51
|
Whitlock JM, Hartzell HC. Anoctamins/TMEM16 Proteins: Chloride Channels Flirting with Lipids and Extracellular Vesicles. Annu Rev Physiol 2016; 79:119-143. [PMID: 27860832 DOI: 10.1146/annurev-physiol-022516-034031] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anoctamin (ANO)/TMEM16 proteins exhibit diverse functions in cells throughout the body and are implicated in several human diseases. Although the founding members ANO1 (TMEM16A) and ANO2 (TMEM16B) are Ca2+-activated Cl- channels, most ANO paralogs are Ca2+-dependent phospholipid scramblases that serve as channels facilitating the movement (scrambling) of phospholipids between leaflets of the membrane bilayer. Phospholipid scrambling significantly alters the physical properties of the membrane and its landscape and has vast downstream signaling consequences. In particular, phosphatidylserine exposed on the external leaflet of the plasma membrane functions as a ligand for receptors vital for cell-cell communication. A major consequence of Ca2+-dependent scrambling is the release of extracellular vesicles that function as intercellular messengers by delivering signaling proteins and noncoding RNAs to alter target cell function. We discuss the physiological implications of Ca2+-dependent phospholipid scrambling, the extracellular vesicles associated with this activity, and the roles of ANOs in these processes.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| |
Collapse
|
52
|
Membrane repair of human skeletal muscle cells requires Annexin-A5. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2267-79. [PMID: 27286750 DOI: 10.1016/j.bbamcr.2016.06.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 11/20/2022]
Abstract
Defect in membrane repair contributes to the development of limb girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. In healthy skeletal muscle, unraveling membrane repair mechanisms requires to establish an exhaustive list of the components of the resealing machinery. Here we show that human myotubes rendered deficient for Annexin-A5 (AnxA5) suffer from a severe defect in membrane resealing. This defect is rescued by the addition of recombinant AnxA5 while an AnxA5 mutant, which is unable to form 2D protein arrays, has no effect. Using correlative light and electron microscopy, we show that AnxA5 binds to the edges of the torn membrane, as early as a few seconds after sarcolemma injury, where it probably self-assembles into 2D arrays. In addition, we observed that membrane resealing is associated with the presence of a cluster of lipid vesicles at the wounded site. AnxA5 is present at the surface of these vesicles and may thus participate in plugging the cell membrane disruption. Finally, we show that AnxA5 behaves similarly in myotubes from a muscle cell line established from a patient suffering from LGMD2B, a myopathy due to dysferlin mutations, which indicates that trafficking of AnxA5 during sarcolemma damage is independent of the presence of dysferlin.
Collapse
|
53
|
Sánchez-Chapul L, Ángel-Muñoz MD, Ruano-Calderón L, Luna-Angulo A, Coral-Vázquez R, Hernández-Hernández Ó, Magaña JJ, León-Hernández SR, Escobar-Cedillo RE, Vargas S. Dysferlin quantification in monocytes for rapid screening for dysferlinopathies. Muscle Nerve 2016; 54:1064-1071. [PMID: 27104310 DOI: 10.1002/mus.25156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
INTRODUCTION In this study, we determined normal levels of dysferlin expression in CD14+ monocytes by flow cytometry (FC) as a screening tool for dysferlinopathies. METHODS Monocytes from 183 healthy individuals and 29 patients were immunolabeled, run on an FACScalibur flow cytometer, and analyzed by FlowJo software. RESULTS The relative quantity of dysferlin was expressed as mean fluorescence intensity (MFI). Performance of this diagnostic test was assessed by calculating likelihood ratios at different MFI cut-off points, which allowed definition of 4 disease classification groups in a simplified algorithm. CONCLUSION The MFI value may differentiate patients with dysferlinopathy from healthy individuals; it may be a useful marker for screening purposes. Muscle Nerve 54: 1064-1071, 2016.
Collapse
Affiliation(s)
- Laura Sánchez-Chapul
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Miguel Del Ángel-Muñoz
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Luis Ruano-Calderón
- Subdirección de Enseñanza y Capacitación, Investigación y Calidad en Salud, Secretaría de Salud del Estado de Durango (SSED), Durango City, Mexico
| | - Alexandra Luna-Angulo
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Ramón Coral-Vázquez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional (ESM-IPN), Mexico City, Mexico
| | | | - Jonathan J Magaña
- Laboratorio de Medicina Genómica, Servicio de Genética, (INR), Mexico City, Mexico
| | | | | | - Steven Vargas
- Laboratorio de Patología Experimental, Instituto Nacional de Neurología y Neurocirugía (INNN) "Manuel Velasco Suárez", Mexico City, Mexico
| |
Collapse
|
54
|
Griffin DA, Johnson RW, Whitlock JM, Pozsgai ER, Heller KN, Grose WE, Arnold WD, Sahenk Z, Hartzell HC, Rodino-Klapac LR. Defective membrane fusion and repair in Anoctamin5-deficient muscular dystrophy. Hum Mol Genet 2016; 25:1900-1911. [PMID: 26911675 PMCID: PMC5062581 DOI: 10.1093/hmg/ddw063] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/22/2016] [Indexed: 11/15/2022] Open
Abstract
Limb-girdle muscular dystrophies are a genetically diverse group of diseases characterized by chronic muscle wasting and weakness. Recessive mutations in ANO5 (TMEM16E) have been directly linked to several clinical phenotypes including limb-girdle muscular dystrophy type 2L and Miyoshi myopathy type 3, although the pathogenic mechanism has remained elusive. ANO5 is a member of the Anoctamin/TMEM16 superfamily that encodes both ion channels and regulators of membrane phospholipid scrambling. The phenotypic overlap of ANO5 myopathies with dysferlin-associated muscular dystrophies has inspired the hypothesis that ANO5, like dysferlin, may be involved in the repair of muscle membranes following injury. Here we show that Ano5-deficient mice have reduced capacity to repair the sarcolemma following laser-induced damage, exhibit delayed regeneration after cardiotoxin injury and suffer from defective myoblast fusion necessary for the proper repair and regeneration of multinucleated myotubes. Together, these data suggest that ANO5 plays an important role in sarcolemmal membrane dynamics. Genbank Mouse Genome Informatics accession no. 3576659.
Collapse
Affiliation(s)
- Danielle A Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - Ryan W Johnson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric R Pozsgai
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA and
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - William E Grose
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - W David Arnold
- Department of Neurology, Department of Physical Medicine and Rehabilitation, Department of Neuroscience and
| | - Zarife Sahenk
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Department of Neurology
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Louise R Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA and
| |
Collapse
|
55
|
Urao N, Mirza RE, Heydemann A, Garcia J, Koh TJ. Thrombospondin-1 levels correlate with macrophage activity and disease progression in dysferlin deficient mice. Neuromuscul Disord 2016; 26:240-51. [PMID: 26927626 DOI: 10.1016/j.nmd.2016.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/24/2015] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
Dysferlinopathy is associated with accumulation of thrombospondin (TSP)-1 and macrophages, both of which may contribute to the pathogenesis of the disease. The purpose of this study was to determine whether TSP-1 levels can predict macrophage activity and disease progression in dysferlin deficient BlaJ mice, focusing on the early disease process. In 3 month-old BlaJ mice, muscle TSP-1 levels exhibited strong positive correlations with both accumulation of F4/80hi macrophages and with their in vivo phagocytic activity in psoas muscles as measured by magnetic resonance imaging and flow cytometry. Muscle TSP-1 levels also exhibited a strong negative correlation with muscle mass and strong positive correlations with histological measurements of muscle fiber infiltration and regeneration. Over the course of disease progression from 3 to 12 months of age, muscle TSP-1 levels showed more complicated relationships with macrophage activity and an inverse relationship with muscle mass. Importantly, blood TSP-1 levels showed strong correlations with macrophage activity and muscle degeneration, particularly early in disease progression in BlaJ mice. These data indicate that TSP-1 may contribute to a destructive macrophage response in dysferlinopathy and pose the intriguing possibility that TSP-1 levels may serve as a biomarker for disease progression.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and 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
| | - Ahlke Heydemann
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jesus Garcia
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
56
|
Wei B, Wei H, Jin JP. Dysferlin deficiency blunts β-adrenergic-dependent lusitropic function of mouse heart. J Physiol 2015; 593:5127-44. [PMID: 26415898 DOI: 10.1113/jp271225] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/22/2015] [Indexed: 11/08/2022] Open
Abstract
Dysferlin is a cell membrane bound protein with a role in the repair of skeletal and cardiac muscle cells. Deficiency of dysferlin leads to limb-girdle muscular dystrophy 2B (LGMD2B) and Miyoshi myopathy. In cardiac muscle, dysferlin is located at the intercalated disc and transverse tubule membranes. Loss of dysferlin causes death of cardiomyocytes, notably in ageing hearts, leading to dilated cardiomyopathy and heart failure in LGM2B patients. To understand the primary pathogenesis and pathophysiology of dysferlin cardiomyopathy, we studied cardiac phenotypes of young adult dysferlin knockout mice and found early myocardial hypertrophy with largely compensated baseline cardiac function. Cardiomyocytes isolated from dysferlin-deficient mice showed normal shortening and re-lengthening velocities in the absence of external load with normal peak systolic Ca(2+) but slower Ca(2+) re-sequestration than wild-type controls. The effects of isoproterenol on relaxation velocity, left ventricular systolic pressure and stroke volume were blunted in dysferlin-deficient mouse hearts compared with that in wild-type hearts. Young dysferlin-deficient mouse hearts expressed normal isoforms of myofilament proteins whereas the phosphorylation of ventricular myosin light chain 2 was significantly increased, implying a molecular response to the impaired lusitropic function. These early phenotypes of diastolic cardiac dysfunction and blunted lusitropic response of cardiac muscle to β-adrenergic stimulation indicate a novel pathogenic mechanism of dysferlin cardiomyopathy.
Collapse
Affiliation(s)
- Bin Wei
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Hongguang Wei
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - J-P Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
57
|
Babiychuk EB, Draeger A. Defying death: Cellular survival strategies following plasmalemmal injury by bacterial toxins. Semin Cell Dev Biol 2015; 45:39-47. [PMID: 26481974 DOI: 10.1016/j.semcdb.2015.10.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
The perforation of the plasmalemma by pore-forming toxins causes an influx of Ca(2+) and an efflux of cytoplasmic constituents. In order to ensure survival, the cell needs to identify, plug and remove lesions from its membrane. Quarantined by membrane folds and isolated by membrane fusion, the pores are removed from the plasmalemma and expelled into the extracellular space. Outward vesiculation and microparticle shedding seem to be the strategies of choice to eliminate toxin-perforated membrane regions from the plasmalemma of host cells. Depending on the cell type and the nature of injury, the membrane lesion can also be taken up by endocytosis and degraded internally. Host cells make excellent use of an initial, moderate rise in intracellular [Ca(2+)], which triggers containment of the toxin-inflicted damage and resealing of the damaged plasmalemma. Additional Ca(2+)-dependent defensive cellular actions range from the release of effector molecules in order to warn neighbouring cells, to the activation of caspases for the initiation of apoptosis in order to eliminate heavily damaged, dysregulated cells. Injury to the plasmalemma by bacterial toxins can be prevented by the early sequestration of bacterial toxins. Artificial liposomes can act as a decoy system preferentially binding and neutralizing bacterial toxins.
