA target prediction software, TargetScan, revealed dysferlin (DYSF) and protein kinase cAMP-activated catalytic subunit alpha (PRKACA), as target genes of miR-92a-1-5p. Overexpression of miR-92a-1-5p in BeWo cells showed reduction in forskolin-induced transcripts for DYSF and PRKACA. Further, reduction in DYSF (~2.6-fold) at protein level and PRKACA-encoded protein kinase A catalytic subunit alpha (PKAC-α; ~1.6-fold) were also observed. CONCLUSION These observations suggest that miR-92a-1-5p regulates forskolin-mediated BeWo cell fusion and hCG secretion by regulating PKA signaling pathway and dysferlin expression.
Collapse
Affiliation(s)
- Richa Dubey
- Reproductive Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Sudha S Malhotra
- Reproductive Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Satish K Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
181
|
Dial AG, Rooprai P, Lally JS, Bujak AL, Steinberg GR, Ljubicic V. The role of AMP‐activated protein kinase in the expression of the dystrophin‐associated protein complex in skeletal muscle. FASEB J 2018; 32:2950-2965. [DOI: 10.1096/fj.201700868rrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Athan G. Dial
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| | - Paul Rooprai
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| | - James S. Lally
- Department of MedicineMcMaster University Hamilton Ontario Canada
| | - Adam L. Bujak
- Department of MedicineMcMaster University Hamilton Ontario Canada
| | - Gregory R. Steinberg
- Department of MedicineMcMaster University Hamilton Ontario Canada
- Department of Biochemistry and Biomedical SciencesMcMaster University Hamilton Ontario Canada
| | - Vladimir Ljubicic
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| |
Collapse
|
182
|
Hu YY, Lian YJ, Xu HL, Zheng YK, Li CF, Zhang JW, Yan SP. Novel, de novo dysferlin gene mutations in a patient with Miyoshi myopathy. Neurosci Lett 2018; 664:107-109. [DOI: 10.1016/j.neulet.2017.10.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 11/29/2022]
|
183
|
An Overview of Recent Advances and Clinical Applications of Exon Skipping and Splice Modulation for Muscular Dystrophy and Various Genetic Diseases. Methods Mol Biol 2018; 1828:31-55. [PMID: 30171533 DOI: 10.1007/978-1-4939-8651-4_2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Exon skipping is a therapeutic approach that is feasible for various genetic diseases and has been studied and developed for over two decades. This approach uses antisense oligonucleotides (AON) to modify the splicing of pre-mRNA to correct the mutation responsible for a disease, or to suppress a particular gene expression, as in allergic diseases. Antisense-mediated exon skipping is most extensively studied in Duchenne muscular dystrophy (DMD) and has developed from in vitro proof-of-concept studies to clinical trials targeting various single exons such as exon 45 (casimersen), exon 53 (NS-065/NCNP-01, golodirsen), and exon 51 (eteplirsen). Eteplirsen (brand name Exondys 51), is the first approved antisense therapy for DMD in the USA, and provides a treatment option for ~14% of all DMD patients, who are amenable to exon 51 skipping. Eteplirsen is granted accelerated approval and marketing authorization by the US Food and Drug Administration (FDA), on the condition that additional postapproval trials show clinical benefit. Permanent exon skipping achieved at the DNA level using clustered regularly interspaced short palindromic repeats (CRISPR) technology holds promise in current preclinical trials for DMD. In hopes of achieving clinical success parallel to DMD, exon skipping and splice modulation are also being studied in other muscular dystrophies, such as Fukuyama congenital muscular dystrophy (FCMD), dysferlinopathy including limb-girdle muscular dystrophy type 2B (LGMD2B), Miyoshi myopathy (MM), and distal anterior compartment myopathy (DMAT), myotonic dystrophy, and merosin-deficient congenital muscular dystrophy type 1A (MDC1A). This chapter also summarizes the development of antisense-mediated exon skipping therapy in diseases such as Usher syndrome, dystrophic epidermolysis bullosa, fibrodysplasia ossificans progressiva (FOP), and allergic diseases.
