Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
JCI Insight ; 8(9)2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36943412

RESUMEN

Phosphoinositides (PIs) are membrane lipids that regulate signal transduction and vesicular trafficking. X-linked centronuclear myopathy (XLCNM), also called myotubular myopathy, results from loss-of-function mutations in the MTM1 gene, which encodes the myotubularin phosphatidylinositol 3-phosphate (PtdIns3P) lipid phosphatase. No therapy for this disease is currently available. Previous studies showed that loss of expression of the class II phosphoinositide 3-kinase (PI3K) PI3KC2ß (PI3KC2B) protein improved the phenotypes of an XLCNM mouse model. PI3Ks are well known to have extensive scaffolding functions and the importance of the catalytic activity of this PI3K for rescue remains unclear. Here, using PI3KC2ß kinase-dead mice, we show that the selective inactivation of PI3KC2ß kinase activity is sufficient to fully prevent muscle atrophy and weakness, histopathology, and sarcomere and triad disorganization in Mtm1-knockout mice. This rescue correlates with normalization of PtdIns3P level and mTORC1 activity, a key regulator of protein synthesis and autophagy. Conversely, lack of PI3KC2ß kinase activity did not rescue the histopathology of the BIN1 autosomal CNM mouse model. Overall, these findings support the development of specific PI3KC2ß kinase inhibitors to cure myotubular myopathy.


Asunto(s)
Miopatías Estructurales Congénitas , Fosfatidilinositol 3-Quinasas , Animales , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositoles , Mutación , Ratones Noqueados , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35217605

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Dinamina II/fisiología , Atrofia Muscular/fisiopatología , Enfermedades Musculares/patología , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Dinamina II/genética , Dinamina II/metabolismo , Humanos , Ratones , Ratones Noqueados , Unión Proteica
3.
Mol Ther ; 30(2): 868-880, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34371181

RESUMEN

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.


Asunto(s)
Dinamina II , Miopatías Estructurales Congénitas , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Regulación hacia Abajo , Dinamina II/genética , Mamíferos , Ratones , Músculo Esquelético/metabolismo , Mutación , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/terapia , Proteínas del Tejido Nervioso/genética , Fenotipo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
4.
Mol Ther ; 29(8): 2514-2534, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-33940157

RESUMEN

Omics analyses are powerful methods to obtain an integrated view of complex biological processes, disease progression, or therapy efficiency. However, few studies have compared different disease forms and different therapy strategies to define the common molecular signatures representing the most significant implicated pathways. In this study, we used RNA sequencing and mass spectrometry to profile the transcriptomes and proteomes of mouse models for three forms of centronuclear myopathies (CNMs), untreated or treated with either a drug (tamoxifen), antisense oligonucleotides reducing the level of dynamin 2 (DNM2), or following modulation of DNM2 or amphiphysin 2 (BIN1) through genetic crosses. Unsupervised analysis and differential gene and protein expression were performed to retrieve CNM molecular signatures. Longitudinal studies before, at, and after disease onset highlighted potential disease causes and consequences. Main pathways in the common CNM disease signature include muscle contraction, regeneration and inflammation. The common therapy signature revealed novel potential therapeutic targets, including the calcium regulator sarcolipin. We identified several novel biomarkers validated in muscle and/or plasma through RNA quantification, western blotting, and enzyme-linked immunosorbent assay (ELISA) assays, including ANXA2 and IGFBP2. This study validates the concept of using multi-omics approaches to identify molecular signatures common to different disease forms and therapeutic strategies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Miopatías Estructurales Congénitas/tratamiento farmacológico , Oligonucleótidos Antisentido/uso terapéutico , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteómica/métodos , Tamoxifeno/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Dinamina II/antagonistas & inhibidores , Humanos , Estudios Longitudinales , Espectrometría de Masas , Ratones , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Análisis de Secuencia de ARN , Proteínas Supresoras de Tumor/antagonistas & inhibidores
5.
Dis Model Mech ; 13(11)2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-32994313

