Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Hum Mutat ; 41(9): 1507-1513, 2020 09.
Article in English | MEDLINE | ID: mdl-32557990

ABSTRACT

Recessively inherited limb girdle muscular dystrophy (LGMD) type 2A is the most common LGMD worldwide. Here, we report the first single missense variant in CAPN3 causing dominantly inherited calpainopathy. A 43-year-old proband, his father and two sons were heterozygous for a c.1715G>C p.(Arg572Pro) variant in CAPN3. Affected family members had at least three of the following; muscle pain, a LGMD2A pattern of muscle weakness and wasting, muscle fat replacement on magnetic resonance imaging, myopathic muscle biopsy, and elevated creatine kinase. Total calpain 3 protein expression was 4 ± 3% of normal. In vitro analysis of c.1715G>C and the previously described c.643_663del variant indicated that the mutant proteins lack autolytic and proteolytic activity and decrease the quantity of wild-type CAPN3 protein. Our findings suggest that dominantly inherited calpainopathy is not unique to the previously reported c.643_663del mutation of CAPN3, and that dominantly inherited calpainopathy should be considered for other single variations in CAPN3.


Subject(s)
Calpain/genetics , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Adolescent , Adult , Aged , Child , Humans , Male , Middle Aged , Pedigree , Young Adult
2.
Sci Transl Med ; 11(520)2019 11 27.
Article in English | MEDLINE | ID: mdl-31776291

ABSTRACT

Limb-girdle muscular dystrophy type 2A (LGMD2A or LGMDR1) is a neuromuscular disorder caused by mutations in the calpain 3 gene (CAPN3). Previous experiments using adeno-associated viral (AAV) vector-mediated calpain 3 gene transfer in mice indicated cardiac toxicity associated with the ectopic expression of the calpain 3 transgene. Here, we performed a preliminary dose study in a severe double-knockout mouse model deficient in calpain 3 and dysferlin. We evaluated safety and biodistribution of AAV9-desmin-hCAPN3 vector administration to nonhuman primates (NHPs) with a dose of 3 × 1013 viral genomes/kg. Vector administration did not lead to observable adverse effects or to detectable toxicity in NHP. Of note, the transgene expression did not produce any abnormal changes in cardiac morphology or function of injected animals while reaching therapeutic expression in skeletal muscle. Additional investigation on the underlying causes of cardiac toxicity observed after gene transfer in mice and the role of titin in this phenomenon suggest species-specific titin splicing. Mice have a reduced capacity for buffering calpain 3 activity compared to NHPs and humans. Our studies highlight a complex interplay between calpain 3 and titin binding sites and demonstrate an effective and safe profile for systemic calpain 3 vector delivery in NHP, providing critical support for the clinical potential of calpain 3 gene therapy in humans.


Subject(s)
Calpain/genetics , Calpain/therapeutic use , Cardiotoxicity/etiology , Connectin/genetics , Genetic Therapy/adverse effects , Muscle Proteins/genetics , Muscle Proteins/therapeutic use , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/therapy , RNA Splicing/genetics , Animals , Binding Sites , Biomarkers/blood , Cardiotoxicity/blood , Connectin/chemistry , Dependovirus/genetics , Dysferlin/deficiency , Dysferlin/metabolism , Enzyme Stability , Gene Expression Regulation , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/blood , Muscular Dystrophies, Limb-Girdle/pathology , Myocardium/metabolism , Myocardium/pathology , Primates , Protein Domains , Proteolysis , Species Specificity , Tissue Distribution , Transgenes
3.
Muscle Nerve ; 56(1): 129-135, 2017 07.
Article in English | MEDLINE | ID: mdl-27759885

ABSTRACT

INTRODUCTION: We report the genetic analysis of a large series of 76 Algerian patients from 65 unrelated families who presented with early onset severe muscular dystrophy and a clinical phenotype resembling limb-girdle muscular dystrophy type 2C. METHODS: To define the genetic basis of the diseases in these families, we undertook a series of analyses of the γ-sarcoglycan (SGCG) and DMD genes. RESULTS: Fifteen families were shown to carry SGCG variants. Only 2 kinds of causative mutations were identified in the population, mostly in the homozygous state: the well-known c.525delT and the previously described c.87dupT. In the DMD gene, 12 distinctive patterns of deletion were identified, mostly affecting the dystrophin central region. CONCLUSIONS: Our data suggest that a simple molecular screen consisting of 2 allele-specific polymerase chain reactions (PCRs) and a set of 3 multiplex PCRs can diagnose half of the patients who present with progressive muscular dystrophy in the developing nation of Algeria. Muscle Nerve 56: 129-135, 2017.