Collapse
|
184
|
McElhanon KE, Bhattacharya S. Altered membrane integrity in the progression of muscle diseases. Life Sci 2017; 192:166-172. [PMID: 29183798 DOI: 10.1016/j.lfs.2017.11.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/12/2017] [Accepted: 11/22/2017] [Indexed: 12/27/2022]
Abstract
Sarcolemmal integrity is orchestrated through the interplay of preserving membrane strength and fast tracking the membrane repair process during an event of compromised membrane fragility. Several molecular players have been identified that act in a concerted fashion to maintain the barrier function of the muscle membrane. Substantial research findings in the field of muscle biology point out the importance of maintaining membrane integrity as a key contributory factor to cellular homeostasis. Innumerable data on the progression of membrane pathology associated with compromised muscle membrane integrity support targeting sarcolemmal integrity in skeletal and cardiac muscle as a model therapeutic strategy to alleviate some of the pathologic conditions. This review will discuss strategies that researchers have undertaken to compensate for an imbalance in sarcolemma membrane fragility and membrane repair to maintain muscle membrane integrity.
Collapse
Affiliation(s)
- Kevin E McElhanon
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 W. 12th Ave, Columbus, OH 43210-1252, United States
| | - Sayak Bhattacharya
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 W. 12th Ave, Columbus, OH 43210-1252, United States.
| |
Collapse
|
185
|
Ma J, Pichavant C, du Bois H, Bhakta M, Calos MP. DNA-Mediated Gene Therapy in a Mouse Model of Limb Girdle Muscular Dystrophy 2B. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:123-131. [PMID: 29159199 PMCID: PMC5684445 DOI: 10.1016/j.omtm.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 11/26/2022]
Abstract
Mutations in the gene for dysferlin cause a degenerative disorder of skeletal muscle known as limb girdle muscular dystrophy 2B. To achieve gene delivery of plasmids encoding dysferlin to hind limb muscles of dysferlin knockout mice, we used a vascular injection method that perfused naked plasmid DNA into all major muscle groups of the hind limb. We monitored delivery by luciferase live imaging and western blot, confirming strong dysferlin expression that persisted over the 3-month time course of the experiment. Co-delivery of the follistatin gene, which may promote muscle growth, was monitored by ELISA. Immunohistochemistry documented the presence of dysferlin in muscle fibers in treated limbs, and PCR confirmed the presence of plasmid DNA. Because dysferlin is involved in repair of the sarcolemmal membrane, dysferlin loss leads to fragile sarcolemmal membranes that can be detected by permeability to Evan’s blue dye. We showed that after gene therapy with a plasmid encoding both dysferlin and follistatin, statistically significant reduction in Evan’s blue dye permeability was present in hamstring muscles. These results suggest that vascular delivery of plasmids carrying these therapeutic genes may lead to simple and effective approaches for improving the clinical condition of limb girdle muscular dystrophy 2B.