RESUMEN

Skeletal muscle development and regeneration are tightly regulated processes. How the intracellular organization of muscle fibers is achieved during these steps is unclear. Here, we focus on the cellular and physiological roles of amphiphysin 2 (BIN1), a membrane remodeling protein mutated in both congenital and adult centronuclear myopathies (CNM), that is ubiquitously expressed and has skeletal muscle-specific isoforms. We created and characterized constitutive muscle-specific and inducible Bin1 homozygous and heterozygous knockout mice targeting either ubiquitous or muscle-specific isoforms. Constitutive Bin1-deficient mice died at birth from lack of feeding due to a skeletal muscle defect. T-tubules and other organelles were misplaced and altered, supporting a general early role for BIN1 in intracellular organization, in addition to membrane remodeling. Although restricted deletion of Bin1 in unchallenged adult muscles had no impact, the forced switch from the muscle-specific isoforms to the ubiquitous isoforms through deletion of the in-frame muscle-specific exon delayed muscle regeneration. Thus, ubiquitous BIN1 function is necessary for muscle development and function, whereas its muscle-specific isoforms fine tune muscle regeneration in adulthood, supporting that BIN1 CNM with congenital onset are due to developmental defects, whereas later onset may be due to regeneration defects.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Desarrollo de Músculos/fisiología , Músculo Esquelético/fisiología , Proteínas del Tejido Nervioso/metabolismo , Regeneración/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Animales Recién Nacidos , Exones/genética , Conducta Alimentaria , Homocigoto , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/ultraestructura , Especificidad de Órganos , Isoformas de Proteínas/metabolismo , Eliminación de Secuencia , Análisis de Supervivencia
6.
JCI Insight ; 5(18)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32809972

RESUMEN

Classical dynamins are large GTPases regulating membrane and cytoskeleton dynamics, and they are linked to different pathological conditions ranging from neuromuscular diseases to encephalopathy and cancer. Dominant dynamin 2 (DNM2) mutations lead to either mild adult onset or severe autosomal dominant centronuclear myopathy (ADCNM). Our objectives were to better understand the pathomechanism of severe ADCNM and test a potential therapy. Here, we created the Dnm2SL/+ mouse line harboring the common S619L mutation found in patients with severe ADCNM and impairing the conformational switch regulating dynamin self-assembly and membrane remodeling. The Dnm2SL/+ mouse faithfully reproduces severe ADCNM hallmarks with early impaired muscle function and force, together with myofiber hypotrophy. It revealed swollen mitochondria lacking cristae as the main ultrastructural defect and potential cause of the disease. Patient analysis confirmed this structural hallmark. In addition, DNM2 reduction with antisense oligonucleotides after disease onset efficiently reverted locomotor and force defects after only 3 weeks of treatment. Most histological defects including mitochondria alteration were partially or fully rescued. Overall, this study highlights an efficient approach to revert the severe form of dynamin-related centronuclear myopathy. These data also reveal that the dynamin conformational switch is key for muscle function and should be targeted for future therapeutic developments.


Asunto(s)
Dinamina II/fisiología , Mitocondrias/patología , Músculo Esquelético/patología , Mutación , Miopatías Estructurales Congénitas/prevención & control , Oligonucleótidos Antisentido/farmacología , Animales , Dinamina II/antagonistas & inhibidores , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/etiología , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología
7.
Mol Ther Methods Clin Dev ; 17: 1178-1189, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32514412