Collapse
|
58
|
Schilling JM, Patel HH. Non-canonical roles for caveolin in regulation of membrane repair and mitochondria: implications for stress adaptation with age. J Physiol 2015; 594:4581-9. [PMID: 26333003 DOI: 10.1113/jp270591] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 08/04/2015] [Indexed: 12/22/2022] Open
Abstract
Many different theories of ageing have been proposed but none has served the unifying purpose of defining a molecular target that can limit the structural and functional decline associated with age that ultimately leads to the demise of the organism. We propose that the search for a molecule with these unique properties must account for regulation of the signalling efficiency of multiple cellular functions that degrade with age due to a loss of a particular protein. We suggest caveolin as one such molecule that serves as a regulator of key processes in signal transduction. We define a particular distinction between cellular senescence and ageing and propose that caveolin plays a distinct role in each of these processes. Caveolin is traditionally thought of as a membrane-localized protein regulating signal transduction via membrane enrichment of specific signalling molecules. Ultimately we focus on two non-canonical roles for caveolin - membrane repair and regulation of mitochondrial function - which may be novel features of stress adaptation, especially in the setting of ageing. The end result of preserving membrane structure and mitochondrial function is maintenance of homeostatic signalling, preserving barrier function, and regulating energy production for cell survival and resilient ageing.
Collapse
Affiliation(s)
- Jan M Schilling
- VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA.,Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hemal H Patel
- VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA.,Department of Anesthesiology, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
59
|
Cai C, Lin P, Zhu H, Ko JK, Hwang M, Tan T, Pan Z, Korichneva I, Ma J. Zinc Binding to MG53 Protein Facilitates Repair of Injury to Cell Membranes. J Biol Chem 2015; 290:13830-9. [PMID: 25869134 DOI: 10.1074/jbc.m114.620690] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Indexed: 01/19/2023] Open
Abstract
Zinc is an essential trace element that participates in a wide range of biological functions, including wound healing. Although Zn(2+) deficiency has been linked to compromised wound healing and tissue repair in human diseases, the molecular mechanisms underlying Zn(2+)-mediated tissue repair remain unknown. Our previous studies established that MG53, a TRIM (tripartite motif) family protein, is an essential component of the cell membrane repair machinery. Domain homology analysis revealed that MG53 contains two Zn(2+)-binding motifs. Here, we show that Zn(2+) binding to MG53 is indispensable to assembly of the cell membrane repair machinery. Live cell imaging illustrated that Zn(2+) entry from extracellular space is essential for translocation of MG53-containing vesicles to the acute membrane injury sites for formation of a repair patch. The effect of Zn(2+) on membrane repair is abolished in mg53(-/-) muscle fibers, suggesting that MG53 functions as a potential target for Zn(2+) during membrane repair. Mutagenesis studies suggested that both RING and B-box motifs of MG53 constitute Zn(2+)-binding domains that contribute to MG53-mediated membrane repair. Overall, this study establishes a base for Zn(2+) interaction with MG53 in protection against injury to the cell membrane.
Collapse
Affiliation(s)
- Chuanxi Cai
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, the Center for Cardiovascular Sciences, Albany Medical College, Albany, New York 12208
| | - Peihui Lin
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, the Department of Surgery
| | - Hua Zhu
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, the Department of Surgery
| | - Jae-Kyun Ko
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | - Moonsun Hwang
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854
| | | | - Zui Pan
- Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, and
| | - Irina Korichneva
- the Laboratory of Cellular and Molecular Physiology, Department of Sciences, University of Picardie, Amiens 80000, France
| | - Jianjie Ma
- From the Department of Physiology and Biophysics, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, the Department of Surgery,
| |
Collapse
|
60
|
Bouter A, Carmeille R, Gounou C, Bouvet F, Degrelle SA, Evain-Brion D, Brisson AR. Review: Annexin-A5 and cell membrane repair. Placenta 2015; 36 Suppl 1:S43-9. [PMID: 25701430 DOI: 10.1016/j.placenta.2015.01.193] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 11/16/2022]
Abstract
Annexins are soluble proteins that bind to biological membranes containing negatively charged phospholipids, principally phosphatidylserine, in a Ca(2+)-dependent manner. Annexin-A5 (AnxA5), the smallest member of the annexin family, presents unique properties of membrane binding and self-assembly into ordered two-dimensional (2D) arrays on membrane surfaces. We have previously reported that AnxA5 plays a central role in the machinery of membrane repair by enabling rapid resealing of plasma membrane disruption in murine perivascular cells. AnxA5 promotes membrane repair via the formation of a protective 2D bandage at membrane damaged site. Here, we review current knowledge on cell membrane repair and present recent findings on the role of AnxA5 in membrane resealing of human trophoblasts.
Collapse
Affiliation(s)
- A Bouter
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France.
| | - R Carmeille
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| | - C Gounou
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| | - F Bouvet
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| | - S A Degrelle
- Fondation PremUP, Paris F-75006, France; INSERM, U1139, Paris F-75006, France; Université Paris Descartes, UMR-S1139 Sorbonne Paris Cité, Paris F-75006, France
| | - D Evain-Brion
- Fondation PremUP, Paris F-75006, France; INSERM, U1139, Paris F-75006, France; Université Paris Descartes, UMR-S1139 Sorbonne Paris Cité, Paris F-75006, France
| | - A R Brisson
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| |
Collapse
|
61
|
Carmeille R, Degrelle SA, Plawinski L, Bouvet F, Gounou C, Evain-Brion D, Brisson AR, Bouter A. Annexin-A5 promotes membrane resealing in human trophoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2033-44. [PMID: 25595530 DOI: 10.1016/j.bbamcr.2014.12.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/25/2014] [Accepted: 12/27/2014] [Indexed: 01/12/2023]
Abstract
Annexin-A5 (AnxA5) is the smallest member of the annexins, a group of soluble proteins that bind to membranes containing negatively-charged phospholipids, principally phosphatidylserine, in a Ca(2+)-dependent manner. AnxA5 presents unique properties of binding and self-assembling on membrane surfaces, forming highly ordered two-dimensional (2D) arrays. We showed previously that AnxA5 plays a central role in the machinery of cell membrane repair of murine perivascular cells, promoting the resealing of membrane damages via the formation of 2D protein arrays at membrane disrupted sites and preventing the extension of membrane ruptures. As the placenta is one of the richest source of AnxA5 in humans, we investigated whether AnxA5 was involved in membrane repair in this organ. We addressed this question at the level of human trophoblasts, either mononucleated cytotrophoblasts or multinucleated syncytiotrophoblasts, in choriocarcinoma cells and primary trophoblasts. Using established procedure of laser irradiation and fluorescence microscopy, we observed that both human cytotrophoblasts and syncytiotrophoblasts repair efficiently a μm²-size disruption. Compared to wild-type cells, AnxA5-deficient trophoblasts exhibit severe defect of membrane repair. Through specifically binding to the disrupted site as early as a few seconds after membrane wounding, AnxA5 promotes membrane resealing of injured human trophoblasts. In addition, we observed that a large membrane area containing the disrupted site was released in the extracellular milieu. We propose mechanisms ensuring membrane resealing and subsequent lesion removal in human trophoblasts. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Romain Carmeille
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France
| | - Séverine A Degrelle
- Fondation PremUP, Paris, F-75006, France; INSERM, U1139, Paris, F-75006, France; Université Paris Descartes, UMR-S1139 Sorbonne Pris Cité, Paris, F-75006, France
| | - Laurent Plawinski
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France
| | - Flora Bouvet
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France
| | - Céline Gounou
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France
| | - Danièle Evain-Brion
- Fondation PremUP, Paris, F-75006, France; INSERM, U1139, Paris, F-75006, France; Université Paris Descartes, UMR-S1139 Sorbonne Pris Cité, Paris, F-75006, France
| | - Alain R Brisson
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France
| | - Anthony Bouter
- Institute of Chemistry and Biology of Membranes and Nano-objects, UMR 5248, CNRS, University of Bordeaux, IPB, F-33600 Pessac, France.
| |
Collapse
|
62
|
Dominov JA, Uyan O, Sapp PC, McKenna-Yasek D, Nallamilli BRR, Hegde M, Brown RH. A novel dysferlin mutant pseudoexon bypassed with antisense oligonucleotides. Ann Clin Transl Neurol 2014; 1:703-20. [PMID: 25493284 PMCID: PMC4241797 DOI: 10.1002/acn3.96] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022] Open
Abstract
Objective Mutations in dysferlin (DYSF), a Ca2+-sensitive ferlin family protein important for membrane repair, vesicle trafficking, and T-tubule function, cause Miyoshi myopathy, limb-girdle muscular dystrophy type 2B, and distal myopathy. More than 330 pathogenic DYSF mutations have been identified within exons or near exon–intron junctions. In ~17% of patients who lack normal DYSF, only a single disease-causing mutation has been identified. We studied one family with one known mutant allele to identify both the second underlying genetic defect and potential therapeutic approaches. Methods We sequenced the full DYSF cDNA and investigated antisense oligonucleotides (AONs) as a tool to modify splicing of the mRNA transcripts in order to process out mutant sequences. Results We identified a novel pseudoexon between exons 44 and 45, (pseudoexon 44.1, PE44.1), which inserts an additional 177 nucleotides into the mRNA and 59 amino acids within the conserved C2F domain of the DYSF protein. Two unrelated dysferlinopathy patients were also found to carry this mutation. Using AONs targeting PE44.1, we blocked the abnormal splicing event, yielding normal, full-length DYSF mRNA, and increased DYSF protein expression. Interpretation This is the first report of a deep intronic mutation in DYSF that alters mRNA splicing to include a mutant peptide fragment within a key DYSF domain. We report that AON-mediated exon-skipping restores production of normal, full-length DYSF in patients’ cells in vitro, offering hope that this approach will be therapeutic in this genetic context, and providing a foundation for AON therapeutics targeting other pathogenic DYSF alleles.