Collapse
Affiliation(s)
- Julia Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Christophe Pichavant
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Haley du Bois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Mital Bhakta
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Michele P Calos
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| |
Collapse
|
186
|
Woolger N, Bournazos A, Sophocleous RA, Evesson FJ, Lek A, Driemer B, Sutton RB, Cooper ST. Limited proteolysis as a tool to probe the tertiary conformation of dysferlin and structural consequences of patient missense variant L344P. J Biol Chem 2017; 292:18577-18591. [PMID: 28904177 DOI: 10.1074/jbc.m117.790014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/13/2017] [Indexed: 12/19/2022] Open
Abstract
Dysferlin is a large transmembrane protein that plays a key role in cell membrane repair and underlies a recessive form of inherited muscular dystrophy. Dysferlinopathy is characterized by absence or marked reduction of dysferlin protein with 43% of reported pathogenic variants being missense variants that span the length of the dysferlin protein. The unique structure of dysferlin, with seven tandem C2 domains separated by linkers, suggests dysferlin may dynamically associate with phospholipid membranes in response to Ca2+ signaling. However, the overall conformation of the dysferlin protein is uncharacterized. To dissect the structural architecture of dysferlin, we have applied the method of limited proteolysis, which allows nonspecific digestion of unfolded peptides by trypsin. Using five antibodies spanning the dysferlin protein, we identified a highly reproducible jigsaw map of dysferlin fragments protected from digestion. Our data infer a modular architecture of four tertiary domains: 1) C2A, which is readily removed as a solo domain; 2) midregion C2B-C2C-Fer-DysF, commonly excised as an intact module, with subdigestion to different fragments suggesting several dynamic folding options; 3) C-terminal four-C2 domain module; and 4) calpain-cleaved mini-dysferlinC72, which is particularly resistant to proteolysis. Importantly, we reveal a patient missense variant, L344P, that largely escapes proteasomal surveillance and shows subtle but clear changes in tertiary conformation. Accompanying evidence from immunohistochemistry and flow cytometry using antibodies with conformationally sensitive epitopes supports proteolysis data. Collectively, we provide insight into the structural topology of dysferlin and show how a single missense mutation within dysferlin can exert local changes in tertiary conformation.
Collapse
Affiliation(s)
- Natalie Woolger
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - Adam Bournazos
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - Reece A Sophocleous
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - Frances J Evesson
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - Angela Lek
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - Birgit Driemer
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia.,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| | - R Bryan Sutton
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Sandra T Cooper
- From the Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Locked Bag 4001, Westmead 2145, Australia, .,Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney 2006, Australia, and
| |
Collapse
|
187
|
Angelini C, Fanin M. Limb girdle muscular dystrophies: clinical-genetical diagnostic update and prospects for therapy. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1367283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Corrado Angelini
- Department of Neurodegenerative Disorders, Neuromuscular Center, San Camillo Hospital IRCCS, Venice, Italy
| | - Marina Fanin
- Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
188
|
Quattrocelli M, Salamone IM, Page PG, Warner JL, Demonbreun AR, McNally EM. Intermittent Glucocorticoid Dosing Improves Muscle Repair and Function in Mice with Limb-Girdle Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2520-2535. [PMID: 28823869 DOI: 10.1016/j.ajpath.2017.07.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Isabella M Salamone
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Patrick G Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
189
|
A muscle-specific protein 'myoferlin' modulates IL-6/STAT3 signaling by chaperoning activated STAT3 to nucleus. Oncogene 2017; 36:6374-6382. [PMID: 28745314 PMCID: PMC5690845 DOI: 10.1038/onc.2017.245] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/08/2017] [Accepted: 06/12/2017] [Indexed: 01/03/2023]
Abstract
Myoferlin, a member of ferlin family of proteins, was first discovered as a candidate gene for muscular dystrophy and cardiomyopathy. Recently, myoferlin was shown to be also expressed in endothelial and cancer cells where it was shown to modulate vascular endothelial growth factor (VEGFR)-2 and epidermal growth factor receptor (EGFR) signaling by enhancing their stability and recycling. Based on these reports, we hypothesized that myoferlin might be regulating IL-6 signaling by modulating IL-6R stabilization and recycling. However, in our immunoprecipitation (IP) experiments, we did not observe myoferlin binding with IL-6R. Instead, we made a novel discovery that in resting cells myoferlin was bound to EHD2 protein and when cells were treated with IL-6, myoferlin dissociated from EHD2 and binds to activated STAT3. Interestingly, myoferlin depletion did not affect STAT3 phosphorylation, but completely blocked STAT3 translocation to nucleus. In addition, inhibition of STAT3 phosphorylation by phosphorylation-defective STAT3 mutants or JAK inhibitor blocked STAT3 binding to myoferlin and nuclear translocation. Myoferlin knockdown significantly decreased IL-6-mediated tumor cell migration, tumorsphere formation and ALDH-positive cancer stem cell population, in vitro. Furthermore, myoferlin knockdown significantly decreased IL-6-meditated tumor growth and tumor metastasis. Based on these results, we have proposed a novel model for the role of myoferlin in chaperoning phosphorylated STAT3 to the nucleus.
