Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217605

ABSTRACT

The mechanoenzyme dynamin 2 (DNM2) is crucial for intracellular organization and trafficking. DNM2 is mutated in dominant centronuclear myopathy (DNM2-CNM), a muscle disease characterized by defects in organelle positioning in myofibers. It remains unclear how the in vivo functions of DNM2 are regulated in muscle. Moreover, there is no therapy for DNM2-CNM to date. Here, we overexpressed human amphiphysin 2 (BIN1), a membrane remodeling protein mutated in other CNM forms, in Dnm2RW/+ and Dnm2RW/RW mice modeling mild and severe DNM2-CNM, through transgenesis or with adeno-associated virus (AAV). Increasing BIN1 improved muscle atrophy and main histopathological features of Dnm2RW/+ mice and rescued the perinatal lethality and survival of Dnm2RW/RW mice. In vitro experiments showed that BIN1 binds and recruits DNM2 to membrane tubules, and that the BIN1-DNM2 complex regulates tubules fission. Overall, BIN1 is a potential therapeutic target for dominant centronuclear myopathy linked to DNM2 mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dynamin II/physiology , Muscular Atrophy/physiopathology , Muscular Diseases/pathology , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Dynamin II/genetics , Dynamin II/metabolism , Humans , Mice , Mice, Knockout , Protein Binding
2.
Mol Ther ; 30(2): 868-880, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34371181

ABSTRACT

Mutations in the BIN1 (Bridging Interactor 1) gene, encoding the membrane remodeling protein amphiphysin 2, cause centronuclear myopathy (CNM) associated with severe muscle weakness and myofiber disorganization and hypotrophy. There is no available therapy, and the validation of therapeutic proof of concept is impaired by the lack of a faithful and easy-to-handle mammalian model. Here, we generated and characterized the Bin1mck-/- mouse through Bin1 knockout in skeletal muscle. Bin1mck-/- mice were viable, unlike the constitutive Bin1 knockout, and displayed decreased muscle force and most histological hallmarks of CNM, including myofiber hypotrophy and intracellular disorganization. Notably, Bin1mck-/- myofibers presented strong defects in mitochondria and T-tubule networks associated with deficient calcium homeostasis and excitation-contraction coupling at the triads, potentially representing the main pathomechanisms. Systemic injection of antisense oligonucleotides (ASOs) targeting Dnm2 (Dynamin 2), which codes for dynamin 2, a BIN1 binding partner regulating membrane fission and mutated in other forms of CNM, improved muscle force and normalized the histological Bin1mck-/- phenotypes within 5 weeks. Overall, we generated a faithful mammalian model for CNM linked to BIN1 defects and validated Dnm2 ASOs as a first translatable approach to efficiently treat BIN1-CNM.


Subject(s)
Dynamin II , Myopathies, Structural, Congenital , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Down-Regulation , Dynamin II/genetics , Mammals , Mice , Muscle, Skeletal/metabolism , Mutation , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Nerve Tissue Proteins/genetics , Phenotype , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
BMC Neurol ; 22(1): 115, 2022 Mar 24.
Article in English | MEDLINE | ID: mdl-35331153

ABSTRACT

BACKGROUND: Hereditary spastic paraplegias (HSPs) are progressively debilitating neurodegenerative disorders that follow heterogenous patterns of Mendelian inheritance. Available epidemiological evidence provides limited incidence and prevalence data, especially at the genetic subtype level, preventing a realistic estimation of the true social burden of the disease. The objectives of this study were to (1) review the literature on epidemiology of HSPs; and (2) develop an epidemiological model of the prevalence of HSP, focusing on four common HSP genetic subtypes at the country and region-level. METHODS: A model was constructed estimating the incidence at birth, survival, and prevalence of four genetic subtypes of HSP based on the most appropriate published literature. The key model parameters were assessed by HSP clinical experts, who provided feedback on the validity of assumptions. A model was then finalized and validated through comparison of outputs against available evidence. The global, regional, and national prevalence and patient pool were calculated per geographic region and per genetic subtype. RESULTS: The HSP global prevalence was estimated to be 3.6 per 100,000 for all HSP forms, whilst the estimated global prevalence per genetic subtype was 0.90 (SPG4), 0.22 (SPG7), 0.34 (SPG11), and 0.13 (SPG15), respectively. This equates to an estimated 3365 (SPG4) and 872 (SPG11) symptomatic patients, respectively, in the USA. CONCLUSIONS: This is the first epidemiological model of HSP prevalence at the genetic subtype-level reported at multiple geographic levels. This study offers additional data to better capture the burden of illness due to mutations in common genes causing HSP, that can inform public health policy and healthcare service planning, especially in regions with higher estimated prevalence of HSP.