RESUMEN

Myotubular myopathy, also called X-linked centronuclear myopathy (XL-CNM), is a severe congenital disease targeted for therapeutic trials. To date, biomarkers to monitor disease progression and therapy efficacy are lacking. The Mtm1 -/y mouse is a faithful model for XL-CNM, due to myotubularin 1 (MTM1) loss-of-function mutations. Using both an unbiased approach (RNA sequencing [RNA-seq]) and a directed approach (qRT-PCR and protein level), we identified decreased Mstn levels in Mtm1 -/y muscle, leading to low levels of myostatin in muscle and plasma. Myostatin (Mstn or growth differentiation factor 8 [Gdf8]) is a protein released by myocytes and inhibiting muscle growth and differentiation. Decreasing Dnm2 by genetic cross with Dnm2 +/- mice or by antisense oligonucleotides blocked or postponed disease progression and resulted in an increase in circulating myostatin. In addition, plasma myostatin levels inversely correlated with disease severity and with Dnm2 mRNA levels in muscles. Altered Mstn levels were associated with a generalized disruption of the myostatin pathway. Importantly, in two different forms of CNMs we identified reduced circulating myostatin levels in plasma from patients. This provides evidence of a blood-based biomarker that may be used to monitor disease state in XL-CNM mice and patients and supports monitoring circulating myostatin during clinical trials for myotubular myopathy.

8.
Sci Transl Med ; 11(484)2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894500

RESUMEN

Centronuclear myopathies (CNMs) are severe diseases characterized by muscle weakness and myofiber atrophy. Currently, there are no approved treatments for these disorders. Mutations in the phosphoinositide 3-phosphatase myotubularin (MTM1) are responsible for X-linked CNM (XLCNM), also called myotubular myopathy, whereas mutations in the membrane remodeling Bin/amphiphysin/Rvs protein amphiphysin 2 [bridging integrator 1 (BIN1)] are responsible for an autosomal form of the disease. Here, we investigated the functional relationship between MTM1 and BIN1 in healthy skeletal muscle and in the physiopathology of CNM. Genetic overexpression of human BIN1 efficiently rescued the muscle weakness and life span in a mouse model of XLCNM. Exogenous human BIN1 expression with adeno-associated virus after birth also prevented the progression of the disease, suggesting that human BIN1 overexpression can compensate for the lack of MTM1 expression in this mouse model. Our results showed that MTM1 controls cell adhesion and integrin localization in mammalian muscle. Alterations in this pathway in Mtm1 -/y mice were associated with defects in myofiber shape and size. BIN1 expression rescued integrin and laminin alterations and restored myofiber integrity, supporting the idea that MTM1 and BIN1 are functionally linked and necessary for focal adhesions in skeletal muscle. The results suggest that BIN1 modulation might be an effective strategy for treating XLCNM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesiones Focales/patología , Miopatías Estructurales Congénitas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Animales Recién Nacidos , Adhesiones Focales/metabolismo , Humanos , Integrina beta1/metabolismo , Longevidad , Masculino , Ratones Transgénicos , Fuerza Muscular , Músculos/patología , Músculos/fisiopatología , Músculos/ultraestructura , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/fisiopatología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(43): 11066-11071, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30291191

RESUMEN

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.


Asunto(s)
Dinamina II/genética , Miopatías Estructurales Congénitas/genética , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Mutación/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética
10.
Mol Ther ; 26(4): 1082-1092, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29506908

RESUMEN

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.


Asunto(s)
Dependovirus/genética , Dinamina II/genética , Terapia Genética , Vectores Genéticos/genética , Miopatías Estructurales Congénitas/genética , ARN Interferente Pequeño/genética , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Inmunohistoquímica , Inyecciones Intramusculares , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Fenotipo , Interferencia de ARN , ARN Mensajero , Resultado del Tratamiento
11.
J Clin Invest ; 127(12): 4477-4487, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29130937