Collapse
|
190
|
Treatment with Recombinant Human MG53 Protein Increases Membrane Integrity in a Mouse Model of Limb Girdle Muscular Dystrophy 2B. Mol Ther 2017; 25:2360-2371. [PMID: 28750735 DOI: 10.1016/j.ymthe.2017.06.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022] Open
Abstract
Limb girdle muscular dystrophy type 2B (LGMD2B) and other dysferlinopathies are degenerative muscle diseases that result from mutations in the dysferlin gene and have limited treatment options. The dysferlin protein has been linked to multiple cellular functions including a Ca2+-dependent membrane repair process that reseals disruptions in the sarcolemmal membrane. Recombinant human MG53 protein (rhMG53) can increase the membrane repair process in multiple cell types both in vitro and in vivo. Here, we tested whether rhMG53 protein can improve membrane repair in a dysferlin-deficient mouse model of LGMD2B (B6.129-Dysftm1Kcam/J). We found that rhMG53 can increase the integrity of the sarcolemmal membrane of isolated muscle fibers and whole muscles in a Ca2+-independent fashion when assayed by a multi-photon laser wounding assay. Intraperitoneal injection of rhMG53 into mice before acute eccentric treadmill exercise can decrease the release of intracellular enzymes from skeletal muscle and decrease the entry of immunoglobulin G and Evans blue dye into muscle fibers in vivo. These results indicate that short-term rhMG53 treatment can ameliorate one of the underlying defects in dysferlin-deficient muscle by increasing sarcolemmal membrane integrity. We also provide evidence that rhMG53 protein increases membrane integrity independently of the canonical dysferlin-mediated, Ca2+-dependent pathway known to be important for sarcolemmal membrane repair.
Collapse
|
191
|
Dastur RS, Gaitonde PS, Kachwala M, Nallamilli BRR, Ankala A, Khadilkar SV, Atchayaram N, Gayathri N, Meena AK, Rufibach L, Shira S, Hegde M. Detection of Dysferlin Gene Pathogenic Variants in the Indian Population in Patients Predicted to have a Dysferlinopathy Using a Blood-based Monocyte Assay and Clinical Algorithm: A Model for Accurate and Cost-effective Diagnosis. Ann Indian Acad Neurol 2017; 20:302-308. [PMID: 28904466 PMCID: PMC5586129 DOI: 10.4103/aian.aian_129_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophy (LGMD) is the most common adult-onset class of muscular dystrophies in India, but a majority of suspected LGMDs in India remain unclassified to the genetic subtype level. The next-generation sequencing (NGS)-based approaches have allowed molecular characterization and subtype diagnosis in a majority of these patients in India. MATERIALS AND METHODS (I) To select probable dysferlinopathy (LGMD2B) cases from other LGMD subtypes using two screening methods (i) to determine the status of dysferlin protein expression in blood (peripheral blood mononuclear cell) by monocyte assay (ii) using a predictive algorithm called automated LGMD diagnostic assistant (ALDA) to obtain possible LGMD subtypes based on clinical symptoms. (II) Identification of gene pathogenic variants by NGS for 34 genes associated with LGMD or LGMD like muscular dystrophies, in cases showing: absence of dysferlin protein by the monocyte assay and/or a typical dysferlinopathy phenotype, with medium to high predictive scores using the ALDA tool. RESULTS Out of the 125 patients screened by NGS, 96 were confirmed with two dysferlin variants, of which 84 were homozygous. Single dysferlin pathogenic variants were seen in 4 patients, whereas 25 showed no variants in the dysferlin gene. CONCLUSION In this study, 98.2% of patients with absence of the dysferlin protein showed one or more variants in the dysferlin gene and hence has a high predictive significance in diagnosing dysferlinopathies. However, collection of blood samples from all over India for protein analysis is expensive. Our analysis shows that the use of the "ALDA tool" could be a cost-effective alternative method. Identification of dysferlin pathogenic variants by NGS is the ultimate method for diagnosing dysferlinopathies though follow-up with the monocyte assay can be useful to understand the phenotype in relation to the dysferlin protein expression and also be a useful biomarker for future clinical trials.