Subject(s)
Spastic Paraplegia, Hereditary , ATPases Associated with Diverse Cellular Activities/genetics , Humans , Incidence , Infant, Newborn , Metalloendopeptidases/genetics , Mutation , Prevalence , Proteins/genetics , Spastic Paraplegia, Hereditary/epidemiology , Spastic Paraplegia, Hereditary/genetics
4.
Hum Mol Genet ; 28(24): 4067-4077, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31628461

ABSTRACT

Dynamin 2 (DNM2) is a ubiquitously expressed GTPase implicated in many cellular functions such as membrane trafficking and cytoskeleton regulation. Dominant mutations in DNM2 result in tissue-specific diseases affecting peripheral nerves (Charcot-Marie-Tooth neuropathy, CMT) or skeletal muscles (centronuclear myopathy, CNM). However, the reason for this tissue specificity is unknown, and it remains unclear if these diseases share a common pathomechanism. To compare the disease pathophysiological mechanisms in skeletal muscle, we exogenously expressed wild-type DNM2 (WT-DNM2), the DNM2-CMT mutation K562E or DNM2-CNM mutations R465W and S619L causing adult and neonatal forms, respectively, by intramuscular adeno-associated virus (AAV) injections. All muscles expressing exogenous WT-DNM2 and CNM or CMT mutations exhibited reduced muscle force. However, only expression of CNM mutations and WT-DNM2 correlated with CNM-like histopathological hallmarks of nuclei centralization and reduced fiber size. The extent of alterations correlated with clinical severity in patients. Ultrastructural and immunofluorescence analyses highlighted defects of the triads, mitochondria and costameres as major causes of the CNM phenotype. Despite the reduction in force upon expression of the DNM2-CMT mutation, muscle histology and ultrastructure were almost normal. However, the neuromuscular junction was affected in all DNM2-injected muscles, with the DNM2-CMT mutation inducing the most severe alterations, potentially explaining the reduction in force observed with this mutant. In conclusion, expression of WT and CNM mutants recreate a CNM-like phenotype, suggesting CNM mutations are gain-of-function. Histological, ultrastructural and molecular analyses pointed to key pathways uncovering the different pathomechanisms involved in centronuclear myopathy or Charcot-Marie-Tooth neuropathy linked to DNM2 mutations.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Dynamin II/genetics , Myopathies, Structural, Congenital/genetics , Animals , Charcot-Marie-Tooth Disease/metabolism , Dynamin II/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Mutation , Myopathies, Structural, Congenital/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiopathology , Peripheral Nerves/metabolism , Peripheral Nerves/physiopathology , Phenotype
5.
Proc Natl Acad Sci U S A ; 115(43): 11066-11071, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30291191

ABSTRACT

Centronuclear myopathies (CNM) are a group of severe muscle diseases for which no effective therapy is currently available. We have previously shown that reduction of the large GTPase DNM2 in a mouse model of the X-linked form, due to loss of myotubularin phosphatase MTM1, prevents the development of the skeletal muscle pathophysiology. As DNM2 is mutated in autosomal dominant forms, here we tested whether DNM2 reduction can rescue DNM2-related CNM in a knock-in mouse harboring the p.R465W mutation (Dnm2RW/+) and displaying a mild CNM phenotype similar to patients with the same mutation. A single intramuscular injection of adeno-associated virus-shRNA targeting Dnm2 resulted in reduction in protein levels 5 wk post injection, with a corresponding improvement in muscle mass and fiber size distribution, as well as an improvement in histopathological CNM features. To establish a systemic treatment, weekly i.p. injections of antisense oligonucleotides targeting Dnm2 were administered to Dnm2RW/+mice for 5 wk. While muscle mass, histopathology, and muscle ultrastructure were perturbed in Dnm2RW/+mice compared with wild-type mice, these features were indistinguishable from wild-type mice after reducing DNM2. Therefore, DNM2 knockdown via two different strategies can efficiently correct the myopathy due to DNM2 mutations, and it provides a common therapeutic strategy for several forms of centronuclear myopathy. Furthermore, we provide an example of treating a dominant disease by targeting both alleles, suggesting that this strategy may be applied to other dominant diseases.