RESUMEN

Regulation of skeletal muscle development and organization is a complex process that is not fully understood. Here, we focused on amphiphysin 2 (BIN1, also known as bridging integrator-1) and dynamin 2 (DNM2), two ubiquitous proteins implicated in membrane remodeling and mutated in centronuclear myopathies (CNMs). We generated Bin1-/- Dnm2+/- mice to decipher the physiological interplay between BIN1 and DNM2. While Bin1-/- mice die perinatally from a skeletal muscle defect, Bin1-/- Dnm2+/- mice survived at least 18 months, and had normal muscle force and intracellular organization of muscle fibers, supporting BIN1 as a negative regulator of DNM2. We next characterized muscle-specific isoforms of BIN1 and DNM2. While BIN1 colocalized with and partially inhibited DNM2 activity during muscle maturation, BIN1 had no effect on the isoform of DNM2 found in adult muscle. Together, these results indicate that BIN1 and DNM2 regulate muscle development and organization, function through a common pathway, and define BIN1 as a negative regulator of DNM2 in vitro and in vivo during muscle maturation. Our data suggest that DNM2 modulation has potential as a therapeutic approach for patients with CNM and BIN1 defects. As BIN1 is implicated in cancers, arrhythmia, and late-onset Alzheimer disease, these findings may trigger research directions and therapeutic development for these common diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Dinamina II/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Dinamina II/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/patología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Proteínas del Tejido Nervioso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Supresoras de Tumor/genética
12.
Hum Mol Genet ; 26(19): 3736-3748, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28934386

RESUMEN

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.


Asunto(s)
Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Miopatías Estructurales Congénitas/enzimología , Miopatías Estructurales Congénitas/metabolismo , Fenotipo , Dominios Proteicos , Isoformas de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética
13.
Nat Commun ; 8: 15661, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28589938

RESUMEN

Centronuclear myopathies (CNM) are non-dystrophic muscle diseases for which no effective therapy is currently available. The most severe form, X-linked CNM, is caused by myotubularin 1 (MTM1) loss-of-function mutations, while the main autosomal dominant form is due to dynamin2 (DNM2) mutations. We previously showed that genetic reduction of DNM2 expression in Mtm1 knockout (Mtm1KO) mice prevents development of muscle pathology. Here we show that systemic delivery of Dnm2 antisense oligonucleotides (ASOs) into Mtm1KO mice efficiently reduces DNM2 protein level in muscle and prevents the myopathy from developing. Moreover, systemic ASO injection into severely affected mice leads to reversal of muscle pathology within 2 weeks. Thus, ASO-mediated DNM2 knockdown can efficiently correct muscle defects due to loss of MTM1, providing an attractive therapeutic strategy for this disease.


Asunto(s)
Dinamina II/genética , Miopatías Estructurales Congénitas/genética , Oligonucleótidos Antisentido/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Modelos Animales de Enfermedad , Dinamina II/metabolismo , Femenino , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Contracción Muscular , Músculo Esquelético/metabolismo , Mutación , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/terapia , Fenotipo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Recombinación Genética
14.
J Clin Invest ; 124(3): 1350-63, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24569376

RESUMEN

Centronuclear myopathies (CNM) are congenital disorders associated with muscle weakness and abnormally located nuclei in skeletal muscle. An autosomal dominant form of CNM results from mutations in the gene encoding dynamin 2 (DNM2), and loss-of-function mutations in the gene encoding myotubularin (MTM1) result in X-linked CNM (XLCNM, also called myotubular myopathy), which promotes severe neonatal hypotonia and early death. Currently, no effective treatments exist for XLCNM. Here, we found increased DNM2 levels in XLCNM patients and a mouse model of XLCNM (Mtm1(-/y)). Generation of Mtm1(-/y) mice that were heterozygous for Dnm2 revealed that reduction of DNM2 in XLCNM mice restored life span, whole-body strength, and diaphragm function and increased muscle strength. Additionally, classic CNM-associated histological features, including fiber atrophy and nuclei mispositioning, were absent or reduced. Ultrastructural analysis revealed improvement of sarcomere organization and triad structures. Skeletal muscle-specific decrease of Dnm2 during embryogenesis or in young mice after disease onset revealed that the rescue associated with downregulation of Dnm2 is cell autonomous and is able to stop and potentially revert XLCNM progression. These data indicate that MTM1 and DNM2 regulate muscle organization and force through a common pathway. Furthermore, despite DNM2 being a key mechanoenzyme, its reduction is beneficial for XLCNM and represents a potential therapeutic approach for patients.