Collapse
Affiliation(s)
- Rashna Sam Dastur
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders, Atlanta, Georgia, USA
| | | | - Munira Kachwala
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders, Atlanta, Georgia, USA
| | - Babi R. R. Nallamilli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arunkanth Ankala
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Satish V. Khadilkar
- Department of Neurology, Sir J J Group of Hospitals, Grant Medical College, Mumbai, Maharashtra, India
| | | | - N. Gayathri
- Department of Neurology, NIMHANS, Bengaluru, Karnataka
| | - A. K. Meena
- Department of Neurology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | | | | | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
192
|
Lukyanenko V, Muriel JM, Bloch RJ. Coupling of excitation to Ca 2+ release is modulated by dysferlin. J Physiol 2017; 595:5191-5207. [PMID: 28568606 DOI: 10.1113/jp274515] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Dysferlin, the protein missing in limb girdle muscular dystrophy 2B and Miyoshi myopathy, concentrates in transverse tubules of skeletal muscle, where it stabilizes voltage-induced Ca2+ transients against loss after osmotic shock injury (OSI). Local expression of dysferlin in dysferlin-null myofibres increases transient amplitude to control levels and protects them from loss after OSI. Inhibitors of ryanodine receptors (RyR1) and L-type Ca2+ channels protect voltage-induced Ca2+ transients from loss; thus both proteins play a role in injury in dysferlin's absence. Effects of Ca2+ -free medium and S107, which inhibits SR Ca2+ leak, suggest the SR as the primary source of Ca2+ responsible for the loss of the Ca2+ transient upon injury. Ca2+ waves were induced by OSI and suppressed by exogenous dysferlin. We conclude that dysferlin prevents injury-induced SR Ca2+ leak. ABSTRACT Dysferlin concentrates in the transverse tubules of skeletal muscle and stabilizes Ca2+ transients when muscle fibres are subjected to osmotic shock injury (OSI). We show here that voltage-induced Ca2+ transients elicited in dysferlin-null A/J myofibres were smaller than control A/WySnJ fibres. Regional expression of Venus-dysferlin chimeras in A/J fibres restored the full amplitude of the Ca2+ transients and protected against OSI. We also show that drugs that target ryanodine receptors (RyR1: dantrolene, tetracaine, S107) and L-type Ca2+ channels (LTCCs: nifedipine, verapamil, diltiazem) prevented the decrease in Ca2+ transients in A/J fibres following OSI. Diltiazem specifically increased transients by ∼20% in uninjured A/J fibres, restoring them to control values. The fact that both RyR1s and LTCCs were involved in OSI-induced damage suggests that damage is mediated by increased Ca2+ leak from the sarcoplasmic reticulum (SR) through the RyR1. Congruent with this, injured A/J fibres produced Ca2+ sparks and Ca2+ waves. S107 (a stabilizer of RyR1-FK506 binding protein coupling that reduces Ca2+ leak) or local expression of Venus-dysferlin prevented OSI-induced Ca2+ waves. Our data suggest that dysferlin modulates SR Ca2+ release in skeletal muscle, and that in its absence OSI causes increased RyR1-mediated Ca2+ leak from the SR into the cytoplasm.