Subject(s)
Dynamin II/genetics , Myopathies, Structural, Congenital/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Mutation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics
6.
Int J Mol Sci ; 22(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34768808

ABSTRACT

Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.


Subject(s)
Myopathies, Structural, Congenital/physiopathology , Animals , Autophagy , Dynamins , Humans , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/therapy , Nerve Tissue Proteins , Protein Tyrosine Phosphatases, Non-Receptor , Ryanodine Receptor Calcium Release Channel
7.
Hum Mol Genet ; 26(19): 3736-3748, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28934386

ABSTRACT

Myotubularins (MTMs) are active or dead phosphoinositides phosphatases defining a large protein family conserved through evolution and implicated in different neuromuscular diseases. Loss-of-function mutations in MTM1 cause the severe congenital myopathy called myotubular myopathy (or X-linked centronuclear myopathy) while mutations in the MTM1-related protein MTMR2 cause a recessive Charcot-Marie-Tooth peripheral neuropathy. Here we aimed to determine the functional specificity and redundancy of MTM1 and MTMR2, and to assess their abilities to compensate for a potential therapeutic strategy. Using molecular investigations and heterologous expression of human MTMs in yeast cells and in Mtm1 knockout mice, we characterized several naturally occurring MTMR2 isoforms with different activities. We identified the N-terminal domain as responsible for functional differences between MTM1 and MTMR2. An N-terminal extension observed in MTMR2 is absent in MTM1, and only the short MTMR2 isoform lacking this N-terminal extension behaved similarly to MTM1 in yeast and mice. Moreover, adeno-associated virus-mediated exogenous expression of several MTMR2 isoforms ameliorates the myopathic phenotype owing to MTM1 loss, with increased muscle force, reduced myofiber atrophy, and reduction of the intracellular disorganization hallmarks associated with myotubular myopathy. Noteworthy, the short MTMR2 isoform provided a better rescue when compared with the long MTMR2 isoform. In conclusion, these results point to the molecular basis for MTMs functional specificity. They also provide the proof-of-concept that expression of the neuropathy-associated MTMR2 gene improves the MTM1-associated myopathy, thus identifying MTMR2 as a novel therapeutic target for myotubular myopathy.


Subject(s)
Myopathies, Structural, Congenital/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Humans , Male , Mice , Mice, Knockout , Mutation , Myopathies, Structural, Congenital/enzymology , Myopathies, Structural, Congenital/metabolism , Phenotype , Protein Domains , Protein Isoforms , Protein Tyrosine Phosphatases, Non-Receptor/genetics
8.
Mol Ther ; 26(4): 1082-1092, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29506908

ABSTRACT

Myotubular myopathy, or X-linked centronuclear myopathy, is a severe muscle disorder representing a significant burden for patients and their families. It is clinically characterized by neonatal and severe muscle weakness and atrophy. Mutations in the myotubularin (MTM1) gene cause myotubular myopathy, and no specific curative treatment is available. We previously found that dynamin 2 (DNM2) is upregulated in both Mtm1 knockout and patient muscle samples, whereas its reduction through antisense oligonucleotides rescues the clinical and histopathological features of this myopathy in mice. Here, we propose a novel approach targeting Dnm2 mRNA. We screened and validated in vitro and in vivo several short hairpin RNA (shRNA) sequences that efficiently target Dnm2 mRNA. A single intramuscular injection of AAV-shDnm2 resulted in long-term reduction of DNM2 protein level and restored muscle force, mass, histology, and myofiber ultrastructure and prevented molecular defects linked to the disease. Our results demonstrate a robust DNM2 knockdown and provide an alternative strategy based on reduction of DNM2 to treat myotubular myopathy.