Subject(s)
Dystrophin/genetics , Muscular Dystrophies/genetics , Mutation/genetics , Sarcoglycanopathies/genetics , Sarcoglycans/genetics , Adolescent , Adult , Algeria , Child , Child, Preschool , Cohort Studies , Family Health , Female , Genetic Testing , Humans , Male , Statistics, Nonparametric , Young Adult
4.
Neurology ; 85(24): 2126-35, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26581302

ABSTRACT

OBJECTIVE: To identify the genetic defects present in 3 families with muscular dystrophy, contractures, and calpain 3 deficiency. METHODS: We performed targeted exome sequencing on one patient presenting a deficiency in calpain 3 on Western blot but for which mutations in the gene had been excluded. The identification of a homozygous truncating mutation in the M-line part of titin prompted us to sequence this region in 2 additional patients presenting similar clinical and biochemical characteristics. RESULTS: The 3 patients shared similar features: coexistence of limb-girdle weakness and early-onset diffuse joint contractures without cardiomyopathy. The biopsies showed rimmed vacuoles, a dystrophic pattern, and secondary reduction in calpain 3. We identified a novel homozygous mutation in the exon Mex3 of the TTN gene in the first patient. At protein level, this mutation introduces a stop codon at the level of Mex3. Interestingly, we identified truncating mutations in both alleles in the same region of the TTN gene in patients from 2 additional families. Molecular protein analyses confirm loss of the C-ter part of titin. CONCLUSIONS: Our study broadens the phenotype of titinopathies with the report of a new clinical entity with prominent contractures and no cardiac abnormality and where the recessive mutations lead to truncation of the M-line titin and secondary calpain 3 deficiency.


Subject(s)
Cardiomyopathies , Connectin/genetics , Frameshift Mutation/genetics , Muscular Dystrophy, Emery-Dreifuss/diagnosis , Muscular Dystrophy, Emery-Dreifuss/genetics , Phenotype , Female , Humans , Male , Middle Aged , Young Adult
5.
Mol Ther Methods Clin Dev ; 2: 15009, 2015.
Article in English | MEDLINE | ID: mdl-26029720

ABSTRACT

Recombinant adeno-associated virus (rAAV) is currently the best vector for gene delivery into the skeletal muscle. However, the 5-kb packaging size of this virus is a major obstacle for large gene transfer. This past decade, many different strategies were developed to circumvent this issue (concatemerization-splicing, overlapping vectors, hybrid dual or fragmented AAV). Loss of function mutations in the DYSF gene whose coding sequence is 6.2kb lead to progressive muscular dystrophies (LGMD2B: OMIM_253601; MM: OMIM_254130; DMAT: OMIM_606768). In this study, we compared large gene transfer techniques to deliver the DYSF gene into the skeletal muscle. After rAAV8s intramuscular injection into dysferlin deficient mice, we showed that the overlap strategy is the most effective approach to reconstitute a full-length messenger. After systemic administration, the level of dysferlin obtained on different muscles corresponded to 0.5- to 2-fold compared to the normal level. We further demonstrated that the overlapping vector set was efficient to correct the histopathology, resistance to eccentric contractions and whole body force in the dysferlin deficient mice. Altogether, these data indicate that using overlapping vectors could be a promising approach for a potential clinical treatment of dysferlinopathies.