Collapse
Affiliation(s)
- Janice A Dominov
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Ozgün Uyan
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Peter C Sapp
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Diane McKenna-Yasek
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Babi R R Nallamilli
- Department of Human Genetics, Emory University School of Medicine Atlanta, Georgia, 30322
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine Atlanta, Georgia, 30322
| | - Robert H Brown
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| |
Collapse
|
63
|
Li X, Xiao Y, Cui Y, Tan T, Narasimhulu CA, Hao H, Liu L, Zhang J, He G, Verfaillie CM, Lei M, Parthasarathy S, Ma J, Zhu H, Liu Z. Cell membrane damage is involved in the impaired survival of bone marrow stem cells by oxidized low-density lipoprotein. J Cell Mol Med 2014; 18:2445-53. [PMID: 25256620 PMCID: PMC4302650 DOI: 10.1111/jcmm.12424] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022] Open
Abstract
Cell therapy with bone marrow stem cells (BMSCs) remains a viable option for tissue repair and regeneration. A major challenge for cell therapy is the limited cell survival after implantation. This study was to investigate the effect of oxidized low-density lipoprotein (ox-LDL, naturally present in human blood) on BMSC injury and the effect of MG53, a tissue repair protein, for the improvement of stem cell survival. Rat bone marrow multipotent adult progenitor cells (MAPCs) were treated with ox-LDL, which caused significant cell death as reflected by the increased LDH release to the media. Exposure of MAPCs to ox-LDL led to entry of fluorescent dye FM1-43 measured under confocal microscope, suggesting damage to the plasma membrane. Ox-LDL also generated reactive oxygen species (ROS) as measured with electron paramagnetic resonance spectroscopy. While antioxidant N-acetylcysteine completely blocked ROS production from ox-LDL, it failed to prevent ox-LDL-induced cell death. When MAPCs were treated with the recombinant human MG53 protein (rhMG53) ox-LDL induced LDH release and FM1-43 dye entry were significantly reduced. In the presence of rhMG53, the MAPCs showed enhanced cell survival and proliferation. Our data suggest that membrane damage induced by ox-LDL contributed to the impaired survival of MAPCs. rhMG53 treatment protected MAPCs against membrane damage and enhanced their survival which might represent a novel means for improving efficacy for stem cell-based therapy for treatment of diseases, especially in setting of hyperlipidemia.
Collapse
Affiliation(s)
- Xin Li
- Xiangya Hospital of Central South University, Changsha, Hunan, China; Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Neuromuscular pathology case. J Clin Neuromuscul Dis 2014; 16:15-9. [PMID: 25137511 DOI: 10.1097/cnd.0000000000000043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
65
|
Emerging role of polymerase-1 and transcript release factor (PTRF/ Cavin-1) in health and disease. Cell Tissue Res 2014; 357:505-13. [PMID: 25107607 DOI: 10.1007/s00441-014-1964-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 07/04/2014] [Indexed: 01/06/2023]
Abstract
Polymerase-1 and release transcript factor (PTRF) was initially reported to be involved in the termination of the transcription process. More recently, it has been implicated in the formation of caveolae, cave-like structures in the plasma membrane. The effects of PTRF related to caveolae suggest that this protein may play important roles in health and disease. PTRF is highly expressed in various cells, including adipocytes, osteoblasts and muscle (cardiac, skeletal and smooth) cells. The role of PTRF in prostate cancer has been recently reviewed but there is growing evidence that PTRF is involved in other physiological processes such as cell repair and the regulation of glucose and lipid metabolism and, furthermore, altered expression of PTRF may be associated with disease. This review discusses the emerging role of PTRF in health and disease.
Collapse
|
66
|
Kim SC, Kellett T, Wang S, Nishi M, Nagre N, Zhou B, Flodby P, Shilo K, Ghadiali SN, Takeshima H, Hubmayr RD, Zhao X. TRIM72 is required for effective repair of alveolar epithelial cell wounding. Am J Physiol Lung Cell Mol Physiol 2014; 307:L449-59. [PMID: 25106429 DOI: 10.1152/ajplung.00172.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms for lung cell repair are largely unknown. Previous studies identified tripartite motif protein 72 (TRIM72) from striated muscle and linked its function to tissue repair. In this study, we characterized TRIM72 expression in lung tissues and investigated the role of TRIM72 in repair of alveolar epithelial cells. In vivo injury of lung cells was introduced by high tidal volume ventilation, and repair-defective cells were labeled with postinjury administration of propidium iodide. Primary alveolar epithelial cells were isolated and membrane wounding and repair were labeled separately. Our results show that absence of TRIM72 increases susceptibility to deformation-induced lung injury whereas TRIM72 overexpression is protective. In vitro cell wounding assay revealed that TRIM72 protects alveolar epithelial cells through promoting repair rather than increasing resistance to injury. The repair function of TRIM72 in lung cells is further linked to caveolin 1. These data suggest an essential role for TRIM72 in repair of alveolar epithelial cells under plasma membrane stress failure.
Collapse
Affiliation(s)
- Seong Chul Kim
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Kellett
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shaohua Wang
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Miyuki Nishi
- Department of Biological Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Nagaraja Nagre
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Beiyun Zhou
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Southern California, Los Angeles, California
| | - Per Flodby
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Southern California, Los Angeles, California
| | - Konstantin Shilo
- Thoracic Pathology Division, Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Samir N Ghadiali
- Biomedical Engineering Department, College of Engineering, The Ohio State University, Columbus, Ohio; and
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Rolf D Hubmayr
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Xiaoli Zhao
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio; Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
67
|
Defour A, Van der Meulen JH, Bhat R, Bigot A, Bashir R, Nagaraju K, Jaiswal JK. Dysferlin regulates cell membrane repair by facilitating injury-triggered acid sphingomyelinase secretion. Cell Death Dis 2014; 5:e1306. [PMID: 24967968 PMCID: PMC4079937 DOI: 10.1038/cddis.2014.272] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/08/2014] [Accepted: 05/20/2014] [Indexed: 01/17/2023]
Abstract
Dysferlin deficiency compromises the repair of injured muscle, but the underlying cellular mechanism remains elusive. To study this phenomenon, we have developed mouse and human myoblast models for dysferlinopathy. These dysferlinopathic myoblasts undergo normal differentiation but have a deficit in their ability to repair focal injury to their cell membrane. Imaging cells undergoing repair showed that dysferlin-deficit decreased the number of lysosomes present at the cell membrane, resulting in a delay and reduction in injury-triggered lysosomal exocytosis. We find repair of injured cells does not involve formation of intracellular membrane patch through lysosome-lysosome fusion; instead, individual lysosomes fuse with the injured cell membrane, releasing acid sphingomyelinase (ASM). ASM secretion was reduced in injured dysferlinopathic cells, and acute treatment with sphingomyelinase restored the repair ability of dysferlinopathic myoblasts and myofibers. Our results provide the mechanism for dysferlin-mediated repair of skeletal muscle sarcolemma and identify ASM as a potential therapy for dysferlinopathy.
Collapse
Affiliation(s)
- A Defour
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - J H Van der Meulen
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - R Bhat
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - A Bigot
- Institut de Myologie, UM76 Université Pierre et Marie Curie, U974 INSERM, UMR7215 CNRS, GH Pitié-Salpétrière, 47 bd de l'Hôpital, Paris, France
| | - R Bashir
- School of Biological and Biochemical Sciences, University of Durham, Durham, UK
| | - K Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - J K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
68
|
Disciglio V, Rizzo CL, Mencarelli MA, Mucciolo M, Marozza A, Di Marco C, Massarelli A, Canocchi V, Baldassarri M, Ndoni E, Frullanti E, Amabile S, Anderlid BM, Metcalfe K, Le Caignec C, David A, Fryer A, Boute O, Joris A, Greco D, Pecile V, Battini R, Novelli A, Fichera M, Romano C, Mari F, Renieri A. Interstitial 22q13 deletions not involving SHANK3 gene: A new contiguous gene syndrome. Am J Med Genet A 2014; 164A:1666-76. [DOI: 10.1002/ajmg.a.36513] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 01/03/2014] [Indexed: 01/15/2023]
Affiliation(s)
| | - Caterina Lo Rizzo
- Medical Genetics; University of Siena; Siena Italy
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| | - Maria Antonietta Mencarelli
- Medical Genetics; University of Siena; Siena Italy
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| | | | - Annabella Marozza
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| | - Chiara Di Marco
- Medical Genetics; University of Siena; Siena Italy
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| | | | | | | | - Enea Ndoni
- Medical Genetics; University of Siena; Siena Italy
| | | | | | - Britt Marie Anderlid
- Department of Molecular Medicine and Surgery; CMM, Karolinska Institutet and Hospital Stockholm; Sweden
| | - Kay Metcalfe
- Manchester Academic Health Sciences Centre; Manchester Biomedical Research Centre; St Mary's Hospital; Manchester United Kingdom
| | | | - Albert David
- CHU Nantes; Service de genetique medicale; Nantes Cedex France
| | - Alan Fryer
- Department of Clinical Genetics; Alder Hey Children's Hospital; Liverpool, and Liverpool Women's Hospital; Liverpool United Kingdom
| | - Odile Boute
- Service de Génétique Clinique; Hôpital Jeanne de Flandre; Lille France
| | - Andrieux Joris
- Institut de Génétique Médicale; Hôpital Jeanne de Flandre; Lille France
| | - Donatella Greco
- Unit of Pediatrics and Medical Genetics; IRCCS Associazione Oasi Maria Santissima; Troina Italy
| | - Vanna Pecile
- Medical Genetics; Institute for Maternal and Child Health IRCCS “Burlo Garofalo”; Trieste Italy
| | - Roberta Battini
- Department of Developmental Neuroscience; IRCCS Stella Maris; Calambrone Italy
| | - Antonio Novelli
- IRCCS Casa Sollievo della Sofferenza Hospital; Mendel Institute; Rome Italy
| | - Marco Fichera
- Laboratory of Genetic Diagnosis; IRCCS Associazione Oasi Maria Santissima; Troina Italy
- Medical Genetics; University of Catania; Catania Italy
| | - Corrado Romano
- Unit of Pediatrics and Medical Genetics; IRCCS Associazione Oasi Maria Santissima; Troina Italy
| | - Francesca Mari
- Medical Genetics; University of Siena; Siena Italy
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| | - Alessandra Renieri
- Medical Genetics; University of Siena; Siena Italy
- Genetica Medica; Azienda Ospedaliera Universitaria Senese; Siena Italy
| |
Collapse
|
69
|
Grounds MD, Terrill JR, Radley-Crabb HG, Robertson T, Papadimitriou J, Spuler S, Shavlakadze T. Lipid accumulation in dysferlin-deficient muscles. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1668-76. [PMID: 24685690 DOI: 10.1016/j.ajpath.2014.02.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/21/2014] [Accepted: 02/25/2014] [Indexed: 02/01/2023]
Abstract
Dysferlin is a membrane associated protein involved in vesicle trafficking and fusion. Defects in dysferlin result in limb-girdle muscular dystrophy type 2B and Miyoshi myopathy in humans and myopathy in A/J(dys-/-) and BLAJ mice, but the pathomechanism of the myopathy is not understood. Oil Red O staining showed many lipid droplets within the psoas and quadriceps muscles of dysferlin-deficient A/J(dys-/-) mice aged 8 and 12 months, and lipid droplets were also conspicuous within human myofibers from patients with dysferlinopathy (but not other myopathies). Electron microscopy of 8-month-old A/J(dys-/-) psoas muscles confirmed lipid droplets within myofibers and showed disturbed architecture of myofibers. In addition, the presence of many adipocytes was confirmed, and a possible role for dysferlin in adipocytes is suggested. Increased expression of mRNA for a gene involved in early lipogenesis, CCAAT/enhancer binding protein-δ, in 3-month-old A/J(dys-/-) quadriceps (before marked histopathology is evident), indicates early induction of lipogenesis/adipogenesis within dysferlin-deficient muscles. Similar results were seen for dysferlin-deficient BLAJ mice. These novel observations of conspicuous intermyofibrillar lipid and progressive adipocyte replacement in dysferlin-deficient muscles present a new focus for investigating the mechanisms that result in the progressive decline of muscle function in dysferlinopathies.