Collapse
Affiliation(s)
- Valeriy Lukyanenko
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joaquin M Muriel
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
193
|
Limb-Girdle Muscular Dystrophy 2B and Miyoshi Presentations of Dysferlinopathy. Am J Med Sci 2017; 353:484-491. [DOI: 10.1016/j.amjms.2016.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 11/20/2022]
|
194
|
Piper AK, Ross SE, Redpath GM, Lemckert FA, Woolger N, Bournazos A, Greer PA, Sutton RB, Cooper ST. Enzymatic cleavage of myoferlin releases a dual C2-domain module linked to ERK signalling. Cell Signal 2017; 33:30-40. [PMID: 28192161 PMCID: PMC5995151 DOI: 10.1016/j.cellsig.2017.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/07/2017] [Indexed: 01/17/2023]
Abstract
Myoferlin and dysferlin are closely related members of the ferlin family of Ca2+-regulated vesicle fusion proteins. Dysferlin is proposed to play a role in Ca2+-triggered vesicle fusion during membrane repair. Myoferlin regulates endocytosis, recycling of growth factor receptors and adhesion proteins, and is linked to the metastatic potential of cancer cells. Our previous studies establish that dysferlin is cleaved by calpains during membrane injury, with the cleavage motif encoded by alternately-spliced exon 40a. Herein we describe the cleavage of myoferlin, yielding a membrane-associated dual C2 domain 'mini-myoferlin'. Myoferlin bears two enzymatic cleavage sites: a canonical cleavage site encoded by exon 38 within the C2DE domain; and a second cleavage site in the linker adjacent to C2DE, encoded by alternately-spliced exon 38a, homologous to dysferlin exon 40a. Both myoferlin cleavage sites, when introduced into dysferlin, can functionally substitute for exon 40a to confer Ca2+-triggered calpain cleavage in response to membrane injury. However, enzymatic cleavage of myoferlin is complex, showing both constitutive or Ca2+-enhanced cleavage in different cell lines, that is not solely dependent on calpains-1 or -2. The functional impact of myoferlin cleavage was explored through signalling protein phospho-protein arrays revealing specific activation of ERK1/2 by ectopic expression of cleavable myoferlin, but not an uncleavable isoform. In summary, we molecularly define two enzymatic cleavage sites within myoferlin and demonstrate 'mini-myoferlin' can be detected in human breast cancer tumour samples and cell lines. These data further illustrate that enzymatic cleavage of ferlins is an evolutionarily preserved mechanism to release functionally specialized mini-modules.
Collapse
Affiliation(s)
- Ann-Katrin Piper
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Samuel E Ross
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Gregory M Redpath
- EMBL Australia Node in Single Molecule Science, School of Medical Science, University of New South Wales, Sydney, NSW, Australia
| | - Frances A Lemckert
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Natalie Woolger
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Adam Bournazos
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Peter A Greer
- Department of Pathology and Molecular Medicine, Queen's University, Division of Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada
| | - Roger B Sutton
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sandra T Cooper
- Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Discipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, Australia.
| |
Collapse
|
195
|
Demonbreun AR, McNally EM. Muscle cell communication in development and repair. Curr Opin Pharmacol 2017; 34:7-14. [PMID: 28419894 DOI: 10.1016/j.coph.2017.03.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/25/2017] [Accepted: 03/06/2017] [Indexed: 12/18/2022]
Abstract
Under basal conditions, postnatal skeletal muscle displays little cell turnover. With injury, muscle initiates a rapid repair response to reseal damaged membrane, reactivating many developmental pathways to facilitate muscle regeneration and prevent tissue loss. Muscle precursor cells become activated accompanied by differentiation and fusion during both muscle growth and regeneration; inter-cellular communication is required for successful completion of these processes. Cellular communication is mediated by lipids, fusogenic membrane proteins, and exosomes. Muscle-derived exosomes carry proteins and micro RNAs as cargo. Secreted factors such as IGF-1, TGFβ, and myostatin are also released by muscle cells providing local signaling cues to modulate muscle fusion and regeneration. Proteins that regulate myoblast fusion also participate in membrane repair and regeneration. Here we will review methods of muscle cell communication focusing on proteins that mediate membrane fusion, exosomes, and autocrine factors.