6.
Eur J Hum Genet ; 23(7): 929-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25351777

ABSTRACT

Defects in TRIM32 were reported in limb-girdle muscular dystrophy type 2H (LGMD2H), sarcotubular myopathies (STM) and in Bardet-Biedl syndrome. Few cases have been described to date in LGMD2H/STM, but this gene is not systematically analysed because of the absence of specific signs and difficulties in protein analysis. By using high-throughput variants screening techniques, we identified variants in TRIM32 in two patients presenting nonspecific LGMD. We report the first case of total inactivation by homozygous deletion of the entire TRIM32 gene. Of interest, the deletion removes part of the ASTN2 gene, a large gene in which TRIM32 is nested. Despite the total TRIM32 gene inactivation, the patient does not present a more severe phenotype. However, he developed a mild progressive cognitive impairment that may be related to the loss of function of ASTN2 because association between ASTN2 heterozygous deletions and neurobehavioral disorders was previously reported. Regarding genomic characteristics at breakpoint of the deleted regions of TRIM32, we found a high density of repeated elements, suggesting a possible hotspot. These observations illustrate the importance of high-throughput technologies for identifying molecular defects in LGMD, confirm that total loss of function of TRIM32 is not associated with a specific phenotype and that TRIM32/ASTN2 inactivation could be associated with cognitive impairment.


Subject(s)
Comparative Genomic Hybridization/methods , Gene Deletion , High-Throughput Nucleotide Sequencing/methods , Muscular Dystrophies, Limb-Girdle/genetics , Transcription Factors/genetics , Adult , Base Sequence , DNA Mutational Analysis/methods , Family Health , Female , Humans , Male , Muscular Dystrophies, Limb-Girdle/pathology , Pedigree , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
7.
Mol Ther ; 22(6): 1176-1187, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24618805

ABSTRACT

Muscular dystrophies are a group of genetically distinct diseases for which no treatment exists. While gene transfer approach is being tested for several of these diseases, such strategies can be hampered when the size of the corresponding complementary DNA (cDNA) exceeds the packaging capacity of adeno-associated virus vectors. This issue concerns, in particular, dysferlinopathies and titinopathies that are due to mutations in the dysferlin (DYSF) and titin (TTN) genes. We investigated the efficacy of RNA trans-splicing as a mode of RNA therapy for these two types of diseases. Results obtained with RNA trans-splicing molecules designed to target the 3' end of mouse titin and human dysferlin pre-mRNA transcripts indicated that trans-splicing of pre-mRNA generated from minigene constructs or from the endogenous genes was achieved. Collectively, these results provide the first demonstration of DYSF and TTN trans-splicing reprogramming in vitro and in vivo. However, in addition to these positive results, we uncovered a possible issue of the technique in the form of undesirable translation of RNA pre-trans-splicing molecules, directly from open reading frames present on the molecule or associated with internal alternative cis-splicing. These events may hamper the efficiency of the trans-splicing process and/or lead to toxicity.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscular Dystrophies/therapy , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Precursors/genetics , RNA, Messenger/metabolism , Alternative Splicing , Animals , Cell Line , Dysferlin , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Muscular Dystrophies/genetics , Open Reading Frames , Trans-Splicing
8.
Circulation ; 128(10): 1094-104, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23908349

ABSTRACT

BACKGROUND: Genetic defects in calpain3 (CAPN3) lead to limb-girdle muscular dystrophy type 2A, a disease of the skeletal muscle that affects predominantly the proximal limb muscles. We previously demonstrated the potential of adeno-associated virus-mediated transfer of the CAPN3 gene to correct the pathological signs in a murine model for limb-girdle muscular dystrophy type 2A after intramuscular and locoregional administrations. METHODS AND RESULTS: Here, we showed that intravenous injection of calpain3-expressing vector in mice can induce mortality in a dose-dependent manner. An anatomopathological investigation revealed large areas of fibrosis in the heart that we related to unregulated proteolytic activity of calpain3. To circumvent this toxicity, we developed new adeno-associated virus vectors with skeletal muscle-restricted expression by using new muscle-specific promoters that include the CAPN3 promoter itself and by introducing a target sequence of the cardiac-specific microRNA-208a in the cassette. Our results show that CAPN3 transgene expression can be successfully suppressed in the cardiac tissue, preventing the cardiac toxicity, whereas expression of the transgene in skeletal muscle reverts the pathological signs of calpain3 deficiency. CONCLUSIONS: The molecular strategies used in this study may be useful for any gene transfer strategy with potential toxicity in the heart.