Collapse
Affiliation(s)
- Miranda D Grounds
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia.
| | - Jessica R Terrill
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| | - Hannah G Radley-Crabb
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia; CHIRI Biosciences Research Precinct, School of Biomedical Sciences, Curtin University, Perth, Australia
| | - Terry Robertson
- Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - John Papadimitriou
- Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Berlin, Germany
| | - Tea Shavlakadze
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| |
Collapse
|
70
|
Touznik A, Lee JJA, Yokota T. New developments in exon skipping and splice modulation therapies for neuromuscular diseases. Expert Opin Biol Ther 2014; 14:809-19. [PMID: 24620745 DOI: 10.1517/14712598.2014.896335] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Antisense oligonucleotide (AON) therapy is a form of treatment for genetic or infectious diseases using small, synthetic DNA-like molecules called AONs. Recent advances in the development of AONs that show improved stability and increased sequence specificity have led to clinical trials for several neuromuscular diseases. Impressive preclinical and clinical data are published regarding the usage of AONs in exon-skipping and splice modulation strategies to increase dystrophin production in Duchenne muscular dystrophy (DMD) and survival of motor neuron (SMN) production in spinal muscular atrophy (SMA). AREAS COVERED In this review, we focus on the current progress and challenges of exon-skipping and splice modulation therapies. In addition, we discuss the recent failure of the Phase III clinical trials of exon 51 skipping (drisapersen) for DMD. EXPERT OPINION The main approach of AON therapy in DMD and SMA is to rescue ('knock up' or increase) target proteins through exon skipping or exon inclusion; conversely, most conventional antisense drugs are designed to knock down (inhibit) the target. Encouraging preclinical data using this 'knock up' approach are also reported to rescue dysferlinopathies, including limb-girdle muscular dystrophy type 2B, Miyoshi myopathy, distal myopathy with anterior tibial onset and Fukuyama congenital muscular dystrophy.
Collapse
Affiliation(s)
- Aleksander Touznik
- University of Alberta, Faculty of Medicine and Dentistry, Department of Medical Genetics , Edmonton, Alberta , Canada
| | | | | |
Collapse
|
71
|
Abstract
Skeletal muscle continuously adapts to changes in its mechanical environment through modifications in gene expression and protein stability that affect its physiological function and mass. However, mechanical stresses commonly exceed the parameters that induce adaptations, producing instead acute injury. Furthermore, the relatively superficial location of many muscles in the body leaves them further vulnerable to acute injuries by exposure to extreme temperatures, contusions, lacerations or toxins. In this article, the molecular, cellular, and mechanical factors that underlie muscle injury and the capacity of muscle to repair and regenerate are presented. Evidence shows that muscle injuries that are caused by eccentric contractions result from direct mechanical damage to myofibrils. However, muscle pathology following other acute injuries is largely attributable to damage to the muscle cell membrane. Many feaures in the injury-repair-regeneration cascade relate to the unregulated influx of calcium through membrane lesions, including: (i) activation of proteases and hydrolases that contribute muscle damage, (ii) activation of enzymes that drive the production of mitogens and motogens for muscle and immune cells involved in injury and repair, and (iii) enabling protein-protein interactions that promote membrane repair. Evidence is also presented to show that the myogenic program that is activated by acute muscle injury and the inflammatory process that follows are highly coordinated, with myeloid cells playing a central role in modulating repair and regeneration. The early-invading, proinflammatory M1 macrophages remove debris caused by injury and express Th1 cytokines that play key roles in regulating the proliferation, migration, and differentiation of satellite cells. The subsequent invasion by anti-inflammatory, M2 macrophages promotes tissue repair and attenuates inflammation. Although this system provides an effective mechanism for muscle repair and regeneration following acute injury, it is dysregulated in chronic injuries. In this article, the process of muscle injury, repair and regeneration that occurs in muscular dystrophy is used as an example of chronic muscle injury, to highlight similarities and differences between the injury and repair processes that occur in acutely and chronically injured muscle.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA.
| |
Collapse
|
72
|
Krajacic P, Pistilli EE, Tanis JE, Khurana TS, Lamitina ST. FER-1/Dysferlin promotes cholinergic signaling at the neuromuscular junction in C. elegans and mice. Biol Open 2013; 2:1245-52. [PMID: 24244862 PMCID: PMC3828772 DOI: 10.1242/bio.20135637] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/25/2013] [Indexed: 12/28/2022] Open
Abstract
Dysferlin is a member of the evolutionarily conserved ferlin gene family. Mutations in Dysferlin lead to Limb Girdle Muscular Dystrophy 2B (LGMD2B), an inherited, progressive and incurable muscle disorder. However, the molecular mechanisms underlying disease pathogenesis are not fully understood. We found that both loss-of-function mutations and muscle-specific overexpression of C. elegans fer-1, the founding member of the Dysferlin gene family, caused defects in muscle cholinergic signaling. To determine if Dysferlin-dependent regulation of cholinergic signaling is evolutionarily conserved, we examined the in vivo physiological properties of skeletal muscle synaptic signaling in a mouse model of Dysferlin-deficiency. In addition to a loss in muscle strength, Dysferlin −/− mice also exhibited a cholinergic deficit manifested by a progressive, frequency-dependent decrement in their compound muscle action potentials following repetitive nerve stimulation, which was observed in another Dysferlin mouse model but not in a Dysferlin-independent mouse model of muscular dystrophy. Oral administration of Pyridostigmine bromide, a clinically used acetylcholinesterase inhibitor (AchE.I) known to increase synaptic efficacy, reversed the action potential defect and restored in vivo muscle strength to Dysferlin −/− mice without altering muscle pathophysiology. Our data demonstrate a previously unappreciated role for Dysferlin in the regulation of cholinergic signaling and suggest that such regulation may play a significant pathophysiological role in LGMD2B disease.
Collapse
Affiliation(s)
- Predrag Krajacic
- Department of Physiology, Richards Research Building A702, University of Pennsylvania , Philadelphia, PA 19104 , USA ; Pennsylvania Muscle Institute, 700A Clinical Research Building, University of Pennsylvania , Philadelphia, PA 19104 , USA
| | | | | | | | | |
Collapse
|
73
|
Coletti D, Teodori L, Lin Z, Beranudin JF, Adamo S. Restoration versus reconstruction: cellular mechanisms of skin, nerve and muscle regeneration compared. Regen Med Res 2013; 1:4. [PMID: 25984323 PMCID: PMC4375925 DOI: 10.1186/2050-490x-1-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/20/2013] [Indexed: 01/24/2023] Open
Abstract
In tissues characterized by a high turnover or following acute injury, regeneration replaces damaged cells and is involved in adaptation to external cues, leading to homeostasis of many tissues during adult life. An understanding of the mechanics underlying tissue regeneration is highly relevant to regenerative medicine-based interventions. In order to investigate the existence a leitmotif of tissue regeneration, we compared the cellular aspects of regeneration of skin, nerve and skeletal muscle, three organs characterized by different types of anatomical and functional organization. Epidermis is a stratified squamous epithelium that migrates from the edge of the wound on the underlying dermis to rebuild lost tissue. Peripheral neurons are elongated cells whose neurites are organized in bundles, within an endoneurium of connective tissue; they either die upon injury or undergo remodeling and axon regrowth. Skeletal muscle is characterized by elongated syncytial cells, i.e. muscle fibers, that can temporarily survive in broken pieces; satellite cells residing along the fibers form new fibers, which ultimately fuse with the old ones as well as with each other to restore the previous organization. Satellite cell asymmetrical division grants a reservoir of undifferentiated cells, while other stem cell populations of muscle and non-muscle origin participate in muscle renewal. Following damage, all the tissues analyzed here go through three phases: inflammation, regeneration and maturation. Another common feature is the occurrence of cellular de-differentiation and/or differentiation events, including gene transcription, which are typical of embryonic development. Nonetheless, various strategies are used by different tissues to replace their lost parts. The epidermis regenerates ex novo, whereas neurons restore their missing parts; muscle fibers use a mixed strategy, based on the regrowth of missing parts through reconstruction by means of newborn fibers. The choice of either strategy is influenced by the anatomical, physical and chemical features of the cells as well as by the extracellular matrix typical of a given tissue, which points to the existence of differential, evolutionary-based mechanisms for specific tissue regeneration. The shared, ordered sequence of steps that characterize the regeneration processes examined suggests it may be possible to model this extremely important phenomenon to reproduce multicellular organisms.