Subject(s)
Dependovirus/genetics , Dynamin II/genetics , Genetic Therapy , Genetic Vectors/genetics , Myopathies, Structural, Congenital/genetics , RNA, Small Interfering/genetics , Animals , Disease Models, Animal , Gene Knockdown Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Immunohistochemistry , Injections, Intramuscular , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myopathies, Structural, Congenital/pathology , Myopathies, Structural, Congenital/therapy , Phenotype , RNA Interference , RNA, Messenger , Treatment Outcome
9.
PLoS Genet ; 12(2): e1005709, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26872257

ABSTRACT

The 16p11.2 600 kb BP4-BP5 deletion and duplication syndromes have been associated with developmental delay; autism spectrum disorders; and reciprocal effects on the body mass index, head circumference and brain volumes. Here, we explored these relationships using novel engineered mouse models carrying a deletion (Del/+) or a duplication (Dup/+) of the Sult1a1-Spn region homologous to the human 16p11.2 BP4-BP5 locus. On a C57BL/6N inbred genetic background, Del/+ mice exhibited reduced weight and impaired adipogenesis, hyperactivity, repetitive behaviors, and recognition memory deficits. In contrast, Dup/+ mice showed largely opposite phenotypes. On a F1 C57BL/6N × C3B hybrid genetic background, we also observed alterations in social interaction in the Del/+ and the Dup/+ animals, with other robust phenotypes affecting recognition memory and weight. To explore the dosage effect of the 16p11.2 genes on metabolism, Del/+ and Dup/+ models were challenged with high fat and high sugar diet, which revealed opposite energy imbalance. Transcriptomic analysis revealed that the majority of the genes located in the Sult1a1-Spn region were sensitive to dosage with a major effect on several pathways associated with neurocognitive and metabolic phenotypes. Whereas the behavioral consequence of the 16p11 region genetic dosage was similar in mice and humans with activity and memory alterations, the metabolic defects were opposite: adult Del/+ mice are lean in comparison to the human obese phenotype and the Dup/+ mice are overweight in comparison to the human underweight phenotype. Together, these data indicate that the dosage imbalance at the 16p11.2 locus perturbs the expression of modifiers outside the CNV that can modulate the penetrance, expressivity and direction of effects in both humans and mice.


Subject(s)
Chromosome Deletion , Chromosome Duplication/genetics , Cognition , Adiposity , Alleles , Animals , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Behavior, Animal , Body Weight , Brain/metabolism , Brain/physiopathology , Chromosomes, Mammalian/genetics , Craniofacial Abnormalities/genetics , Diet, High-Fat , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Gene Rearrangement/genetics , Hippocampus/physiopathology , Memory , Mice, Inbred C57BL , Motor Activity , Phenotype , Synaptic Transmission/genetics , Syndrome , Weaning
10.
Hum Mol Genet ; 23(3): 618-36, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24087791

ABSTRACT

Utrophin is a potential therapeutic target for the fatal muscle disease, Duchenne muscular dystrophy (DMD). In adult skeletal muscle, utrophin is restricted to the neuromuscular and myotendinous junctions and can compensate for dystrophin loss in mdx mice, a mouse model of DMD, but requires sarcolemmal localization. NFATc1-mediated transcription regulates utrophin expression and the LIM protein, FHL1 which promotes muscle hypertrophy, is a transcriptional activator of NFATc1. By generating mdx/FHL1-transgenic mice, we demonstrate that FHL1 potentiates NFATc1 activation of utrophin to ameliorate the dystrophic pathology. Transgenic FHL1 expression increased sarcolemmal membrane stability, reduced muscle degeneration, decreased inflammation and conferred protection from contraction-induced injury in mdx mice. Significantly, FHL1 expression also reduced progressive muscle degeneration and fibrosis in the diaphragm of aged mdx mice. FHL1 enhanced NFATc1 activation of the utrophin promoter and increased sarcolemmal expression of utrophin in muscles of mdx mice, directing the assembly of a substitute utrophin-glycoprotein complex, and revealing a novel FHL1-NFATc1-utrophin signaling axis that can functionally compensate for dystrophin.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Animals , Diaphragm/physiopathology , Dystrophin/genetics , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Contraction , Muscle Proteins/metabolism , Muscle, Skeletal/physiopathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic , Sarcolemma/metabolism , Signal Transduction , Utrophin/genetics , Utrophin/metabolism
11.
PLoS Genet ; 9(6): e1003430, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23754947