Asunto(s)
Dinamina II/genética , Expresión Génica , Miopatías Estructurales Congénitas/metabolismo , Animales , Diafragma/fisiopatología , Regulación hacia Abajo , Dinamina II/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Contracción Muscular , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Fenotipo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
15.
PLoS One ; 8(6): e67527, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826317

RESUMEN

Congenital myopathies are severe muscle disorders affecting adults as well as children in all populations. The diagnosis of congenital myopathies is constrained by strong clinical and genetic heterogeneity. Moreover, the majority of patients present with unspecific histological features, precluding purposive molecular diagnosis and demonstrating the need for an alternative and more efficient diagnostic approach. We used exome sequencing complemented by histological and ultrastructural analysis of muscle biopsies to identify the causative mutations in eight patients with clinically different skeletal muscle pathologies, ranging from a fatal neonatal myopathy to a mild and slowly progressive myopathy with adult onset. We identified RYR1 (ryanodine receptor) mutations in six patients and NEB (nebulin) mutations in two patients. We found novel missense and nonsense mutations, unraveled small insertions/deletions and confirmed their impact on splicing and mRNA/protein stability. Histological and ultrastructural findings of the muscle biopsies of the patients validated the exome sequencing results. We provide the evidence that an integrated strategy combining exome sequencing with clinical and histopathological investigations overcomes the limitations of the individual approaches to allow a fast and efficient diagnosis, accelerating the patient's access to a better healthcare and disease management. This is of particular interest for the diagnosis of congenital myopathies, which involve very large genes like RYR1 and NEB as well as genetic and phenotypic heterogeneity.


Asunto(s)
Enfermedades Musculares/congénito , Enfermedades Musculares/diagnóstico , Adulto , Secuencia de Bases , Biopsia , Análisis Mutacional de ADN , Exoma/genética , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Músculos/patología , Músculos/ultraestructura , Enfermedades Musculares/genética , Mutación/genética , Linaje , Fenotipo , Análisis de Secuencia de ADN
16.
FASEB J ; 27(8): 3384-94, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23695157

RESUMEN

Mutations in the phosphoinositide phosphatase myotubularin (MTM1) results in X-linked myotubular/centronuclear myopathy (XLMTM), characterized by a severe decrease in muscle mass and strength in patients and murine models. However, the molecular mechanism involved in the muscle hypotrophy is unclear. Here we show that the IGF1R/Akt pathway is affected in Mtm1-deficient murine muscles, characterized by an increase in IGF1 receptor and Akt levels in both the presymptomatic and symptomatic phases. Moreover, up-regulation of atrogenes was observed in the presymptomatic phase of the myopathy, supporting overactivation of the ubiquitin-proteasome pathway. In parallel, the autophagy machinery was affected as indicated by the increase in the number of autophagosomes and of autophagy markers, such as LC3 and P62. However, phosphorylation of FOXO3a and mTOR were abnormal at late but not at early stages of the disease, suggesting that myotubularin acts both upstream in the IGF1R/Akt pathway and downstream on the balance between the autophagy and ubiquitin-proteasome pathways in vivo. Adeno-associated virus-mediated delivery of Mtm1 into Mtm1-null muscles rescued muscle mass and normalized the expression levels of IGF1 receptor, the ubiquitin-proteasome pathway, and autophagy markers. These data support the hypothesis that the unbalanced regulation of the ubiquitin proteasome pathway and the autophagy machinery is a primary cause of the XLMTM pathogenesis.


Asunto(s)
Autofagia , Miopatías Estructurales Congénitas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia , Transducción de Señal , Ubiquitina/metabolismo , Animales , Western Blotting , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/genética , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR/metabolismo
17.
Eur J Hum Genet ; 21(6): 637-42, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23092955

RESUMEN

Heterozygous mutations in dynamin 2 (DNM2) have been linked to dominant Charcot-Marie-Tooth neuropathy and centronuclear myopathy. We report the first homozygous mutation in the DNM2 protein p.Phe379Val, in three consanguineous patients with a lethal congenital syndrome associating akinesia, joint contractures, hypotonia, skeletal abnormalities, and brain and retinal hemorrhages. In vitro membrane tubulation, trafficking and GTPase assays are consistent with an impact of the DNM2p.Phe379Val mutation on endocytosis. Although DNM2 has been previously implicated in axonal and muscle maintenance, the clinical manifestation in our patients taken together with our expression analysis profile during mouse embryogenesis and knockdown approaches in zebrafish resulting in defects in muscle organization and angiogenesis support a pleiotropic role for DNM2 during fetal development in vertebrates and humans.


Asunto(s)
Anomalías Congénitas/genética , Dinamina II/genética , Homocigoto , Mutación Missense/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Secuencia de Bases , Secuencia Conservada/genética , Análisis Mutacional de ADN , Dinamina II/química , Dinamina II/metabolismo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Humanos , Recién Nacido , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Embarazo , Síndrome
18.
Hum Mutat ; 33(6): 949-59, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22396310

RESUMEN

Centronuclear myopathy (CNM) is a genetically heterogeneous disorder associated with general skeletal muscle weakness, type I fiber predominance and atrophy, and abnormally centralized nuclei. Autosomal dominant CNM is due to mutations in the large GTPase dynamin 2 (DNM2), a mechanochemical enzyme regulating cytoskeleton and membrane trafficking in cells. To date, 40 families with CNM-related DNM2 mutations have been described, and here we report 60 additional families encompassing a broad genotypic and phenotypic spectrum. In total, 18 different mutations are reported in 100 families and our cohort harbors nine known and four new mutations, including the first splice-site mutation. Genotype-phenotype correlation hypotheses are drawn from the published and new data, and allow an efficient screening strategy for molecular diagnosis. In addition to CNM, dissimilar DNM2 mutations are associated with Charcot-Marie-Tooth (CMT) peripheral neuropathy (CMTD1B and CMT2M), suggesting a tissue-specific impact of the mutations. In this study, we discuss the possible clinical overlap of CNM and CMT, and the biological significance of the respective mutations based on the known functions of dynamin 2 and its protein structure. Defects in membrane trafficking due to DNM2 mutations potentially represent a common pathological mechanism in CNM and CMT.


Asunto(s)
Dinamina II/genética , Genes Dominantes , Estudios de Asociación Genética , Mutación , Miopatías Estructurales Congénitas/genética , Secuencia de Aminoácidos , Dinamina II/química , Humanos , Datos de Secuencia Molecular , Miopatías Estructurales Congénitas/diagnóstico , Polimorfismo Genético , Alineación de Secuencia
20.
Neuromuscul Disord ; 20(6): 375-81, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20434914

RESUMEN

X-linked centronuclear myopathy (XLMTM), also called myotubular myopathy, is a severe congenital myopathy characterized by generalized hypotonia and weakness at birth and the typical histological finding of centralization of myo-nuclei. It is caused by mutations in the MTM1 gene encoding the 3-phosphoinositides phosphatase myotubularin. Mutations in dynamin 2 and amphiphysin 2 genes lead to autosomal forms of centronuclear myopathy (CNM). While XLMTM is the most frequent and severe form of CNM, no mutations are found in about 30% of patients by sequencing all MTM1 exons. Moreover, the impact of MTM1 sequence variants is sometimes difficult to assess. It is thus important to devise a complete molecular diagnostic strategy that includes analysis of the myotubularin transcript and protein expression. We therefore developed novel antibodies against human myotubularin and showed that they are able to detect the endogenous protein by direct Western blot from muscle samples and from cultured cells. In conjunction with RT-PCR analysis we validated the consequences of missense and splice mutations on transcript integrity and protein level. We also detected and characterized a novel deep intronic mutation consisting of a single nucleotide change that induces exonisation of a conserved intronic sequence. Patients with centronuclear myopathy and no molecular diagnosis should be investigated for MTM1 defects at the cDNA and protein level.


Asunto(s)
Genes Ligados a X/genética , Intrones/genética , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/genética , Secuencia de Bases , Western Blotting , Células Cultivadas , ADN/genética , Exones/genética , Humanos , Datos de Secuencia Molecular , Mutación/genética , Mutación/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...