Collapse
Affiliation(s)
- Dario Coletti
- UPMC Univ Paris 06, UR4 Ageing, Stress, Inflammation, 75005 Paris, France ; Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology & Medical Embryology, 00161 Rome, Italy ; Interuniversity Institute of Myology, Kragujevac, Italy
| | - Laura Teodori
- ENEA-Frascati, UTAPRAD-DIM, Diagnostics and Metrology Laboratory, 00044 Rome, Italy
| | - Zhenlin Lin
- UPMC Univ Paris 06, UR4 Ageing, Stress, Inflammation, 75005 Paris, France
| | | | - Sergio Adamo
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology & Medical Embryology, 00161 Rome, Italy ; Interuniversity Institute of Myology, Kragujevac, Italy
| |
Collapse
|
74
|
Abstract
The distal myopathies are a heterogeneous group of genetic disorders defined by a predominant distal weakness at onset or throughout the evolution of the disease and by pathological data supporting a myopathic process. The number of genes associated with distal myopathies continues to increase. Fourteen distinct distal myopathies are currently defined by their gene and causative mutations, compared to just five entities delineated on clinical grounds two decades ago. The known proteins affected in the distal myopathies are of many types and include a significant number of sarcomeric proteins. The useful indicators for clinicians to direct towards a correct molecular diagnosis are the mode of inheritance, the age at onset, the pattern of muscle involvement, the serum creatine kinase level and the muscle pathology findings. This review gives an overview of the clinical and genetic characteristics of the currently identified distal myopathies with emphasis on some recent findings.
Collapse
|
75
|
Ceco E, McNally EM. Modifying muscular dystrophy through transforming growth factor-β. FEBS J 2013; 280:4198-209. [PMID: 23551962 PMCID: PMC3731412 DOI: 10.1111/febs.12266] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/11/2013] [Accepted: 03/20/2013] [Indexed: 12/31/2022]
Abstract
Muscular dystrophy arises from ongoing muscle degeneration and insufficient regeneration. This imbalance leads to loss of muscle, with replacement by scar or fibrotic tissue, resulting in muscle weakness and, eventually, loss of muscle function. Human muscular dystrophy is characterized by a wide range of disease severity, even when the same genetic mutation is present. This variability implies that other factors, both genetic and environmental, modify the disease outcome. There has been an ongoing effort to define the genetic and molecular bases that influence muscular dystrophy onset and progression. Modifier genes for muscle disease have been identified through both candidate gene approaches and genome-wide surveys. Multiple lines of experimental evidence have now converged on the transforming growth factor-β (TGF-β) pathway as a modifier for muscular dystrophy. TGF-β signaling is upregulated in dystrophic muscle as a result of a destabilized plasma membrane and/or an altered extracellular matrix. Given the important biological role of the TGF-β pathway, and its role beyond muscle homeostasis, we review modifier genes that alter the TGF-β pathway and approaches to modulate TGF-β activity to ameliorate muscle disease.
Collapse
Affiliation(s)
- Ermelinda Ceco
- Committee on Cell Physiology, University of Chicago, IL 60637, USA
| | | |
Collapse
|
76
|
Xiong Y, Ai D, Meng P, Wei M, Hong Y, Zhang M, Huang L, Fu Z, Shi Y, Lin J. Cloning, expression, and preliminary characterization of the dysferlin tegument protein in Schistosoma japonicum. Parasitol Int 2013; 62:522-9. [PMID: 23892179 DOI: 10.1016/j.parint.2013.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 07/11/2013] [Accepted: 07/12/2013] [Indexed: 12/11/2022]
Abstract
The schistosomal tegument is a dynamic host-interactive layer. Proteins exposed to the host on the tegumental surface are important for completion of the parasitic lifecycle. Dysferlin is a member of the ferlin family and is involved in plasma membrane repair. Based on the results of a proteomics study of tegument surface proteins of Schistosoma japonicum in our laboratory, dysferlin was identified as a tegumental protein of S. japonicum. The gene encoding S. japonicum dysferlin (SjDF), which codes for several Ca(2+) binding sites, was cloned, expressed in Escherichia coli, and characterized. Western blot analysis revealed that recombinant SjDF had good immunogenicity. Real-time RT-PCR analysis showed that SjDF was upregulated mainly in adult worms and the transcription level in 42-day-old female worms was significantly higher than that in males. Immunofluorescence analysis revealed that SjDF was mainly distributed in the tegument at various developmental stages. Experimental mice were treated with praziquantel and at 35days post-infection, we noted that damage to the tegument and subtegument worsened and did not recover at 36h post-treatment in the high-dose group and was accompanied by downregulation of SjDF mRNA, while the damage was less severe and recovered by this time in the low-dose group, and accompanied by upregulation of SjDF. Our results suggested that SjDF is a tegumental protein that may be important in schistosomal development and may participate in the repair process in muscle and tegument, and could present a viable vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- Yanian Xiong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Flix B, de la Torre C, Castillo J, Casal C, Illa I, Gallardo E. Dysferlin interacts with calsequestrin-1, myomesin-2 and dynein in human skeletal muscle. Int J Biochem Cell Biol 2013; 45:1927-38. [PMID: 23792176 DOI: 10.1016/j.biocel.2013.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/24/2013] [Accepted: 06/09/2013] [Indexed: 11/25/2022]
Abstract
Dysferlinopathies are a group of progressive muscular dystrophies characterized by mutations in the gene DYSF. These mutations cause scarcity or complete absence of dysferlin, a protein that is expressed in skeletal muscle and plays a role in membrane repair. Our objective was to unravel the proteins that constitute the dysferlin complex and their interaction within the complex using immunoprecipitation assays (IP), blue native gel electrophoresis (BN) in healthy adult skeletal muscle and healthy cultured myotubes, and fluorescence lifetime imaging-fluorescence resonance energy transfer (FLIM-FRET) analysis in healthy myotubes. The combination of immunoprecipitations and blue native electrophoresis allowed us to identify previously reported partners of dysferlin - such as caveolin-3, AHNAK, annexins, or Trim72/MG53 - and new interacting partners. Fluorescence lifetime imaging showed a direct interaction of dysferlin with Trim72/MG53, AHNAK, cytoplasmic dynein, myomesin-2 and calsequestrin-1, but not with caveolin-3 or dystrophin. In conclusion, although IP and BN are useful tools to identify the proteins in a complex, techniques such as fluorescence lifetime imaging analysis are needed to determine the direct and indirect interactions of these proteins within the complex. This knowledge may help us to better understand the roles of dysferlin in muscle tissue and identify new genes involved in muscular dystrophies in which the responsible gene is unknown.
Collapse
Affiliation(s)
- Bàrbara Flix
- Servei de Neurologia, Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
78
|
Perturbations in skeletal muscle sarcomere structure in patients with heart failure and Type 2 diabetes: restorative effects of (−)-epicatechinrich cocoa. Clin Sci (Lond) 2013; 125:383-9. [DOI: 10.1042/cs20130023] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
HF (heart failure) and T2D (Type 2 diabetes) associate with detrimental alterations in SkM (skeletal muscle) structure/function. We have demonstrated recently that (−)-ERC (epicatechin-rich cocoa) improves SkM mitochondrial structure [Taub, Ramirez-Sanchez, Ciaraldi, Perkins, Murphy, Naviaux, Hogan, Ceballos, Maisel, Henry et al. (2012) Clin. Trans. Sci. 5, 43–47]. We hypothesized that an improved mitochondrial structure may facilitate the reversal of detrimental alterations in sarcomeric microstructure. In a pilot study, five patients with HF and T2D consumed ERC for 3 months; treadmill testing [V̇O2max (maximum oxygen consumption)] and SkM biopsies were performed. Western blot analysis, immunohistochemistry and electron microscopy were used. We report severe perturbations in components of the DAPC (dystrophin-associated protein complex) as well as sarcomeric microstructure at baseline. ERC induced recovery/enhancement of DAPC protein levels, sarcomeric microstructure and, in a co-ordinated fashion, alterations in markers of SkM growth/differentiation consistent with myofibre regeneration. V̇O2max increased (~24%) but did not reach statistical significance. These initial results warrant further rigorous investigation, since the use of ERC (or pure epicatechin) may represent a safe and novel means of improving muscle function.
Collapse
|
79
|
Muscular dystrophy in dysferlin-deficient mouse models. Neuromuscul Disord 2013; 23:377-87. [DOI: 10.1016/j.nmd.2013.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 01/09/2013] [Accepted: 02/05/2013] [Indexed: 11/17/2022]
|
80
|
Terrill JR, Radley-Crabb HG, Iwasaki T, Lemckert FA, Arthur PG, Grounds MD. Oxidative stress and pathology in muscular dystrophies: focus on protein thiol oxidation and dysferlinopathies. FEBS J 2013; 280:4149-64. [PMID: 23332128 DOI: 10.1111/febs.12142] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 12/23/2022]
Abstract
The muscular dystrophies comprise more than 30 clinical disorders that are characterized by progressive skeletal muscle wasting and degeneration. Although the genetic basis for many of these disorders has been identified, the exact mechanism for pathogenesis generally remains unknown. It is considered that disturbed levels of reactive oxygen species (ROS) contribute to the pathology of many muscular dystrophies. Reactive oxygen species and oxidative stress may cause cellular damage by directly and irreversibly damaging macromolecules such as proteins, membrane lipids and DNA; another major cellular consequence of reactive oxygen species is the reversible modification of protein thiol side chains that may affect many aspects of molecular function. Irreversible oxidative damage of protein and lipids has been widely studied in Duchenne muscular dystrophy, and we have recently identified increased protein thiol oxidation in dystrophic muscles of the mdx mouse model for Duchenne muscular dystrophy. This review evaluates the role of elevated oxidative stress in Duchenne muscular dystrophy and other forms of muscular dystrophies, and presents new data that show significantly increased protein thiol oxidation and high levels of lipofuscin (a measure of cumulative oxidative damage) in dysferlin-deficient muscles of A/J mice at various ages. The significance of this elevated oxidative stress and high levels of reversible thiol oxidation, but minimal myofibre necrosis, is discussed in the context of the disease mechanism for dysferlinopathies, and compared with the situation for dystrophin-deficient mdx mice.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
81
|
Tabebordbar M, Wang ET, Wagers AJ. Skeletal muscle degenerative diseases and strategies for therapeutic muscle repair. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:441-75. [PMID: 23121053 DOI: 10.1146/annurev-pathol-011811-132450] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skeletal muscle is a highly specialized, postmitotic tissue that must withstand chronic mechanical and physiological stress throughout life to maintain proper contractile function. Muscle damage or disease leads to progressive weakness and disability, and manifests in more than 100 different human disorders. Current therapies to treat muscle degenerative diseases are limited mostly to the amelioration of symptoms, although promising new therapeutic directions are emerging. In this review, we discuss the pathological basis for the most common muscle degenerative diseases and highlight new and encouraging experimental and clinical opportunities to prevent or reverse these afflictions.
Collapse
Affiliation(s)
- Mohammadsharif Tabebordbar
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | | | | |
Collapse
|
82
|
Hosur V, Kavirayani A, Riefler J, Carney LMB, Lyons B, Gott B, Cox GA, Shultz LD. Dystrophin and dysferlin double mutant mice: a novel model for rhabdomyosarcoma. Cancer Genet 2012; 205:232-41. [PMID: 22682622 DOI: 10.1016/j.cancergen.2012.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 02/29/2012] [Accepted: 03/30/2012] [Indexed: 01/07/2023]
Abstract
Although researchers have yet to establish a link between muscular dystrophy (MD) and sarcomas in human patients, literature suggests that the MD genes dystrophin and dysferlin act as tumor suppressor genes in mouse models of MD. For instance, dystrophin-deficient mdx and dysferlin-deficient A/J mice, models of human Duchenne MD and limb-girdle MD type 2B, respectively, develop mixed sarcomas with variable penetrance and latency. To further establish the correlation between MD and sarcoma development, and to test whether a combined deletion of dystrophin and dysferlin exacerbates MD and augments the incidence of sarcomas, we generated dystrophin and dysferlin double mutant mice (STOCK-Dysf(prmd)Dmd(mdx-5Cv)). Not surprisingly, the double mutant mice develop severe MD symptoms and, moreover, develop rhabdomyosarcoma (RMS) at an average age of 12 months, with an incidence of >90%. Histological and immunohistochemical analyses, using a panel of antibodies against skeletal muscle cell proteins, electron microscopy, cytogenetics, and molecular analysis reveal that the double mutant mice develop RMS. The present finding bolsters the correlation between MD and sarcomas, and provides a model not only to examine the cellular origins but also to identify mechanisms and signal transduction pathways triggering development of RMS.
Collapse
|
83
|
Kobayashi K, Izawa T, Kuwamura M, Yamate J. Dysferlin and animal models for dysferlinopathy. J Toxicol Pathol 2012; 25:135-47. [PMID: 22907980 PMCID: PMC3392904 DOI: 10.1293/tox.25.135] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 03/16/2012] [Indexed: 12/27/2022] Open
Abstract
Dysferlin (DYSF) is involved in the membrane-repair process, in the intracellular vesicle system and in T-tubule development in skeletal muscle. It interacts with mitsugumin 53, annexins, caveolin-3, AHNAK, affixin, S100A10, calpain-3, tubulin and dihydropyridine receptor. Limb-girdle muscular dystrophy 2B (LGMD2B) and Miyoshi myopathy (MM) are muscular dystrophies associated with recessively inherited mutations in the DYSF gene. The diseases are characterized by weakness and muscle atrophy that progress slowly and symmetrically in the proximal muscles of the limb girdles. LGMD2B and MM, which are collectively termed “dysferlinopathy”, both lead to abnormalities in vesicle traffic and membrane repair at the plasma membrane in muscle fibers. SJL/J (SJL) and A/J mice are naturally occurring animal models for dysferlinopathy. Since there has been no an approach to therapy for dysferlinopathy, the immediate development of a therapeutic method for this genetic disorder is desirable. The murine models are useful in verification experiments for new therapies and they are valuable tools for identifying factors that accelerate dystrophic changes in skeletal muscle. It could be possible that the genetic or immunological background in SJL or A/J mice could modify muscle damage in experiments involving these models, because SJL and A/J mice show differences in the progress and prevalent sites of skeletal muscle lesions as well as in the gene-expression profiles of their skeletal muscle. In this review, we provide up-to-date information on the function of dysferlin, the development of possible therapies for muscle dystrophies (including dysferlinopathy) and the detection of new therapeutic targets for dysferlinopathy by means of experiments using animal models for dysferlinopathy.
Collapse
|
84
|
1α,25(OH)(2)-Vitamin D3 increases dysferlin expression in vitro and in a human clinical trial. Mol Ther 2012; 20:1988-97. [PMID: 22910291 DOI: 10.1038/mt.2012.156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dysferlinopathies are a heterogenous group of autosomal recessive inherited muscular dystrophies caused by mutations in DYSF gene. Dysferlin is expressed mainly in skeletal muscle and in monocytes and patients display a severe reduction or absence of protein in both tissues. Vitamin D3 promotes differentiation of the promyelocytic leukemia HL60 cells. We analyzed the effect of vitamin D3 on dysferlin expression in vitro using HL60 cells, monocytes and myotubes from controls and carriers of a single mutation in DYSF. We also performed an observational study with oral vitamin D3 in a cohort of 21 carriers. Fifteen subjects were treated for 1 year and dysferlin expression in monocytes was analysed before and after treatment. Treatment with vitamin D3 increased expression of dysferlin in vitro. The effect of vitamin D3 was mediated by both a nongenomic pathway through MEK/ERK and a genomic pathway involving binding of vitamin D3 receptor to the dysferlin promoter. Carriers treated with vitamin D3 had significantly increased expression of dysferlin in monocytes compared with nontreated carriers (P < 0.05). These findings will have important therapeutic implications since a combination of different molecular strategies together with vitamin D3 uptake could increase dysferlin expression to nonpathological protein levels.
Collapse
|
85
|
Roostalu U, Strähle U. In vivo imaging of molecular interactions at damaged sarcolemma. Dev Cell 2012; 22:515-29. [PMID: 22421042 DOI: 10.1016/j.devcel.2011.12.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 10/25/2011] [Accepted: 12/14/2011] [Indexed: 12/30/2022]
Abstract
Muscle cells have a remarkable capability to repair plasma membrane lesions. Mutations in dysferlin (dysf) are known to elicit a progressive myopathy in humans, probably due to impaired sarcolemmal repair. We show here that loss of Dysf and annexin A6 (Anxa6) function lead to myopathy in zebrafish. By use of high-resolution imaging of myofibers in intact animals, we reveal sequential phases in sarcolemmal repair. Initially, membrane vesicles enriched in Dysf together with cytoplasmic Anxa6 form a tight patch at the lesion independently of one another. In the subsequent steps, annexin A2a (Anxa2a) followed by annexin A1a (Anxa1a) accumulate at the patch; the recruitment of these annexins depends on Dysf and Anxa6. Thus, sarcolemmal repair relies on the ordered assembly of a protein-membrane scaffold. Moreover, we provide several lines of evidence that the membrane for sarcolemmal repair is derived from a specialized plasma membrane compartment.
Collapse
Affiliation(s)
- Urmas Roostalu
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology and University of Heidelberg, Eggenstein-Leopoldshafen, Germany.
| | | |
Collapse
|
86
|
Distinct effects of contraction-induced injury in vivo on four different murine models of dysferlinopathy. J Biomed Biotechnol 2012; 2012:134031. [PMID: 22431915 PMCID: PMC3303924 DOI: 10.1155/2012/134031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/27/2011] [Indexed: 11/17/2022] Open
Abstract
Mutations in the DYSF gene, encoding dysferlin, cause muscular dystrophies in man. We compared 4 dysferlinopathic mouse strains: SJL/J and B10.SJL-Dysfim/AwaJ (B10.SJL), and A/J and B6.A-Dysfprmd/GeneJ (B6.A/J). The former but not the latter two are overtly myopathic and weaker at 3 months of age. Following repetitive large-strain injury (LSI) caused by lengthening contractions, all except B6.A/J showed ~40% loss in contractile torque. Three days later, torque in SJL/J, B10.SJL and controls, but not A/J, recovered nearly completely. B6.A/J showed ~30% torque loss post-LSI and more variable recovery. Pre-injury, all dysferlinopathic strains had more centrally nucleated fibers (CNFs) and all but A/J showed more inflammation than controls. At D3, all dysferlinopathic strains showed increased necrosis and inflammation, but not more CNFs; controls were unchanged. Dystrophin-null DMDmdx mice showed more necrosis and inflammation than all dysferlin-nulls. Torque loss and inflammation on D3 across all strains were linearly related to necrosis. Our results suggest that (1) dysferlin is not required for functional recovery 3 days after LSI; (2) B6.A/J mice recover from LSI erratically; (3) SJL/J and B10.SJL muscles recover rapidly, perhaps due to ongoing myopathy; (4) although they recover function to different levels, all 4 dysferlinopathic strains show increased inflammation and necrosis 3 days after LSI.
Collapse
|
87
|
Lin P, Zhu H, Cai C, Wang X, Cao C, Xiao R, Pan Z, Weisleder N, Takeshima H, Ma J. Nonmuscle myosin IIA facilitates vesicle trafficking for MG53-mediated cell membrane repair. FASEB J 2012; 26:1875-83. [PMID: 22253476 DOI: 10.1096/fj.11-188599] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Repair of injury to the plasma membrane is an essential mechanism for maintenance of cellular homeostasis and integrity that involves coordinated movement of intracellular vesicles to membrane injury sites to facilitate patch formation. We have previously identified MG53 as an essential component of the cell membrane repair machinery. In order for MG53 and intracellular vesicles to translocate to membrane injury sites, motor proteins must be involved. Here, we show that nonmuscle myosin type IIA (NM-IIA) interacts with MG53 to regulate vesicle trafficking during cell membrane repair. In cells that are deficient for NM-IIA expression, MG53 cannot translocate to acute injury sites, whereas rescue of NM-IIA expression in these cells can restore MG53-mediated membrane repair. Compromised cell membrane repair is observed in cells with RNAi-mediated knockdown of NM-IIA expression, or following pharmacological alteration of NM-IIA motor function. Together, our data reveal NM-IIA as a key cytoskeleton motor protein that facilitates vesicle trafficking during MG53-mediated cell membrane repair.
Collapse
Affiliation(s)
- Peihui Lin
- Department of Physiology and Biophysics, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Roche JA, Ru LW, O'Neill AM, Resneck WG, Lovering RM, Bloch RJ. Unmasking potential intracellular roles for dysferlin through improved immunolabeling methods. J Histochem Cytochem 2011; 59:964-75. [PMID: 22043020 DOI: 10.1369/0022155411423274] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mutations in the DYSF gene that severely reduce the levels of the protein dysferlin are implicated in muscle-wasting syndromes known as dysferlinopathies. Although studies of its function in skeletal muscle have focused on its potential role in repairing the plasma membrane, dysferlin has also been found, albeit inconsistently, in the sarcoplasm of muscle fibers. The aim of this article is to study the localization of dysferlin in skeletal muscle through optimized immunolabeling methods. We studied the localization of dysferlin in control rat skeletal muscle using several different methods of tissue collection and subsequent immunolabeling. We then applied our optimized immunolabeling methods on human cadaveric muscle, control and dystrophic human muscle biopsies, and control and dysferlin-deficient mouse muscle. Our data suggest that dysferlin is present in a reticulum of the sarcoplasm, similar but not identical to those containing the dihydropyridine receptors and distinct from the distribution of the sarcolemmal protein dystrophin. Our data illustrate the importance of tissue fixation and antigen unmasking for proper immunolocalization of dysferlin. They suggest that dysferlin has an important function in the internal membrane systems of skeletal muscle, involved in calcium homeostasis and excitation-contraction coupling.