ABSTRACT

Amphiphysin 2, encoded by BIN1, is a key factor for membrane sensing and remodelling in different cell types. Homozygous BIN1 mutations in ubiquitously expressed exons are associated with autosomal recessive centronuclear myopathy (CNM), a mildly progressive muscle disorder typically showing abnormal nuclear centralization on biopsies. In addition, misregulation of BIN1 splicing partially accounts for the muscle defects in myotonic dystrophy (DM). However, the muscle-specific function of amphiphysin 2 and its pathogenicity in both muscle disorders are not well understood. In this study we identified and characterized the first mutation affecting the splicing of the muscle-specific BIN1 exon 11 in a consanguineous family with rapidly progressive and ultimately fatal centronuclear myopathy. In parallel, we discovered a mutation in the same BIN1 exon 11 acceptor splice site as the genetic cause of the canine Inherited Myopathy of Great Danes (IMGD). Analysis of RNA from patient muscle demonstrated complete skipping of exon 11 and BIN1 constructs without exon 11 were unable to promote membrane tubulation in differentiated myotubes. Comparative immunofluorescence and ultrastructural analyses of patient and canine biopsies revealed common structural defects, emphasizing the importance of amphiphysin 2 in membrane remodelling and maintenance of the skeletal muscle triad. Our data demonstrate that the alteration of the muscle-specific function of amphiphysin 2 is a common pathomechanism for centronuclear myopathy, myotonic dystrophy, and IMGD. The IMGD dog is the first faithful model for human BIN1-related CNM and represents a mammalian model available for preclinical trials of potential therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alternative Splicing/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Myopathies, Structural, Congenital/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Base Sequence , Dogs , Exons/genetics , Female , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/veterinary , Organ Specificity , RNA Splice Sites/genetics
12.
J Cell Sci ; 126(Pt 8): 1806-19, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444364

ABSTRACT

The sarcoplasmic reticulum (SR) is a specialized form of endoplasmic reticulum (ER) in skeletal muscle and is essential for calcium homeostasis. The mechanisms involved in SR remodeling and maintenance of SR subdomains are elusive. In this study, we identified myotubularin (MTM1), a phosphoinositide phosphatase mutated in X-linked centronuclear myopathy (XLCNM, or myotubular myopathy), as a key regulator of phosphatidylinositol 3-monophosphate (PtdIns3P) levels at the SR. MTM1 is predominantly located at the SR cisternae of the muscle triads, and Mtm1-deficient mouse muscles and myoblasts from XLCNM patients exhibit abnormal SR/ER networks. In vivo modulation of MTM1 enzymatic activity in skeletal muscle using ectopic expression of wild-type or a dead-phosphatase MTM1 protein leads to differential SR remodeling. Active MTM1 is associated with flat membrane stacks, whereas dead-phosphatase MTM1 mutant promotes highly curved cubic membranes originating from the SR and enriched in PtdIns3P. Overexpression of a tandem FYVE domain with high affinity for PtdIns3P alters the shape of the SR cisternae at the triad. Our findings, supported by the parallel analysis of the Mtm1-null mouse and an in vivo study, reveal a direct function of MTM1 enzymatic activity in SR remodeling and a key role for PtdIns3P in promoting SR membrane curvature in skeletal muscle. We propose that alteration in SR remodeling is a primary cause of X-linked centronuclear myopathy. The tight regulation of PtdIns3P on specific membrane subdomains may be a general mechanism to control membrane curvature.


Subject(s)
Muscle, Skeletal/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Blotting, Western , Cell Line , Immunoprecipitation , Male , Mice , Microscopy, Electron, Transmission , Muscle, Skeletal/ultrastructure , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/genetics
13.
PLoS Genet ; 8(4): e1002595, 2012.
Article in English | MEDLINE | ID: mdl-22496665

ABSTRACT

Proteins involved in membrane remodeling play an essential role in a plethora of cell functions including endocytosis and intracellular transport. Defects in several of them lead to human diseases. Myotubularins, amphiphysins, and dynamins are all proteins implicated in membrane trafficking and/or remodeling. Mutations in myotubularin, amphiphysin 2 (BIN1), and dynamin 2 lead to different forms of centronuclear myopathy, while mutations in myotubularin-related proteins cause Charcot-Marie-Tooth neuropathies. In addition to centronuclear myopathy, dynamin 2 is also mutated in a dominant form of Charcot-Marie-Tooth neuropathy. While several proteins from these different families are implicated in similar diseases, mutations in close homologues or in the same protein in the case of dynamin 2 lead to diseases affecting different tissues. This suggests (1) a common molecular pathway underlying these different neuromuscular diseases, and (2) tissue-specific regulation of these proteins. This review discusses the pathophysiology of the related neuromuscular diseases on the basis of animal models developed for proteins of the myotubularin, amphiphysin, and dynamin families. A better understanding of the common mechanisms between these neuromuscular disorders will lead to more specific health care and therapeutic approaches.