Subject(s)
Calpain/antagonists & inhibitors , Gene Expression Regulation, Enzymologic , Muscle Proteins/antagonists & inhibitors , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/enzymology , Muscular Dystrophies, Limb-Girdle/pathology , Animals , Calpain/biosynthesis , Calpain/genetics , Gene Expression Regulation, Enzymologic/physiology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HEK293 Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics
9.
Hum Gene Ther Clin Dev ; 24(2): 65-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23721401

ABSTRACT

Mutations in dysferlin and anoctamin 5 are the cause of muscular disorders, with the main presentations as limb-girdle muscular dystrophy or Miyoshi type of distal myopathy. Both these proteins have been implicated in sarcolemmal resealing. On the basis of similarities in associated phenotypes and protein functions, we tested the hypothesis that ANO5 protein could compensate for dysferlin absence. We first defined that the main transcript of ANO5 expressed in skeletal muscle is the 22-exon full-length isoform, and we demonstrated that dysferlin-deficient (Dysf (prmd)) mice have lower Ano5 expression levels, an observation that further enhanced the rational of the tested hypothesis. We then showed that AAV-mediated transfer of human ANO5 (hANO5) did not lead to apparent toxicity in wild-type mice. Finally, we demonstrated that AAV-hANO5 injection was not able to compensate for dysferlin deficiency in the Dysf (prmd) mouse model or improve the membrane repair defect seen in the absence of dysferlin. Consequently, overexpressing hANO5 does not seem to provide a valuable therapeutic strategy for dysferlin deficiency.


Subject(s)
Chloride Channels/metabolism , Dependovirus/genetics , Membrane Proteins/metabolism , Muscular Dystrophies, Limb-Girdle/therapy , Animals , Anoctamins , Cell Line , Chloride Channels/genetics , Down-Regulation , Dysferlin , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism
10.
PLoS One ; 7(5): e38036, 2012.
Article in English | MEDLINE | ID: mdl-22666441

ABSTRACT

Mutations in the dysferlin gene are the cause of Limb-girdle Muscular Dystrophy type 2B and Miyoshi Myopathy. The dysferlin protein has been implicated in sarcolemmal resealing, leading to the idea that the pathophysiology of dysferlin deficiencies is due to a deficit in membrane repair. Here, we show using two different approaches that fulfilling membrane repair as asseyed by laser wounding assay is not sufficient for alleviating the dysferlin deficient pathology. First, we generated a transgenic mouse overexpressing myoferlin to test the hypothesis that myoferlin, which is homologous to dysferlin, can compensate for the absence of dysferlin. The myoferlin overexpressors show no skeletal muscle abnormalities, and crossing them with a dysferlin-deficient model rescues the membrane fusion defect present in dysferlin-deficient mice in vitro. However, myoferlin overexpression does not correct muscle histology in vivo. Second, we report that AAV-mediated transfer of a minidysferlin, previously shown to correct the membrane repair deficit in vitro, also fails to improve muscle histology. Furthermore, neither myoferlin nor the minidysferlin prevented myofiber degeneration following eccentric exercise. Our data suggest that the pathogenicity of dysferlin deficiency is not solely related to impairment in sarcolemmal repair and highlight the care needed in selecting assays to assess potential therapies for dysferlinopathies.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/pathology , Genetic Therapy/methods , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Animals , Bystander Effect/genetics , Dependovirus/genetics , Dysferlin , Female , Gene Deletion , Gene Expression Regulation/genetics , Humans , Male , Membrane Fusion/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Muscle Proteins/genetics , Muscles/metabolism , Muscles/pathology , Muscles/physiopathology , Muscular Dystrophies, Limb-Girdle/therapy , Phenotype , Sarcolemma/metabolism , Sarcolemma/pathology , Treatment Outcome
11.
Hum Mol Genet ; 19(23): 4608-24, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20855473