Collapse
Affiliation(s)
- Joseph A Roche
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | | | | | | | | | |
Collapse
|
89
|
Humphrey GW, Mekhedov E, Blank PS, de Morree A, Pekkurnaz G, Nagaraju K, Zimmerberg J. GREG cells, a dysferlin-deficient myogenic mouse cell line. Exp Cell Res 2011; 318:127-35. [PMID: 22020321 DOI: 10.1016/j.yexcr.2011.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 11/29/2022]
Abstract
The dysferlinopathies (e.g. LGMD2b, Myoshi myopathy) are progressive, adult-onset muscle wasting syndromes caused by mutations in the gene coding for dysferlin. Dysferlin is a large (~200kDa) membrane-anchored protein, required for maintenance of plasmalemmal integrity in muscle fibers. To facilitate analysis of dysferlin function in muscle cells, we have established a dysferlin-deficient myogenic cell line (GREG cells) from the A/J mouse, a genetic model for dysferlinopathy. GREG cells have no detectable dysferlin expression, but proliferate normally in growth medium and fuse into functional myotubes in differentiation medium. GREG myotubes exhibit deficiencies in plasma membrane repair, as measured by laser wounding in the presence of FM1-43 dye. Under the wounding conditions used, the majority (~66%) of GREG myotubes lack membrane repair capacity, while no membrane repair deficiency was observed in dysferlin-normal C2C12 myotubes, assayed under the same conditions. We discuss the possibility that the observed heterogeneity in membrane resealing represents genetic compensation for dysferlin deficiency.
Collapse
Affiliation(s)
- Glen W Humphrey
- Program in Physical Biology, Eunice Kennedy Schriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Rezvanpour A, Santamaria-Kisiel L, Shaw GS. The S100A10-annexin A2 complex provides a novel asymmetric platform for membrane repair. J Biol Chem 2011; 286:40174-83. [PMID: 21949189 PMCID: PMC3220529 DOI: 10.1074/jbc.m111.244038] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Membrane repair is mediated by multiprotein complexes, such as that formed between the dimeric EF-hand protein S100A10, the calcium- and phospholipid-binding protein annexin A2, the enlargeosome protein AHNAK, and members of the transmembrane ferlin family. Although interactions between these proteins have been shown, little is known about their structural arrangement and mechanisms of formation. In this work, we used a non-covalent complex between S100A10 and the N terminus of annexin A2 (residues 1-15) and a designed hybrid protein (A10A2), where S100A10 is linked in tandem to the N-terminal region of annexin A2, to explore the binding region, stoichiometry, and affinity with a synthetic peptide from the C terminus of AHNAK. Using multiple biophysical methods, we identified a novel asymmetric arrangement between a single AHNAK peptide and the A10A2 dimer. The AHNAK peptide was shown to require the annexin A2 N terminus, indicating that the AHNAK binding site comprises regions on both S100A10 and annexin proteins. NMR spectroscopy was used to show that the AHNAK binding surface comprised residues from helix IV in S100A10 and the C-terminal portion from the annexin A2 peptide. This novel surface maps to the exposed side of helices IV and IV' of the S100 dimeric structure, a region not identified in any previous S100 target protein structures. The results provide the first structural details of the ternary S100A10 protein complex required for membrane repair.
Collapse
Affiliation(s)
- Atoosa Rezvanpour
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | |
Collapse
|
91
|
Bouter A, Gounou C, Bérat R, Tan S, Gallois B, Granier T, d'Estaintot BL, Pöschl E, Brachvogel B, Brisson AR. Annexin-A5 assembled into two-dimensional arrays promotes cell membrane repair. Nat Commun 2011; 2:270. [PMID: 21468022 PMCID: PMC3104517 DOI: 10.1038/ncomms1270] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 03/09/2011] [Indexed: 11/18/2022] Open
Abstract
Eukaryotic cells possess a universal repair machinery that ensures rapid resealing of plasma membrane disruptions. Before resealing, the torn membrane is submitted to considerable tension, which functions to expand the disruption. Here we show that annexin-A5 (AnxA5), a protein that self-assembles into two-dimensional (2D) arrays on membranes upon Ca2+ activation, promotes membrane repair. Compared with wild-type mouse perivascular cells, AnxA5-null cells exhibit a severe membrane repair defect. Membrane repair in AnxA5-null cells is rescued by addition of AnxA5, which binds exclusively to disrupted membrane areas. In contrast, an AnxA5 mutant that lacks the ability of forming 2D arrays is unable to promote membrane repair. We propose that AnxA5 participates in a previously unrecognized step of the membrane repair process: triggered by the local influx of Ca2+, AnxA5 proteins bind to torn membrane edges and form a 2D array, which prevents wound expansion and promotes membrane resealing. Eukaryotic cell plasma membranes possess a mechanism to repair tears caused by stimuli such as mechanical stress. The authors demonstrate that annexin-A5, when assembled into two-dimensional arrays in the presence of calcium, is required for membrane repair.
Collapse
Affiliation(s)
- Anthony Bouter
- Molecular Imaging and NanoBioTechnology, IECB, UMR-5248 CBMN CNRS-University Bordeaux1-ENITAB, Talence F-33402, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Weisleder N, Takeshima H, Ma J. Mitsugumin 53 (MG53) facilitates vesicle trafficking in striated muscle to contribute to cell membrane repair. Commun Integr Biol 2011; 2:225-6. [PMID: 19641737 DOI: 10.4161/cib.2.3.8077] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Accepted: 02/02/2009] [Indexed: 11/19/2022] Open
Abstract
Repair of the plasma membrane following damage is an important aspect of normal cellular physiology, and disruption of this process is observed in many pathologic states. In a recent series of publications, we resolved that Mitsugumin 53 (MG53) is a novel, muscle-specific member of the tripartite motif/RING B-box Coiled Coil (TRIM/RBCC) family of proteins (TRIM72) that contributes to vesicle trafficking in the course of normal cellular physiology. MG53 can bind phosphatidylserine (PS) with some specificity, and interacts with caveolin-3 (Cav-3) as part of its function in vesicle trafficking. As part of the response to membrane damage in muscle cells, MG53 acts in an oxidation-dependent manner to facilitate vesicle translocation to the sites of membrane injury where these vesicles are involved in patching the membrane. Here we discuss these findings and examine the implications of this work in the field of membrane repair. Further discussion is provided about potential therapeutic applications for MG53.
Collapse
Affiliation(s)
- Noah Weisleder
- Department of Physiology and Biophysics; Robert Wood Johnson Medical School; Piscataway, NJ USA
| | | | | |
Collapse
|
93
|
Yu C, Sharma A, Trane A, Utokaparch S, Leung C, Bernatchez P. Myoferlin gene silencing decreases Tie-2 expression in vitro and angiogenesis in vivo. Vascul Pharmacol 2011; 55:26-33. [PMID: 21586340 DOI: 10.1016/j.vph.2011.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 04/13/2011] [Accepted: 04/18/2011] [Indexed: 01/09/2023]
Abstract
Angiogenesis consists in the growth of new blood vessels from pre-existing ones. Although anti-angiogenesis interventions have been shown to have therapeutic properties in human diseases such as cancer, their effect is only partial and the identification of novel modulators of angiogenesis is warranted. Recently, we reported the unexpected proteomic identification in endothelial cells (EC) of Myoferlin, a member of the Ferlin family of transmembrane proteins. Ferlins are well known to regulate the fusion of lipid vesicles at the plasma membrane in muscle cells, and we showed that Myoferlin gene knockdown not only decreases lipid vesicle fusion in EC but also attenuates Vascular Endothelial Growth Factor (VEGF) Receptor-2 (VEGFR-2) expression. Herein, we show that Myoferlin gene silencing in cultured EC also results in attenuated expression of a second tyrosine kinase receptor, Tie-2, which is another well-described angiogenic receptor. Most importantly, we provide evidence that delivery of a low-volume Myoferlin siRNA preparation in mouse tissues results in attenuated angiogenesis and edema formation. This provides the first evidence that acute Myoferlin knockdown has anti-angiogenic effects and validates Myoferlin as an anti-angiogenesis target. Furthermore, this supports the unexpected but increasingly accepted concept that proper tyrosine kinase receptors expression at the plasma membrane requires Myoferlin.
Collapse
Affiliation(s)
- Carol Yu
- The Providence Heart and Lung Institute, The James Hogg Research Centre, St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
94
|
Hwang M, Ko JK, Weisleder N, Takeshima H, Ma J. Redox-dependent oligomerization through a leucine zipper motif is essential for MG53-mediated cell membrane repair. Am J Physiol Cell Physiol 2011; 301:C106-14. [PMID: 21525429 DOI: 10.1152/ajpcell.00382.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We recently discovered that MG53, a muscle-specific tripartite motif (TRIM) family protein, functions as a sensor of oxidation to nucleate the assembly of cell membrane repair machinery. Our data showed that disulfide bond formation mediated by Cys242 is critical for MG53-mediated translocation of intracellular vesicles toward the injury sites. Here we test the hypothesis that leucine zipper motifs in the coiled-coil domain of MG53 constitute an additional mechanism that facilitates oligomerization of MG53 during cell membrane repair. Two leucine zipper motifs in the coiled-coil domain of MG53 (LZ1 - L176/L183/L190/V197 and LZ2 - L205/L212/L219/L226) are highly conserved across the different animal species. Chemical cross-linking studies show that LZ1 is critical for MG53 homodimerization, whereas LZ2 is not. Mutations of the conserved leucines into alanines in LZ1, not in LZ2, diminish the redox-dependent oligomerization of MG53. Live cell imaging studies demonstrate that the movement of green fluorescent protein (GFP)-tagged MG53 mutants (GFP-LA1 and GFP-LA2) is partially compromised in response to mechanical damage of the cell membrane, and the GFP-LA1/2 double mutant is completely ineffective in translocation toward the injury sites. In addition to the leucine zipper-mediated intermolecular interaction, redox-dependent cross talk between MG53 appears to be an obligatory step for cell membrane repair, since in vivo modification of cysteine residues with alkylating reagents can prevent the movement of MG53 toward the injury sites. Our data show that oxidation of the thiol group of Cys242 and leucine zipper-mediated interaction among the MG53 molecules both contribute to the nucleation process for MG53-mediated cell membrane repair.
Collapse
Affiliation(s)
- Moonsun Hwang
- Department of Physiology and Biophysics, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | | | |
Collapse
|
95
|
Kobayashi K, Izawa T, Kuwamura M, Yamate J. Comparative Gene Expression Analysis in the Skeletal Muscles of Dysferlin-deficient SJL/J and A/J Mice. J Toxicol Pathol 2011; 24:49-62. [PMID: 22272044 PMCID: PMC3234620 DOI: 10.1293/tox.24.49] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 12/01/2010] [Indexed: 11/23/2022] Open
Abstract
Quantitative real-time polymerase chain reaction (qRT-PCR) analysis was conducted to
determine whether or not there are interstrain or site-dependent differences in the gene
expression profiles of skeletal muscles in SJL/J and A/J mice as dysferlinopathy models.
Upon analysis by qRT-PCR, SJL/J mice showed a trend of increased gene expression level of
uncoupling protein 2 in the rectus femoris and longissimus lumborum at 30 weeks of age
when dystrophic lesions became histopathologically pronounced. Heme oxygenase 1 and S100
calcium binding protein A4 were upregulated in the rectus femoris, longissimus lumborum
and abdominal muscles, in which dystrophic lesions occur more commonly in SJL mice. The
gene expression levels of heat shock protein 70 in most muscles of A/J mice were lower
than those of BALB/c mice as control. SJL/J mice exhibited a marked lowering of
decay-accelerating factor 1/CD55 gene expression level in all studied muscles except for
the heart at all ages compared with that of BALB/c mice. This study showed that there were
some interstrain differences in the gene expres sion profiles of skeletal muscles between
SJL/J and A/J mice. Further investigation is required to reveal whether these alterations
of the expression levels are the cause of dystrophic changes or occur subsequent to muscle
damage.
Collapse
Affiliation(s)
- Kinji Kobayashi
- Laboratory of Veterinary Pathology, Division of Veterinary Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-ourai-kita, Izumisano City, Osaka 598-8531, Japan
| | | | | | | |
Collapse
|
96
|
Han R. Muscle membrane repair and inflammatory attack in dysferlinopathy. Skelet Muscle 2011; 1:10. [PMID: 21798087 PMCID: PMC3156633 DOI: 10.1186/2044-5040-1-10] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 03/01/2011] [Indexed: 12/17/2022] Open
Abstract
Repair of plasma membrane tears is an important normal physiological process that enables the cells to survive a variety of physiological and pathological membrane lesions. Dysferlin was the first protein reported to play a crucial role in this repair process in muscle, and recently, several other proteins including Mitsugumin 53 (MG53), annexin and calpain were also found to participate. These findings have now established the framework of the membrane repair mechanism. Defective membrane repair in dysferlin-deficient muscle leads to the development of muscular dystrophy associated with remarkable muscle inflammation. Recent studies have demonstrated a crosstalk between defective membrane repair and immunological attack, thus unveiling a new pathophysiological mechanism of dysferlinopathy. Here I summarize and discuss the latest progress in the molecular mechanisms of membrane repair and the pathogenesis of dysferlinopathy. Discussion about potential therapeutic applications of these findings is also provided.
Collapse
Affiliation(s)
- Renzhi Han
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA.
| |
Collapse
|
97
|
Zhu H, Lin P, De G, Choi KH, Takeshima H, Weisleder N, Ma J. Polymerase transcriptase release factor (PTRF) anchors MG53 protein to cell injury site for initiation of membrane repair. J Biol Chem 2011; 286:12820-4. [PMID: 21343302 DOI: 10.1074/jbc.c111.221440] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Plasma membrane repair is an essential process for maintenance of homeostasis at the cellular and tissue levels, whereas compromised repair capacity contributes to degenerative human diseases. Our recent studies show that MG53 is essential for muscle membrane repair, and defects in MG53 function are linked to muscular dystrophy and cardiac dysfunction. Here we report that polymerase I and transcript release factor (PTRF), a gene known to regulate caveolae membrane structure, is an indispensable component of the membrane repair machinery. PTRF acts as a docking protein for MG53 during membrane repair potentially by binding exposed membrane cholesterol at the injury site. Cells lacking expression of endogenous PTRF show defective trafficking of MG53 to membrane injury sites. A mutation in PTRF associated with human disease results in aberrant nuclear localization of PTRF and disrupts MG53 function in membrane resealing. Although RNAi silencing of PTRF leads to defective muscle membrane repair, overexpression of PTRF can rescue membrane repair defects in dystrophic muscle. Our data suggest that membrane-delimited interaction between MG53 and PTRF contributes to initiation of cell membrane repair, which can be an attractive target for treatment or prevention of tissue injury in human diseases.
Collapse
Affiliation(s)
- Hua Zhu
- Department of Physiology and Biophysics, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
98
|
Swildens J, de Vries AAF, Li Z, Umar S, Atsma DE, Schalij MJ, van der Laarse A. Integrin stimulation favors uptake of macromolecules by cardiomyocytes in vitro. Cell Physiol Biochem 2011; 26:999-1010. [PMID: 21220931 DOI: 10.1159/000324013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2010] [Indexed: 11/19/2022] Open
Abstract
Previously, our research group showed that integrin stimulation induces release of cardiac troponin I from viable neonatal rat ventricular cardiomyocytes (NRCMs), but would it also stimulate uptake of exogenous macromolecules? For this purpose, beating NRCMs were incubated without or with an RGD motif-containing peptide (GRGDS) to stimulate integrins in the presence of Texas Red-conjugated ovalbumin (OTR; 45 kDa) or dextran (DTR; 70 kDa). After incubation periods of 8, 16 and 24 hours endocytosis of red label was quantified by fluorescence microscopy. Uptake of OTR and DTR by NRCMs was intensified by GRGDS treatment (p for trend <0.001 and 0.019, respectively) and increased with duration of incubation (p<0.001 for both). The GRGDS-induced uptake of OTR by NRCMs correlated positively with OTR concentration (p<0.001). Experiments with pharmacological inhibitors of endocytosis indicated that in the absence of GRGDS, NRCMs take up OTR by the clathrin-mediated pathway of endocytosis while the GRGDS-dependent OTR uptake occurs by macropinocytosis. Cultures of NRCMs that were stretched cyclically showed ≍4-fold increased uptake of OTR compared to stationary NRCM cultures. Immunofluorescence microscopy revealed that the dysferlin-positive plasma membrane (PM) areas in beating GRGDS-treated NRCMs were ≍3-fold larger than in contracting NRCMs incubated with vehicle (p<0.001). However, in non-beating NRCMs exposure to GRGDS did not induce larger dysferlin-positive PM areas, nor did it stimulate uptake of OTR. After inhibition of dysferlin expression by short hairpin RNA-mediated RNA interference, OTR uptake by contracting NRCMs could no longer be stimulated via GRGDS treatment. We conclude that in NRCMs, stimulation of integrins by RGD motif-containing peptides or stretch cause uptake of labeled macromolecules. The latter process appears to depend on the contractile behavior of the NRCMs and on the PM repair protein dysferlin, probably because of its role in macropinocytosis.
Collapse
Affiliation(s)
- Jim Swildens
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
Dysferlin is a sarcolemmal protein that plays an important role in patching defects in skeletal membrane by regulating vesicle fusion with the sarcolemma. Mutations in the dysferlin gene can lead to a variety of clinical phenotypes. Affected individuals usually present with early involvement of the posterior calf muscles (Miyoshi myopathy) in their teens or early twenties, but can present with proximal greater than distal weakness similar to other limb-girdle muscular dystrophies (LGMD2B), with anterior tibial weakness, an axial myopathy (e.g., rigid spine syndrome or hyperkyphosis resembling bent spine syndrome), or any combination of the above. Muscle biopsies may be quite inflammatory, often resulting in a misdiagnosis as polymyositis. Unfortunately, there are no medical therapies available at this time.
Collapse
|
100
|
Rayavarapu S, Van der meulen JH, Gordish-Dressman H, Hoffman EP, Nagaraju K, Knoblach SM. Characterization of dysferlin deficient SJL/J mice to assess preclinical drug efficacy: fasudil exacerbates muscle disease phenotype. PLoS One 2010; 5:e12981. [PMID: 20886045 PMCID: PMC2945315 DOI: 10.1371/journal.pone.0012981] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 08/31/2010] [Indexed: 12/21/2022] Open
Abstract
The dysferlin deficient SJL/J mouse strain is commonly used to study dysferlin deficient myopathies. Therefore, we systematically evaluated behavior in relatively young (9-25 weeks) SJL/J mice and compared them to C57BL6 mice to determine which functional end points may be the most effective to use for preclinical studies in the SJL/J strain. SJL/J mice had reduced body weight, lower open field scores, higher creatine kinase levels, and less muscle force than did C57BL6 mice. Power calculations for expected effect sizes indicated that grip strength normalized to body weight and open field activity were the most sensitive indicators of functional status in SJL/J mice. Weight and open field scores of SJL/J mice deteriorated over the course of the study, indicating that progressive myopathy was ongoing even in relatively young (<6 months old) SJL/J mice. To further characterize SJL/J mice within the context of treatment, we assessed the effect of fasudil, a rho-kinase inhibitor, on disease phenotype. Fasudil was evaluated based on previous observations that Rho signaling may be overly activated as part of the inflammatory cascade in SJL/J mice. Fasudil treated SJL/J mice showed increased body weight, but decreased grip strength, horizontal activity, and soleus muscle force, compared to untreated SJL/J controls. Fasudil either improved or had no effect on these outcomes in C57BL6 mice. Fasudil also reduced the number of infiltrating macrophages/monocytes in SJL/J muscle tissue, but had no effect on muscle fiber degeneration/regeneration. These studies provide a basis for standardization of preclinical drug testing trials in the dysferlin deficient SJL/J mice, and identify measures of functional status that are potentially translatable to clinical trial outcomes. In addition, the data provide pharmacological evidence suggesting that activation of rho-kinase, at least in part, may represent a beneficial compensatory response in dysferlin deficient myopathies.
Collapse
Affiliation(s)
- Sree Rayavarapu
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Institute of Biomedical Sciences, The George Washington University, Washington, D.C., United States of America
| | - Jack H. Van der meulen
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
| | - Heather Gordish-Dressman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
| | - Eric P. Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, School of Medicine and Health Sciences, The George Washington University, Washington, D.C., United States of America
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, School of Medicine and Health Sciences, The George Washington University, Washington, D.C., United States of America
- * E-mail: (KN); (SMK)
| | - Susan M. Knoblach
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, D.C., United States of America
- Department of Integrative Systems Biology, School of Medicine and Health Sciences, The George Washington University, Washington, D.C., United States of America
- * E-mail: (KN); (SMK)
| |
Collapse
|