Subject(s)
Cell Membrane , Dynamins , Nerve Tissue Proteins , Neuromuscular Diseases , Protein Tyrosine Phosphatases, Non-Receptor , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Disease Models, Animal , Dynamins/genetics , Dynamins/metabolism , Humans , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Diseases/genetics , Neuromuscular Diseases/metabolism , Phylogeny , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
14.
PLoS Genet ; 8(10): e1002965, 2012.
Article in English | MEDLINE | ID: mdl-23071445

ABSTRACT

Myotubularin MTM1 is a phosphoinositide (PPIn) 3-phosphatase mutated in X-linked centronuclear myopathy (XLCNM; myotubular myopathy). We investigated the involvement of MTM1 enzymatic activity on XLCNM phenotypes. Exogenous expression of human MTM1 in yeast resulted in vacuolar enlargement, as a consequence of its phosphatase activity. Expression of mutants from patients with different clinical progression and determination of PtdIns3P and PtdIns5P cellular levels confirmed the link between vacuolar morphology and MTM1 phosphatase activity, and showed that some disease mutants retain phosphatase activity. Viral gene transfer of phosphatase-dead myotubularin mutants (MTM1(C375S) and MTM1(S376N)) significantly improved most histological signs of XLCNM displayed by a Mtm1-null mouse, at similar levels as wild-type MTM1. Moreover, the MTM1(C375S) mutant improved muscle performance and restored the localization of nuclei, triad alignment, and the desmin intermediate filament network, while it did not normalize PtdIns3P levels, supporting phosphatase-independent roles of MTM1 in maintaining normal muscle performance and organelle positioning in skeletal muscle. Among the different XLCNM signs investigated, we identified only triad shape and fiber size distribution as being partially dependent on MTM1 phosphatase activity. In conclusion, this work uncovers MTM1 roles in the structural organization of muscle fibers that are independent of its enzymatic activity. This underlines that removal of enzymes should be used with care to conclude on the physiological importance of their activity.


Subject(s)
Myopathies, Structural, Congenital/genetics , Phenotype , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Animals , Desmin/metabolism , Disease Models, Animal , Enzyme Activation/genetics , Gene Expression , Humans , Male , Mice , Mice, Knockout , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Mutation , Myopathies, Structural, Congenital/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
15.
Am J Pathol ; 178(5): 2224-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21514436

ABSTRACT

Dynamin 2 (DNM2) is a large GTPase implicated in many cellular functions, including cytoskeleton regulation and endocytosis. Although ubiquitously expressed, DNM2 was found mutated in two genetic disorders affecting different tissues: autosomal dominant centronuclear myopathy (ADCNM; skeletal muscle) and peripheral Charcot-Marie-Tooth neuropathy (peripheral nerve). To gain insight into the function of DNM2 in skeletal muscle and the pathological mechanisms leading to ADCNM, we introduced wild-type DNM2 (WT-DNM2) or R465W DNM2 (RW-DNM2), the most common ADCNM mutation, into adult wild-type mouse skeletal muscle by intramuscular adeno-associated virus injections. We detected altered localization of RW-DNM2 in mouse muscle. Several ADCNM features were present in RW-DNM2 mice: fiber atrophy, nuclear mislocalization, and altered mitochondrial staining, with a corresponding reduction in specific maximal muscle force. The sarcomere and triad structures were also altered. We report similar findings in muscle biopsy specimens from an ADCNM patient with the R465W mutation. In addition, expression of wild-type DNM2 induced some muscle defects, albeit to a lesser extent than RW-DNM2, suggesting that the R465W mutation has enhanced activity in vivo. In conclusion, we show the RW-DNM2 mutation acts in a dominant manner to cause ADCNM in adult muscle, and the disease arises from a primary defect in skeletal muscle rather than secondary to peripheral nerve involvement. Therefore, DNM2 plays important roles in the maintenance of adult muscle fibers.