Collapse
Affiliation(s)
- Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
196
|
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: 17] [Impact Index Per Article: 2.1] [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
|
197
|
Savisaar R, Hurst LD. Estimating the prevalence of functional exonic splice regulatory information. Hum Genet 2017; 136:1059-1078. [PMID: 28405812 PMCID: PMC5602102 DOI: 10.1007/s00439-017-1798-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
In addition to coding information, human exons contain sequences necessary for correct splicing. These elements are known to be under purifying selection and their disruption can cause disease. However, the density of functional exonic splicing information remains profoundly uncertain. Several groups have experimentally investigated how mutations at different exonic positions affect splicing. They have found splice information to be distributed widely in exons, with one estimate putting the proportion of splicing-relevant nucleotides at >90%. These results suggest that splicing could place a major pressure on exon evolution. However, analyses of sequence conservation have concluded that the need to preserve splice regulatory signals only slightly constrains exon evolution, with a resulting decrease in the average human rate of synonymous evolution of only 1–4%. Why do these two lines of research come to such different conclusions? Among other reasons, we suggest that the methods are measuring different things: one assays the density of sites that affect splicing, the other the density of sites whose effects on splicing are visible to selection. In addition, the experimental methods typically consider short exons, thereby enriching for nucleotides close to the splice junction, such sites being enriched for splice-control elements. By contrast, in part owing to correction for nucleotide composition biases and to the assumption that constraint only operates on exon ends, the conservation-based methods can be overly conservative.
Collapse
Affiliation(s)
- Rosina Savisaar
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Laurence D Hurst
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| |
Collapse
|
198
|
Jin SQ, Yu M, Zhang W, Lyu H, Yuan Y, Wang ZX. Dysferlin Gene Mutation Spectrum in a Large Cohort of Chinese Patients with Dysferlinopathy. Chin Med J (Engl) 2017; 129:2287-93. [PMID: 27647186 PMCID: PMC5040013 DOI: 10.4103/0366-6999.190671] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Dysferlinopathy is caused by mutations in the dysferlin (DYSF) gene. Here, we described the genetic features of a large cohort of Chinese patients with this disease. METHODS Eighty-nine index patients were included in the study. DYSF gene analysis was performed by Sanger sequencing in 41 patients and targeted next generation sequencing (NGS) in 48 patients. Multiplex ligation-dependent probe amplification (MLPA) was performed to detect exon duplication/deletion in patients with only one pathogenic mutation. RESULTS Among the 89 index patients, 79 patients were demonstrated to carry two disease-causing (73 cases) or possibly disease-causing mutations (6 cases), including 26 patients with homozygous mutations. We identified 105 different mutations, including 59 novel ones. Notably, in 13 patients in whom only one pathogenic mutation was initially found by Sanger sequencing or NGS, 3 were further identified to carry exon deletions by MLPA. The mutations identified in this study appeared to cluster in the N-terminal region. Mutation types included missense mutations (30.06%), nonsense mutations (17.18%), frameshift mutations (30.67%), in-frame deletions (2.45%), intronic mutations (17.79%), and exonic rearrangement (1.84%). No genotype-phenotype correlation was identified. CONCLUSIONS DYSF mutations in Chinese patients clustered in the N-terminal region of the gene. Exonic rearrangements were found in 23% of patients with only one pathogenic mutation identified by Sanger sequencing or NGS. The novel mutations found in this study greatly expanded the mutational spectrum of dysferlinopathy.
Collapse
Affiliation(s)
- Su-Qin Jin
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - He Lyu
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Zhao-Xia Wang
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
199
|
Umakhanova ZR, Bardakov SN, Mavlikeev MO, Chernova ON, Magomedova RM, Akhmedova PG, Yakovlev IA, Dalgatov GD, Fedotov VP, Isaev AA, Deev RV. Twenty-Year Clinical Progression of Dysferlinopathy in Patients from Dagestan. Front Neurol 2017; 8:77. [PMID: 28337173 PMCID: PMC5340769 DOI: 10.3389/fneur.2017.00077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/20/2017] [Indexed: 11/21/2022] Open
Abstract
To date, over 30 genes with mutations causing limb-girdle muscle dystrophy have been described. Dysferlinopathies are a form of limb-girdle muscle dystrophy type 2B with an incidence ranging from 1:1,300 to 1:200,000 in different populations. In 1996, Dr. S. N. Illarioshkin described a family from the Botlikhsky district of Dagestan, where limb-girdle muscle dystrophy type 2B and Miyoshi myopathy were diagnosed in 12 members from three generations of a large Avar family. In 2000, a previously undescribed mutation in the DYSF gene (c.TG573/574AT; p. Val67Asp) was detected in the affected members of this family. Twenty years later, in this work, we re-examine five known and seven newly affected family members previously diagnosed with dysferlinopathy. We observed disease progression in family members who were previously diagnosed and noted obvious clinical polymorphism of the disease. A typical clinical case is provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ivan A Yakovlev
- Kazan Federal University, Kazan, Russia; Human Stem Cells Institute, Moscow, Russia; Ryazan State Medical University, Ryazan, Russia
| | | | | | | | - Roman V Deev
- Human Stem Cells Institute, Moscow, Russia; Ryazan State Medical University, Ryazan, Russia
| |
Collapse
|
200
|
Kumar B, Brown NV, Swanson BJ, Schmitt AC, Old M, Ozer E, Agrawal A, Schuller DE, Teknos TN, Kumar P. High expression of myoferlin is associated with poor outcome in oropharyngeal squamous cell carcinoma patients and is inversely associated with HPV-status. Oncotarget 2017; 7:18665-77. [PMID: 26919244 PMCID: PMC4951318 DOI: 10.18632/oncotarget.7625] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/11/2016] [Indexed: 11/25/2022] Open
Abstract
Myoferlin (MYOF) is a member of ferlin family of membrane proteins that was originally discovered as a muscle specific protein. Recent studies have shown that myoferlin is also expressed in other cell types including endothelial cells and cancer cells. However, very little is known about the expression and biological role of myoferlin in head and neck cancer. In this study, we examined expression profile of myoferlin in oropharyngeal squamous cell carcinoma (OPSCC) and assessed its correlation with disease progression and patient outcome. In univariate analyses, nuclear MYOF was associated with poor overall survival (p<0.001) and these patients had 5.5 times increased hazard of death (95% Cl 3.4-8.8). Nuclear myoferlin expression was also directly associated with tumor recurrence (p<0.001), perineural invasion (p=0.008), extracapsular spread (p=0.009), higher T-stage (p=0.0015) and distant metastasis (p<0.001). In addition, nuclear MYOF expression was directly associated with IL-6 (p<0.001) and inversely with HPV status (p=0.0014). In a subgroup survival analysis, MYOF nuclear+/IL-6+ group had worst survival (84.6% mortality), whereas MYOF nuclear-/IL-6- had the best survival. Similarly, patients with HPV-negative/MYOF-positive tumors had worse survival compared to HPV-positive/MYOF-negative. Taken together, our results demonstrate for the first time that nuclear myoferlin expression independently predicts poor clinical outcome in OPSCC patients.
Collapse
Affiliation(s)
- Bhavna Kumar
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole V Brown
- Center for Biostatistics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin J Swanson
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA
| | - Alessandra C Schmitt
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA.,Current affiliation: Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30303, USA
| | - Matthew Old
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Enver Ozer
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Amit Agrawal
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - David E Schuller
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Theodoros N Teknos
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Pawan Kumar
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|