ABSTRACT

The dominant tibial muscular dystrophy (TMD) and recessive limb-girdle muscular dystrophy 2J are allelic disorders caused by mutations in the C-terminus of titin, a giant sarcomeric protein. Both clinical presentations were initially identified in a large Finnish family and linked to a founder mutation (FINmaj). To further understand the physiopathology of these two diseases, we generated a mouse model carrying the FINmaj mutation. In heterozygous mice, dystrophic myopathology appears late at 9 months of age in few distal muscles. In homozygous (HO) mice, the first signs appear in the Soleus at 1 month of age and extend to most muscles at 6 months of age. Interestingly, the heart is also severely affected in HO mice. The mutation leads to the loss of the very C-terminal end of titin and to a secondary deficiency of calpain 3, a partner of titin. By crossing the FINmaj model with a calpain 3-deficient model, the TMD phenotype was corrected, demonstrating a participation of calpain 3 in the pathogenesis of this disease.


Subject(s)
Calpain/metabolism , Disease Models, Animal , Distal Myopathies , Muscle Proteins/metabolism , Muscular Dystrophies, Limb-Girdle , Animals , Blotting, Western , Calpain/deficiency , Calpain/genetics , Connectin , DNA Mutational Analysis , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Echocardiography , Genetic Linkage , Genetic Predisposition to Disease , Heterozygote , Mice , Microscopy, Electron , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Mutation , Polymerase Chain Reaction , Protein Kinases/genetics , Sarcomeres/genetics , Sarcomeres/ultrastructure
12.
FEBS J ; 277(20): 4322-37, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20860623

ABSTRACT

A multiprotein complex encompassing a transcription regulator, cardiac ankyrin repeat protein (CARP), and the calpain 3 protease was identified in the N2A elastic region of the giant sarcomeric protein titin. The present study aimed to investigate the function(s) of this complex in the skeletal muscle. We demonstrate that CARP subcellular localization is controlled by the activity of calpain 3: the higher the calpain 3, the more important the sarcomeric retention of CARP. This regulation would occur through cleavage of the N-terminal end of CARP by the protease. We show that, upon CARP over-expression, the transcription factor nuclear factor NF-κB p65 DNA-binding activity decreases. Taken as a whole, CARP and its regulator calpain 3 appear to occupy a central position in the important cell fate-governing NF-κB pathway. Interestingly, the expression of the atrophying protein MURF1, one of NF-κB main targets, remains unchanged in presence of CARP, suggesting that the pathway encompassing calpain 3/CARP/NF-κB does not play a role in muscle atrophy. With NF-κB also having anti-apoptotic effects, the inability of calpain 3 to lower CARP-driven inhibition of NF-κB could reduce muscle cell survival, hence partly accounting for the dystrophic pattern observed in limb girdle muscular dystrophy 2A, a pathology resulting from the protease deficiency.


Subject(s)
Calpain/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Humans , Muscular Dystrophies, Limb-Girdle/pathology , NF-kappa B/antagonists & inhibitors
13.
Hum Mol Genet ; 19(10): 1897-907, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20154340

ABSTRACT

Deficiency of the dysferlin protein presents as two major clinical phenotypes: limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. Dysferlin is known to participate in membrane repair, providing a potential hypothesis to the underlying pathophysiology of these diseases. The size of the dysferlin cDNA prevents its direct incorporation into an adeno-associated virus (AAV) vector for therapeutic gene transfer into muscle. To bypass this limitation, we split the dysferlin cDNA at the exon 28/29 junction and cloned it into two independent AAV vectors carrying the appropriate splicing sequences. Intramuscular injection of the corresponding vectors into a dysferlin-deficient mouse model led to the expression of full-length dysferlin for at least 1 year. Importantly, systemic injection in the tail vein of the two vectors led to a widespread although weak expression of the full-length protein. Injections were associated with an improvement of the histological aspect of the muscle, a reduction in the number of necrotic fibers, restoration of membrane repair capacity and a global improvement in locomotor activity. Altogether, these data support the use of such a strategy for the treatment of dysferlin deficiency.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Membrane Proteins/deficiency , Membrane Proteins/therapeutic use , Muscle Proteins/deficiency , Muscle Proteins/therapeutic use , Muscular Dystrophies, Limb-Girdle/genetics , Animals , Crosses, Genetic , Dysferlin , Female , Injections, Intramuscular , Male , Membrane Proteins/genetics , Membranes/pathology , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/therapy , Mutation , Phenotype , Transgenes , Wound Healing
14.
FEBS J ; 276(3): 669-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19143834

ABSTRACT

In an attempt to identify potential therapeutic targets for the correction of muscle wasting, the gene expression of several pivotal proteins involved in protein metabolism was investigated in experimental atrophy induced by transient or definitive denervation, as well as in four animal models of muscular dystrophies (deficient for calpain 3, dysferlin, alpha-sarcoglycan and dystrophin, respectively). The results showed that: (a) the components of the ubiquitin-proteasome pathway are upregulated during the very early phases of atrophy but do not greatly increase in the muscular dystrophy models; (b) forkhead box protein O1 mRNA expression is augmented in the muscles of a limb girdle muscular dystrophy 2A murine model; and (c) the expression of cardiac ankyrin repeat protein (CARP), a regulator of transcription factors, appears to be persistently upregulated in every condition, suggesting that CARP could be a hub protein participating in common pathological molecular pathway(s). Interestingly, the mRNA level of a cell cycle inhibitor known to be upregulated by CARP in other tissues, p21(WAF1/CIP1), is consistently increased whenever CARP is upregulated. CARP overexpression in muscle fibres fails to affect their calibre, indicating that CARP per se cannot initiate atrophy. However, a switch towards fast-twitch fibres is observed, suggesting that CARP plays a role in skeletal muscle plasticity. The observation that p21(WAF1/CIP1) is upregulated, put in perspective with the effects of CARP on the fibre type, fits well with the idea that the mechanisms at stake might be required to oppose muscle remodelling in skeletal muscle.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Biomarkers/metabolism , Calpain/deficiency , Calpain/genetics , Calpain/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Male , Mice , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/genetics , Muscular Dystrophies/genetics , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/genetics , Signal Transduction , Up-Regulation
15.
Neuromuscul Disord ; 17(2): 148-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17236769

ABSTRACT

Limb-girdle muscular dystrophy type 2A (LGMD2A) is an autosomal recessive muscular disorder caused by mutations in the gene coding for calpain 3, a calcium-dependent protease. We developed an in vitro assay that can detect the proteolytic activity of calpain 3 in a muscle sample. This assay is based on the use of an inactive calpain 3 as a substrate for active calpain 3 molecules. A total of 79 human biopsies have been analysed using an unbiased single blind method. Results were confronted with the molecular diagnosis for confirmation. Proteolytic activity was either reduced or absent in 68% of LGMD2A biopsies. In the remaining 32%, normal proteolytic activity was found despite the presence of calpain 3 mutation(s), suggesting that other calpain 3 properties might be impaired to give rise to the LGMD2A phenotype. Our assay is easily adaptable to routine and appears to be more sensitive than common analysis by immunodetection.


Subject(s)
Calpain/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscular Dystrophies, Limb-Girdle/enzymology , Animals , Blotting, Western , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Heterozygote , Humans , Mice , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , NIH 3T3 Cells , Phenotype , Reproducibility of Results , Tissue Banks , Transfection
16.
Mol Ther ; 13(2): 250-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16290124

ABSTRACT

Calpainopathy (limb-girdle muscular dystrophy type 2A, LGMD2A) is a recessive muscular disorder caused by deficiency in the calcium-dependent cysteine protease calpain 3. To date, no treatment exists for this disease. We evaluated the potential of recombinant adeno-associated virus (rAAV) vectors for gene therapy in a murine model for LGMD2A. To drive the expression of calpain 3, we used rAAV2/1 pseudotyped vectors and muscle-specific promoters to avoid calpain 3 cell toxicity. We report efficient and stable transgene expression in muscle with restoration of the proteolytic activity and without evident toxicity. In addition, calpain 3 was correctly targeted to the sarcomere. Moreover, its presence resulted in improvement of the histological features and in therapeutic efficacy at the physiological levels, including correction of atrophy and full rescue of the contractile force deficits. Our results establish the feasibility of AAV-mediated calpain 3 gene transfer as a therapeutic approach.


Subject(s)
Calpain/genetics , Calpain/therapeutic use , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Muscle Proteins/genetics , Muscle Proteins/therapeutic use , Muscular Dystrophies, Limb-Girdle/therapy , Animals , Calpain/biosynthesis , Calpain/deficiency , Disease Models, Animal , Enzyme Activation/genetics , Enzyme Stability/genetics , Genetic Vectors/therapeutic use , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/biosynthesis , Muscle Proteins/deficiency , Muscle, Skeletal/enzymology , Muscular Dystrophies, Limb-Girdle/metabolism
17.
Genomics ; 84(3): 592-5, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15498466

ABSTRACT

A calpain 3 (Capn3) deficiency model was created by targeted disruption of the mouse Capn3 gene through homologous recombination in ES cells. Analysis of the genotype of pups from heterozygous crosses revealed a transmission ratio distortion (TRD) in favor of homozygous Capn3-deficient mice. This TRD was not observed in a second model of Capn3 deficiency, ruling out a possible involvement of Capn3 deficiency in this phenotype. The molecular nature of the TRD was investigated by quantitative RT-PCR and RACE-PCR analyses. We observed the presence in testis and ovaries of abundant, novel transcripts of the Capn3 gene arising from the antisense strand of the Pgk1-neomycin cassette. Although we could not detect corresponding translation products, our results suggest that the activity of the Pgk1 promoter could be the causative factor of TRD. This first example of TRD induced by an introduced cassette further emphasizes the care that should be taken in interpreting phenotypes of animal models, especially when dealing with reproductive functions, and further supports the rationale of using excisable cassettes in inactivation strategies.


Subject(s)
Calpain/genetics , Chromosome Segregation/genetics , Gene Expression , Mice/genetics , Muscle Proteins/genetics , Transcription, Genetic/genetics , Animals , Blotting, Western , Genotype , Gonads/metabolism , Inheritance Patterns/genetics , Mutagenesis, Insertional , Phosphoglycerate Kinase/genetics , Polymerase Chain Reaction/methods , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Cell Biol ; 23(24): 9127-35, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645524

ABSTRACT

Calpain 3 (Capn3) is known as the skeletal muscle-specific member of the calpains, a family of intracellular nonlysosomal cysteine proteases. This enigmatic protease has many unique features among the calpain family and, importantly, mutations in Capn3 have been shown to be responsible for limb girdle muscular dystrophy type 2A. Here we demonstrate that the Capn3 activation mechanism is similar to the universal activation of caspases and corresponds to an autolysis within the active site of the protease. We undertook a search for substrates in immature muscle cells, as several lines of evidence suggest that Capn3 is mostly in an inactive state in muscle and needs a signal to be activated. In this model, Capn3 proteolytic activity leads to disruption of the actin cytoskeleton and disorganization of focal adhesions through cleavage of several endogenous proteins. In addition, we show that titin, a previously identified Capn3 partner, and filamin C are further substrates of Capn3. Finally, we report that Capn3 colocalizes in vivo with its substrates at various sites along cytoskeletal structures. We propose that Capn3-mediated cleavage produces an adaptive response of muscle cells to external and/or internal stimuli, establishing Capn3 as a muscle cytoskeleton regulator.


Subject(s)
Adaptor Proteins, Signal Transducing , Calpain/metabolism , Muscle, Skeletal/enzymology , Actins/metabolism , Animals , Autolysis , Calpain/chemistry , Calpain/genetics , Catalytic Domain , Cells, Cultured , Connectin , Contractile Proteins/metabolism , Cytoskeletal Proteins , Cytoskeleton/enzymology , Enzyme Activation , Filamins , In Vitro Techniques , Mice , Microfilament Proteins/metabolism , Models, Biological , Muscle Proteins/metabolism , NIH 3T3 Cells , Phosphoproteins/metabolism , Protein Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcolemma/enzymology , Sarcomeres/enzymology , Substrate Specificity , Talin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...