Subject(s)
Dynamin II/genetics , Muscle, Skeletal/pathology , Mutation , Myopathies, Structural, Congenital/genetics , Animals , Blotting, Western , Dynamin II/metabolism , Fluorescent Antibody Technique , Humans , Mice , Microscopy, Electron, Transmission , Muscle Weakness/genetics , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Transduction, Genetic
16.
Dis Model Mech ; 15(7)2022 07 01.
Article in English | MEDLINE | ID: mdl-35642830

ABSTRACT

Generating reliable preclinical data in animal models of disease is essential in therapy development. Here, we performed statistical analysis and joint longitudinal-survival modeling of the progressive phenotype observed in Mtm1-/y mice, a reliable model for myotubular myopathy. Analysis of historical data was used to generate a model for phenotype progression, which was then confirmed with phenotypic data from a new colony of mice derived via in vitro fertilization in an independent animal house, highlighting the reproducibility of disease phenotype in Mtm1-/y mice. These combined data were used to refine the phenotypic parameters analyzed in these mice and improve the model generated for expected disease progression. The disease progression model was then used to test the therapeutic efficacy of Dnm2 targeting. Dnm2 reduction by antisense oligonucleotides blocked or postponed disease development, and resulted in a significant dose-dependent improvement outside the expected disease progression in untreated Mtm1-/y mice. This provides an example of optimizing disease analysis and testing therapeutic efficacy in a preclinical model, which can be applied by scientists testing therapeutic approaches using neuromuscular disease models in different laboratories. This article has an associated First Person interview with the joint first authors of the paper.


Subject(s)
Myopathies, Structural, Congenital , Protein Tyrosine Phosphatases, Non-Receptor , Animals , Disease Progression , Dynamin II/genetics , Humans , Mice , Muscle, Skeletal , Mutation , Myopathies, Structural, Congenital/genetics , Phenotype , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Reproducibility of Results
17.
Neurol Genet ; 8(6): e200027, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36324371

ABSTRACT

Background and Objectives: Centronuclear myopathy (CNM) due to mutations in the dynamin 2 gene, DNM2, is a rare neuromuscular disease about which little is known. The objective of this study was to describe the range of clinical presentations and subsequent natural history of DNM2-related CNM. Methods: Pediatric and adult patients with suspicion for a CNM diagnosis and confirmed heterozygous pathogenic variants in DNM2 were ascertained between December 8, 2000, and May 1, 2019. Data were collected through a retrospective review of genetic testing results, clinical records, and pathology slides combined with patient-reported clinical findings via questionnaires. Results: Forty-two patients with DNM2-related CNM, whose ages ranged from 0.95 to 75.76 years at most recent contact, were enrolled from 34 families in North or South America and Europe. There were 8 different DNM2 pathogenic variants within the cohort. Of the 32 biopsied patients, all had histologic features of CNM. The disease onset was in infancy or childhood in 81% of the cohort, and more than half of the patients had high arched palates, indicative of weakness in utero. Ambulation was affected in nearly all (92%) the patients, and while the rapidity of progression was variable, most (67%) reported a "deteriorating course." Ptosis, ophthalmoparesis, facial weakness, dysphagia, and respiratory insufficiency were commonly reported. One-third of the patients experienced restricted jaw mobility. Certain pathogenic variants appear to correlate with a more severe phenotype. Discussion: DNM2-related CNM has a predominantly early-onset, often congenital, myopathy resulting in progressive difficulty with ambulation and occasionally bulbar and respiratory dysfunction. This detailed characterization of the phenotype provides important information to support clinical trial readiness for future disease-modifying therapies.

18.
Nat Commun ; 13(1): 6849, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36369230

ABSTRACT

Dynamin 2 mechanoenzyme is a key regulator of membrane remodeling and gain-of-function mutations in its gene cause centronuclear myopathies. Here, we investigate the functions of dynamin 2 isoforms and their associated phenotypes and, specifically, the ubiquitous and muscle-specific dynamin 2 isoforms expressed in skeletal muscle. In cell-based assays, we show that a centronuclear myopathy-related mutation in the ubiquitous but not the muscle-specific dynamin 2 isoform causes increased membrane fission. In vivo, overexpressing the ubiquitous dynamin 2 isoform correlates with severe forms of centronuclear myopathy, while overexpressing the muscle-specific isoform leads to hallmarks seen in milder cases of the disease. Previous mouse studies suggested that reduction of the total dynamin 2 pool could be therapeutic for centronuclear myopathies. Here, dynamin 2 splice switching from muscle-specific to ubiquitous dynamin 2 aggravated the phenotype of a severe X-linked form of centronuclear myopathy caused by loss-of-function of the MTM1 phosphatase, supporting the importance of targeting the ubiquitous isoform for efficient therapy in muscle. Our results highlight that the ubiquitous and not the muscle-specific dynamin 2 isoform is the main modifier contributing to centronuclear myopathy pathology.


Subject(s)
Dynamin II , Myopathies, Structural, Congenital , Animals , Mice , Dynamin II/genetics , Muscle, Skeletal/pathology , Mutation , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Phenotype , Protein Isoforms/genetics
19.
J Clin Invest ; 118(3): 904-12, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18274675

ABSTRACT

Reducing body myopathy (RBM) is a rare disorder causing progressive muscular weakness characterized by aggresome-like inclusions in the myofibrils. Identification of genes responsible for RBM by traditional genetic approaches has been impossible due to the frequently sporadic occurrence in affected patients and small family sizes. As an alternative approach to gene identification, we used laser microdissection of intracytoplasmic inclusions identified in patient muscle biopsies, followed by nanoflow liquid chromatography-tandem mass spectrometry and proteomic analysis. The most prominent component of the inclusions was the Xq26.3-encoded four and a half LIM domain 1 (FHL1) protein, expressed predominantly in skeletal but also in cardiac muscle. Mutational analysis identified 4 FHL1 mutations in 2 sporadic unrelated females and in 2 families with severely affected boys and less-affected mothers. Transfection of kidney COS-7 and skeletal muscle C2C12 cells with mutant FHL1 induced the formation of aggresome-like inclusions that incorporated both mutant and wild-type FHL1 and trapped other proteins in a dominant-negative manner. Thus, a novel laser microdissection/proteomics approach has helped identify both inherited and de novo mutations in FHL1, thereby defining a new X-linked protein aggregation disorder of muscle.


Subject(s)
Inclusion Bodies/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Muscular Diseases/genetics , Mutation , Proteomics/methods , Amino Acid Sequence , Genetic Diseases, X-Linked/genetics , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/chemistry , LIM Domain Proteins , Models, Molecular , Molecular Sequence Data , Muscle Proteins/analysis , Muscle Proteins/chemistry , Muscular Diseases/metabolism , Transfection
20.
Orphanet J Rare Dis ; 16(1): 3, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407688

ABSTRACT

BACKGROUND: Centronuclear myopathies are severe rare congenital diseases. The clinical variability and genetic heterogeneity of these myopathies result in major challenges in clinical trial design. Alternative strategies to large placebo-controlled trials that have been used in other rare diseases (e.g., the use of surrogate markers or of historical controls) have limitations that Bayesian statistics may address. Here we present a Bayesian model that uses each patient's own natural history study data to predict progression in the absence of treatment. This prospective multicentre natural history evaluated 4-year follow-up data from 59 patients carrying mutations in the MTM1 or DNM2 genes. METHODS: Our approach focused on evaluation of forced expiratory volume in 1 s (FEV1) in 6- to 18-year-old children. A patient was defined as a responder if an improvement was observed after treatment and the predictive probability of such improvement in absence of intervention was less than 0.01. An FEV1 response was considered clinically relevant if it corresponded to an increase of more than 8%. RESULTS: The key endpoint of a clinical trial using this model is the rate of response. The power of the study is based on the posterior probability that the rate of response observed is greater than the rate of response that would be observed in the absence of treatment predicted based on the individual patient's previous natural history. In order to appropriately control for Type 1 error, the threshold probability by which the difference in response rates exceeds zero was adapted to 91%, ensuring a 5% overall Type 1 error rate for the trial. CONCLUSIONS: Bayesian statistical analysis of natural history data allowed us to reliably simulate the evolution of symptoms for individual patients over time and to probabilistically compare these simulated trajectories to actual observed post-treatment outcomes. The proposed model adequately predicted the natural evolution of patients over the duration of the study and will facilitate a sufficiently powerful trial design that can cope with the disease's rarity. Further research and ongoing dialog with regulatory authorities are needed to allow for more applications of Bayesian statistics in orphan disease research.


Subject(s)
Myopathies, Structural, Congenital , Adolescent , Bayes Theorem , Child , Clinical Trials as Topic , Disease Progression , Humans , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL