Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Nat Rev Mol Cell Biol ; 22(11): 713-732, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34257452

RESUMEN

Neuromuscular disorders comprise a diverse group of human inborn diseases that arise from defects in the structure and/or function of the muscle tissue - encompassing the muscle cells (myofibres) themselves and their extracellular matrix - or muscle fibre innervation. Since the identification in 1987 of the first genetic lesion associated with a neuromuscular disorder - mutations in dystrophin as an underlying cause of Duchenne muscular dystrophy - the field has made tremendous progress in understanding the genetic basis of these diseases, with pathogenic variants in more than 500 genes now identified as underlying causes of neuromuscular disorders. The subset of neuromuscular disorders that affect skeletal muscle are referred to as myopathies or muscular dystrophies, and are due to variants in genes encoding muscle proteins. Many of these proteins provide structural stability to the myofibres or function in regulating sarcolemmal integrity, whereas others are involved in protein turnover, intracellular trafficking, calcium handling and electrical excitability - processes that ensure myofibre resistance to stress and their primary activity in muscle contraction. In this Review, we discuss how defects in muscle proteins give rise to muscle dysfunction, and ultimately to disease, with a focus on pathologies that are most common, best understood and that provide the most insight into muscle biology.


Asunto(s)
Distrofina/genética , Proteínas Musculares/genética , Distrofia Muscular de Duchenne/genética , Enfermedades Neuromusculares/genética , Humanos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología , Mutación/genética , Enfermedades Neuromusculares/patología
2.
Gene Ther ; 30(10-11): 738-746, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37935854

RESUMEN

Despite the ups and downs in the field over three decades, the science of gene therapy has continued to advance and provide enduring treatments for increasing number of diseases. There are active clinical trials approaching a variety of inherited and acquired disorders of different organ systems. Approaches include ex vivo modification of hematologic stem cells (HSC), T lymphocytes and other immune cells, as well as in vivo delivery of genes or gene editing reagents to the relevant target cells by either local or systemic administration. In this article, we highlight success and ongoing challenges in three areas of high activity in gene therapy: inherited blood cell diseases by targeting hematopoietic stem cells, malignant disorders using immune effector cells genetically modified with chimeric antigen receptors, and ophthalmologic, neurologic, and coagulation disorders using in vivo administration of adeno-associated virus (AAV) vectors. In recent years, there have been true cures for many of these diseases, with sustained clinical benefit that exceed those from other medical approaches. Each of these treatments faces ongoing challenges, namely their high one-time costs and the complexity of manufacturing the therapeutic agents, which are biological viruses and cell products, at pharmacologic standards of quality and consistency. New models of reimbursement are needed to make these innovative treatments widely available to patients in need.


Asunto(s)
Terapia Genética , Neoplasias , Humanos , Linfocitos T , Células Madre Hematopoyéticas , Vectores Genéticos/genética , Dependovirus/genética , Edición Génica
3.
J Nanobiotechnology ; 21(1): 303, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37641124

RESUMEN

Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.


Asunto(s)
Distrofina , Músculo Esquelético , Animales , Ratones , Distribución Tisular , Ratones Endogámicos mdx , Regeneración
4.
PLoS Med ; 18(8): e1003734, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34464388

RESUMEN

BACKGROUND: The leading cause of mortality for patients with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome is the development of malignant peripheral nerve sheath tumor (MPNST), an aggressive soft tissue sarcoma. In the setting of NF1, this cancer type frequently arises from within its common and benign precursor, plexiform neurofibroma (PN). Transformation from PN to MPNST is challenging to diagnose due to difficulties in distinguishing cross-sectional imaging results and intralesional heterogeneity resulting in biopsy sampling errors. METHODS AND FINDINGS: This multi-institutional study from the National Cancer Institute and Washington University in St. Louis used fragment size analysis and ultra-low-pass whole genome sequencing (ULP-WGS) of plasma cell-free DNA (cfDNA) to distinguish between MPNST and PN in patients with NF1. Following in silico enrichment for short cfDNA fragments and copy number analysis to estimate the fraction of plasma cfDNA originating from tumor (tumor fraction), we developed a noninvasive classifier that differentiates MPNST from PN with 86% pretreatment accuracy (91% specificity, 75% sensitivity) and 89% accuracy on serial analysis (91% specificity, 83% sensitivity). Healthy controls without NF1 (participants = 16, plasma samples = 16), PN (participants = 23, plasma samples = 23), and MPNST (participants = 14, plasma samples = 46) cohorts showed significant differences in tumor fraction in plasma (P = 0.001) as well as cfDNA fragment length (P < 0.001) with MPNST samples harboring shorter fragments and being enriched for tumor-derived cfDNA relative to PN and healthy controls. No other covariates were significant on multivariate logistic regression. Mutational analysis demonstrated focal NF1 copy number loss in PN and MPNST patient plasma but not in healthy controls. Greater genomic instability including alterations associated with malignant transformation (focal copy number gains in chromosome arms 1q, 7p, 8q, 9q, and 17q; focal copy number losses in SUZ12, SMARCA2, CDKN2A/B, and chromosome arms 6p and 9p) was more prominently observed in MPNST plasma. Furthermore, the sum of longest tumor diameters (SLD) visualized by cross-sectional imaging correlated significantly with paired tumor fractions in plasma from MPNST patients (r = 0.39, P = 0.024). On serial analysis, tumor fraction levels in plasma dynamically correlated with treatment response to therapy and minimal residual disease (MRD) detection before relapse. Study limitations include a modest MPNST sample size despite accrual from 2 major referral centers for this rare malignancy, and lack of uniform treatment and imaging protocols representing a real-world cohort. CONCLUSIONS: Tumor fraction levels derived from cfDNA fragment size and copy number alteration analysis of plasma cfDNA using ULP-WGS significantly correlated with MPNST tumor burden, accurately distinguished MPNST from its benign PN precursor, and dynamically correlated with treatment response. In the future, our findings could form the basis for improved early cancer detection and monitoring in high-risk cancer-predisposed populations.


Asunto(s)
Ácidos Nucleicos Libres de Células/análisis , Neurofibroma Plexiforme/diagnóstico , Neurofibrosarcoma/diagnóstico , Secuenciación Completa del Genoma , Adulto , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Hum Mol Genet ; 28(20): 3431-3442, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31411676

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by mutations in the gene encoding dystrophin. Prior work has shown that DMD progression can vary, depending on the genetic makeup of the patient. Several modifier alleles have been identified including LTBP4 and SPP1. We previously showed that Spp1 exacerbates the DMD phenotype in the mdx mouse model by promoting fibrosis and by skewing macrophage polarization. Here, we studied the mechanisms involved in Spp1's promotion of fibrosis by using both isolated fibroblasts and genetically modified mice. We found that Spp1 upregulates collagen expression in mdx fibroblasts by enhancing TGFß signaling. Spp1's effects on TGFß signaling are through induction of MMP9 expression. MMP9 is a protease that can release active TGFß ligand from its latent complex. In support for activation of this pathway in our model, we showed that treatment of mdx fibroblasts with MMP9 inhibitor led to accumulation of the TGFß latent complex, decreased levels of active TGFß and reduced collagen expression. Correspondingly, we found reduced active TGFß in Spp1-/-mdxB10 and Mmp9-/-mdxB10 muscles in vivo. Taken together with previous observations of reduced fibrosis in both models, these data suggest that Spp1 acts upstream of TGFß to promote fibrosis in mdx muscles. We found that in the context of constitutively upregulated TGFß signaling (such as in the mdxD2 model), ablation of Spp1 has very little effect on fibrosis. Finally, we performed proof-of-concept studies showing that postnatal pharmacological inhibition of Spp1 reduces fibrosis and improves muscle function in mdx mice.


Asunto(s)
Fibrosis/genética , Distrofia Muscular de Duchenne/metabolismo , Osteopontina/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Colágeno Tipo I/biosíntesis , Modelos Animales de Enfermedad , Distrofina/genética , Distrofina/metabolismo , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Fibrosis/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Osteopontina/metabolismo , Cultivo Primario de Células , Regeneración/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/genética
6.
J Physiol ; 598(18): 3927-3939, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33460149

RESUMEN

KEY POINTS: Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. The effect of genetic and pharmacological inhibition of myostatin signalling on the disease phenotype in a mouse model of LGMD R1 (CAPN3 knockout mouse-C3KO) was studied. Inhibition of myostatin signalling in C3KO muscles resulted in significant muscle hypertrophy; however, there were no improvements in muscle strength and exacerbation of exercise intolerance concomitant with further reduction of muscle oxidative capacity was observed. Inhibition of myostatin signalling is unlikely to be a valid therapeutic strategy for LGMD R1. ABSTRACT: Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. There are currently no therapies available to patients. We sought to determine if induction of muscle growth, through myostatin inhibition, represents a viable therapeutic strategy for this disease. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. However, the effect of myostatin depends on the genetic and pathophysiological context and may not be efficacious in all contexts. We found that genetic inhibition of myostatin through overexpression of follistatin (an endogenous inhibitor of myostatin) in our LGMD R1 model (C3KO) resulted in 1.5- to 2-fold increase of muscle mass for the majority of limb muscles. However, muscle strength was not improved and exercise intolerance was exacerbated. Pharmacological inhibition of myostatin, using an anti-myostatin antibody, resulted in statistically significant increases in muscle mass; however, functional testing did not reveal changes in muscle strength nor endurance in treated C3KO mice. Histochemical and biochemical evaluation of follistatin overexpressing mice revealed a reduction in the percentage of oxidative fibres and decreased activation of AMP-activated protein kinase signalling in transgenics compared to C3KO muscles. Our data suggest that muscle hypertrophy, induced by myostatin inhibition, leads to loss of oxidative capacity, which further compromises metabolically impaired C3KO muscles and thus is unlikely to be a valid strategy for treatment of LGMD R1.


Asunto(s)
Distrofia Muscular de Cinturas , Miostatina , Proteínas Quinasas Activadas por AMP , Animales , Calpaína , Tolerancia al Ejercicio , Humanos , Hipertrofia , Ratones , Proteínas Musculares , Músculo Esquelético , Distrofia Muscular de Cinturas/genética , Miostatina/genética
7.
J Biol Chem ; 294(10): 3501-3513, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30602569

RESUMEN

Mutations in superoxide dismutase 1 (SOD1) cause 15-20% of familial amyotrophic lateral sclerosis (fALS) cases. The resulting amino acid substitutions destabilize SOD1's protein structure, leading to its self-assembly into neurotoxic oligomers and aggregates, a process hypothesized to cause the characteristic motor-neuron degeneration in affected individuals. Currently, effective disease-modifying therapy is not available for ALS. Molecular tweezers prevent formation of toxic protein assemblies, yet their protective action has not been tested previously on SOD1 or in the context of ALS. Here, we tested the molecular tweezer CLR01-a broad-spectrum inhibitor of the self-assembly and toxicity of amyloid proteins-as a potential therapeutic agent for ALS. Using recombinant WT and mutant SOD1, we found that CLR01 inhibited the aggregation of all tested SOD1 forms in vitro Next, we examined whether CLR01 could prevent the formation of misfolded SOD1 in the G93A-SOD1 mouse model of ALS and whether such inhibition would have a beneficial therapeutic effect. CLR01 treatment decreased misfolded SOD1 in the spinal cord significantly. However, these histological findings did not correlate with improvement of the disease phenotype. A small, dose-dependent decrease in disease duration was found in CLR01-treated mice, relative to vehicle-treated animals, yet motor function did not improve in any of the treatment groups. These results demonstrate that CLR01 can inhibit SOD1 misfolding and aggregation both in vitro and in vivo, but raise the question whether such inhibition is sufficient for achieving a therapeutic effect. Additional studies in other less aggressive ALS models may be needed to determine the therapeutic potential of this approach.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Hidrocarburos Aromáticos con Puentes/farmacología , Mutación , Organofosfatos/farmacología , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/genética , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Sitios de Unión , Peso Corporal/efectos de los fármacos , Hidrocarburos Aromáticos con Puentes/metabolismo , Modelos Animales de Enfermedad , Ratones , Fuerza Muscular/efectos de los fármacos , Organofosfatos/metabolismo , Agregado de Proteínas/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Superóxido Dismutasa-1/metabolismo , Análisis de Supervivencia
8.
Physiology (Bethesda) ; 34(5): 341-353, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31389773

RESUMEN

This is a review describing advances in CRISPR/Cas-mediated therapies for neuromuscular disorders (NMDs). We explore both CRISPR-mediated editing and dead Cas approaches as potential therapeutic strategies for multiple NMDs. Last, therapeutic considerations, including delivery and off-target effects, are also discussed.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Enfermedades Neuromusculares/genética , Animales , Edición Génica/métodos , Humanos
9.
Hum Mol Genet ; 27(9): 1642-1653, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29528394

RESUMEN

Mutations in CAPN3 cause autosomal recessive limb girdle muscular dystrophy 2A. Calpain 3 (CAPN3) is a calcium dependent protease residing in the myofibrillar, cytosolic and triad fractions of skeletal muscle. At the triad, it colocalizes with calcium calmodulin kinase IIß (CaMKIIß). CAPN3 knock out mice (C3KO) show reduced triad integrity and blunted CaMKIIß signaling, which correlates with impaired transcriptional activation of myofibrillar and oxidative metabolism genes in response to running exercise. These data suggest a role for CAPN3 and CaMKIIß in gene regulation that takes place during adaptation to endurance exercise. To assess whether CAPN3- CaMKIIß signaling influences skeletal muscle remodeling in other contexts, we subjected C3KO and wild type mice to hindlimb unloading and reloading and assessed CaMKIIß signaling and gene expression by RNA-sequencing. After induced atrophy followed by 4 days of reloading, both CaMKIIß activation and expression of inflammatory and cellular stress genes were increased. C3KO muscles failed to activate CaMKIIß signaling, did not activate the same pattern of gene expression and demonstrated impaired growth at 4 days of reloading. Moreover, C3KO muscles failed to activate inducible HSP70, which was previously shown to be indispensible for the inflammatory response needed to promote muscle recovery. Likewise, C3KO showed diminished immune cell infiltration and decreased expression of pro-myogenic genes. These data support a role for CaMKIIß signaling in induction of HSP70 and promotion of the inflammatory response during muscle growth and remodeling that occurs after atrophy, suggesting that CaMKIIß regulates remodeling in multiple contexts: endurance exercise and growth after atrophy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calpaína/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calpaína/genética , Línea Celular , Proteínas HSP70 de Choque Térmico/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética
10.
PLoS Genet ; 13(10): e1007070, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29065150

RESUMEN

Genetic disruption of the dystrophin complex produces muscular dystrophy characterized by a fragile muscle plasma membrane leading to excessive muscle degeneration. Two genetic modifiers of Duchenne Muscular Dystrophy implicate the transforming growth factor ß (TGFß) pathway, osteopontin encoded by the SPP1 gene and latent TGFß binding protein 4 (LTBP4). We now evaluated the functional effect of these modifiers in the context of muscle injury and repair to elucidate their mechanisms of action. We found that excess osteopontin exacerbated sarcolemmal injury, and correspondingly, that loss of osteopontin reduced injury extent both in isolated myofibers and in muscle in vivo. We found that ablation of osteopontin was associated with reduced expression of TGFß and TGFß-associated pathways. We identified that increased TGFß resulted in reduced expression of Anxa1 and Anxa6, genes encoding key components of the muscle sarcolemma resealing process. Genetic manipulation of Ltbp4 in dystrophic muscle also directly modulated sarcolemmal resealing, and Ltbp4 alleles acted in concert with Anxa6, a distinct modifier of muscular dystrophy. These data provide a model in which a feed forward loop of TGFß and osteopontin directly impacts the capacity of muscle to recover from injury, and identifies an intersection of genetic modifiers on muscular dystrophy.


Asunto(s)
Genes Modificadores , Proteínas de Unión a TGF-beta Latente/fisiología , Músculo Esquelético/fisiología , Distrofia Muscular Animal/genética , Osteopontina/metabolismo , Animales , Anexina A1/genética , Anexina A1/metabolismo , Anexina A6/genética , Anexina A6/metabolismo , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Músculo Esquelético/lesiones , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Osteopontina/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Recuperación de la Función , Sarcolema/fisiología
11.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 572-579, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28011285

RESUMEN

Muscular dystrophies are genetic conditions leading to muscle degeneration and often, impaired regeneration. Duchenne Muscular Dystrophy is a prototypical form of muscular dystrophy, and like other forms of genetically inherited muscle diseases, pathological progression is variable. Variability in muscular dystrophy can arise from differences in the manner in which the primary mutation impacts the affected protein's function; however, clinical heterogeneity also derives from secondary mutations in other genes that can enhance or reduce pathogenic features of disease. These genes, called genetic modifiers, regulate the pathophysiological context of dystrophic degeneration and regeneration. Understanding the mechanistic links between genetic modifiers and dystrophic progression sheds light on pathologic remodeling, and provides novel avenues to therapeutically intervene to reduce muscle degeneration. Based on targeted genetic approaches and unbiased genomewide screens, several modifiers have been identified for muscular dystrophy, including extracellular agonists of signaling cascades. This review will focus on identification and possible mechanisms of recently identified modifiers for muscular dystrophy, including osteopontin, latent TGFß binding protein 4 (LTBP4) and Jagged1. Moreover, we will review the investigational approaches that aim to target modifier pathways and thereby counteract dystrophic muscle wasting.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Proteína Jagged-1/genética , Proteínas de Unión a TGF-beta Latente/genética , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Osteopontina/genética , Animales , Anticuerpos Monoclonales/uso terapéutico , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Drogas en Investigación/uso terapéutico , Marcadores Genéticos , Humanos , Proteína Jagged-1/agonistas , Proteína Jagged-1/metabolismo , Proteínas de Unión a TGF-beta Latente/agonistas , Proteínas de Unión a TGF-beta Latente/metabolismo , Terapia Molecular Dirigida , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Osteopontina/antagonistas & inhibidores , Osteopontina/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal
12.
Hum Mol Genet ; 25(11): 2194-2207, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27005420

RESUMEN

Limb girdle muscular dystrophy 2A is due to loss-of-function mutations in the Calpain 3 (CAPN3) gene. Our previous data suggest that CAPN3 helps to maintain the integrity of the triad complex in skeletal muscle. In Capn3 knock-out mice (C3KO), Ca2+ release and Ca2+/calmodulin kinase II (CaMKII) signaling are attenuated. We hypothesized that calpainopathy may result from a failure to transmit loading-induced Ca2+-mediated signals, necessary to up-regulate expression of muscle adaptation genes. To test this hypothesis, we compared transcriptomes of muscles from wild type (WT) and C3KO mice subjected to endurance exercise. In WT mice, exercise induces a gene signature that includes myofibrillar, mitochondrial and oxidative lipid metabolism genes, necessary for muscle adaptation. C3KO muscles fail to activate the same gene signature. Furthermore, in agreement with the aberrant transcriptional profile, we observe a commensurate functional defect in lipid metabolism whereby C3KO muscles fail to release fatty acids from stored triacylglycerol. In conjunction with the defects in oxidative metabolism, C3KO mice demonstrate reduced exercise endurance. Failure to up-regulate genes in C3KO muscles is due, in part, to decreased levels of PGC1α, a transcriptional co-regulator that orchestrates the muscle adaptation response. Destabilization of PGC1α is attributable to decreased p38 MAPK activation via diminished CaMKII signaling. Thus, we elucidate a pathway downstream of Ca2+-mediated CaMKII activation that is dysfunctional in C3KO mice, leading to reduced transcription of genes involved in muscle adaptation. These studies identify a novel mechanism of muscular dystrophy: a blunted transcriptional response to muscle loading resulting in chronic failure to adapt and remodel.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calpaína/genética , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Animales , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Calpaína/biosíntesis , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Mutación , Estrés Oxidativo/genética , Activación Transcripcional/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
13.
Hum Mol Genet ; 25(24): 5395-5406, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798107

RESUMEN

Duchenne muscular dystrophy (DMD) is a genetic disorder that causes progressive muscle weakness, ultimately leading to early mortality in affected teenagers and young adults. Previous work from our lab has shown that a small transmembrane protein called sarcospan (SSPN) can enhance the recruitment of adhesion complex proteins to the cell surface. When human SSPN is expressed at three-fold levels in mdx mice, this increase in adhesion complex abundance improves muscle membrane stability, preventing many of the histopathological changes associated with DMD. However, expressing higher levels of human SSPN (ten-fold transgenic expression) causes a severe degenerative muscle phenotype in wild-type mice. Since SSPN-mediated stabilization of the sarcolemma represents a promising therapeutic strategy in DMD, it is important to determine whether SSPN can be introduced at high levels without toxicity. Here, we show that mouse SSPN (mSSPN) can be overexpressed at 30-fold levels in wild-type mice with no deleterious effects. In mdx mice, mSSPN overexpression improves dystrophic pathology and sarcolemmal stability. We show that these mice exhibit increased resistance to eccentric contraction-induced damage and reduced fatigue following exercise. mSSPN overexpression improved pulmonary function and reduced dystrophic histopathology in the diaphragm. Together, these results demonstrate that SSPN overexpression is well tolerated in mdx mice and improves sarcolemma defects that underlie skeletal muscle and pulmonary dysfunction in DMD.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Distrofia Muscular de Duchenne/genética , Proteínas de Neoplasias/genética , Sarcolema/genética , Animales , Proteínas Portadoras/biosíntesis , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Contracción Muscular/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Proteínas de Neoplasias/biosíntesis , Sarcolema/patología
14.
Hum Mol Genet ; 24(10): 2873-83, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25701873

RESUMEN

Limb girdle muscular dystrophy 2H is caused by mutations in the gene encoding the E3 ubiquitin ligase, TRIM32. Previously, we generated and characterized a Trim32 knockout mouse (T32KO) that displays both neurogenic and myopathic features. The myopathy in these mice is attributable to impaired muscle growth, associated with satellite cell senescence and premature sarcopenia. This satellite cell senescence is due to accumulation of the SUMO ligase PIASy, a substrate of TRIM32. The goal of this investigation was to identify additional substrates of TRIM32 using 2D fluorescence difference gel electrophoresis (2D-DIGE) in order to further explore its role in skeletal muscle. Because TRIM32 is an E3 ubiquitin ligase, we reasoned that TRIM32's substrates would accumulate in its absence. 2D-DIGE identified 19 proteins that accumulate in muscles from the T32KO mouse. We focused on two of these proteins, NDRG2 and TRIM72, due to their putative roles in myoblast proliferation and myogenesis. Follow-up analysis confirmed that both proteins were ubiquitinated by TRIM32 in vitro; however, only NDRG2 accumulated in skeletal muscle and myoblasts in the absence of TRIM32. NDRG2 overexpression in myoblasts led to reduced cell proliferation and delayed cell cycle withdrawal during differentiation. Thus, we identified NDRG2 as a novel target for TRIM32; these findings further corroborate the hypothesis that TRIM32 is involved in control of myogenic cells proliferation and differentiation.


Asunto(s)
Proliferación Celular , Distrofia Muscular de Cinturas/genética , Mioblastos/enzimología , Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Ciclo Celular , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Distrofia Muscular de Cinturas/enzimología , Mioblastos/fisiología , Proteínas/genética , Electroforesis Bidimensional Diferencial en Gel , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
15.
J Biol Chem ; 290(2): 996-1004, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25389288

RESUMEN

Calpains are broadly distributed, calcium-dependent enzymes that induce limited proteolysis in a wide range of substrates. Mutations in the gene encoding the muscle-specific family member calpain 3 (CAPN3) underlie limb-girdle muscular dystrophy 2A. We have shown previously that CAPN3 knockout muscles exhibit attenuated calcium release, reduced calmodulin kinase (CaMKII) signaling, and impaired muscle adaptation to exercise. However, neither the precise role of CAPN3 in these processes nor the mechanisms of CAPN3 activation in vivo have been fully elucidated. In this study, we identify calmodulin (CaM), a known transducer of the calcium signal, as the first positive regulator of CAPN3 autolytic activity. CaM was shown to bind CAPN3 at two sites located in the C2L domain. Biochemical studies using muscle extracts from transgenic mice overexpressing CAPN3 or its inactive mutant revealed that CaM binding enhanced CAPN3 autolytic activation. Furthermore, CaM facilitated CAPN3-mediated cleavage of its in vivo substrate titin in tissue extracts. Therefore, these studies reveal a novel interaction between CAPN3 and CaM and identify CaM as the first positive regulator of CAPN3 activity.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calmodulina/genética , Calpaína/genética , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/genética , Proteolisis , Animales , Señalización del Calcio/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/biosíntesis , Calmodulina/metabolismo , Calpaína/biosíntesis , Humanos , Ratones , Ratones Transgénicos , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología , Mutación
16.
J Health Econ ; 95: 102867, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38461677

RESUMEN

Behavioral adjustments to mitigate increasing risk of STIs can increase or decrease the likelihood of pregnancy. This paper measures the effects of the arrival and spread of AIDS across U.S. cities in the 1980s and 1990s on births and abortions. I show that the AIDS epidemic increased the birth rate by 0.55 percent and the abortion rate by 1.77 percent. I find support for two underlying mechanisms to explain the increase in pregnancies. Some women opted into monogamous partnerships in response to the AIDS epidemic, with a corresponding increase in the marriage rate and improvement in infant health. Others switched from prescription contraceptive methods to condoms. These behavioral changes lowered the incidence of other sexually transmitted infections, but increased both planned and unplanned pregnancies.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida , Tasa de Natalidad , Sexo Seguro , Humanos , Femenino , Embarazo , Tasa de Natalidad/tendencias , Estados Unidos/epidemiología , Síndrome de Inmunodeficiencia Adquirida/epidemiología , Síndrome de Inmunodeficiencia Adquirida/prevención & control , Adulto , Aborto Inducido/estadística & datos numéricos , Adulto Joven , Adolescente , Conducta Sexual , Condones/estadística & datos numéricos , Enfermedades de Transmisión Sexual/epidemiología , Enfermedades de Transmisión Sexual/prevención & control
17.
JCI Insight ; 9(3)2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38175727

RESUMEN

The Murphy Roths Large (MRL) mouse strain has "super-healing" properties that enhance recovery from injury. In mice, the DBA/2J strain intensifies many aspects of muscular dystrophy, so we evaluated the ability of the MRL strain to suppress muscular dystrophy in the Sgcg-null mouse model of limb girdle muscular dystrophy. A comparative analysis of Sgcg-null mice in the DBA/2J versus MRL strains showed greater myofiber regeneration, with reduced structural degradation of muscle in the MRL strain. Transcriptomic profiling of dystrophic muscle indicated strain-dependent expression of extracellular matrix (ECM) and TGF-ß signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized myoscaffolds. Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-ß1 and TGF-ß3 throughout the matrix. Dystrophic myoscaffolds from the MRL background, but not the DBA/2J background, were enriched in myokines like IGF-1 and IL-6. C2C12 myoblasts seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J muscles showed the MRL background induced greater myoblast differentiation compared with dystrophic DBA/2J myoscaffolds. Thus, the MRL background imparts its effect through a highly regenerative ECM, which is active even in muscular dystrophy.


Asunto(s)
Distrofia Muscular de Cinturas , Distrofias Musculares , Ratones , Animales , Ratones Endogámicos DBA , Distrofias Musculares/genética , Músculos , Matriz Extracelular , Ratones Noqueados
18.
bioRxiv ; 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38765987

RESUMEN

Introduction: Limb girdle muscular dystrophies (LGMDs) are a group of genetically heterogeneous autosomal conditions with some degree of phenotypic homogeneity. LGMD is defined as having onset >2 years of age with progressive proximal weakness, elevated serum creatine kinase levels and dystrophic features on muscle biopsy. Advances in massively parallel sequencing have led to a surge in genes linked to LGMD. Methods: The ClinGen Muscular Dystrophies and Myopathies gene curation expert panel (MDM GCEP, formerly Limb Girdle Muscular Dystrophy GCEP) convened to evaluate the strength of evidence supporting gene-disease relationships (GDR) using the ClinGen gene-disease clinical validity framework to evaluate 31 genes implicated in LGMD. Results: The GDR was exclusively LGMD for 17 genes, whereas an additional 14 genes were related to a broader phenotype encompassing congenital weakness. Four genes (CAPN3, COL6A1, COL6A2, COL6A3) were split into two separate disease entities, based on each displaying both dominant and recessive inheritance patterns, resulting in curation of 35 GDRs. Of these, 30 (86%) were classified as Definitive, 4 (11%) as Moderate and 1 (3%) as Limited. Two genes, POMGNT1 and DAG1, though definitively related to myopathy, currently have insufficient evidence to support a relationship specifically with LGMD. Conclusions: The expert-reviewed assertions on the clinical validity of genes implicated in LGMDs form an invaluable resource for clinicians and molecular geneticists. We encourage the global neuromuscular community to publish case-level data that help clarify disputed or novel LGMD associations.

19.
Hum Mol Genet ; 20(20): 3925-32, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21775502

RESUMEN

Mutations in tripartite motif protein 32 (TRIM32) are responsible for several hereditary disorders that include limb girdle muscular dystrophy type 2H (LGMD2H), sarcotubular myopathy (STM) and Bardet Biedl syndrome. Most LGMD2H mutations in TRIM32 are clustered in the NHL ß-propeller domain at the C-terminus and are predicted to interfere with homodimerization. To get insight into TRIM32's role in the pathogenesis of LGMD2H and to create an accurate model of disease, we have generated a knock-in mouse (T32KI) carrying the c.1465G > A (p.D489N) mutation in murine Trim32 corresponding to the human LGMD2H/STM pathogenic mutation c.1459G > A (p.D487N). Our data indicate that T32KI mice have both a myopathic and a neurogenic phenotype, very similar to the one described in the Trim32-null mice that we created previously. Analysis of Trim32 gene expression in T32KI mice revealed normal mRNA levels, but a severe reduction in mutant TRIM32 (D489N) at the protein level. Our results suggest that the D489N pathogenic mutation destabilizes the protein, leading to its degradation, and results in the same mild myopathic and neurogenic phenotype as that found in Trim32-null mice. Thus, one potential mechanism of LGMD2H might be destabilization of mutated TRIM32 protein leading to a null phenotype.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Mutación Missense , Fenotipo , Ubiquitina-Proteína Ligasas/genética , Animales , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Marcación de Gen , Ratones , Ratones Transgénicos , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Miosinas/metabolismo , Isoformas de Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
20.
Hum Mol Genet ; 20(17): 3331-45, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21624972

RESUMEN

Calpain 3 (CAPN3) is a muscle-specific, calcium-dependent proteinase that is mutated in Limb Girdle Muscle Dystrophy type 2A. Most pathogenic missense mutations in LGMD2A affect CAPN3's proteolytic activity; however, two mutations, D705G and R448H, retain activity but nevertheless cause muscular dystrophy. Previously, we showed that D705G and R448H mutations reduce CAPN3s ability to bind to titin in vitro. In this investigation, we tested the consequence of loss of titin binding in vivo and examined whether this loss can be an underlying pathogenic mechanism in LGMD2A. To address this question, we created transgenic mice that express R448H or D705G in muscles, on wild-type (WT) CAPN3 or knock-out background. Both mutants were readily expressed in insect cells, but when D705G was expressed in skeletal muscle, it was not stable enough to study. Moreover, the D705G mutation had a dominant negative effect on endogenous CAPN3 when expressed on a WT background. The R448H protein was stably expressed in muscles; however, it was more rapidly degraded in muscle extracts compared with WT CAPN3. Increased degradation of R448H was due to non-cysteine, cellular proteases acting on the autolytic sites of CAPN3, rather than autolysis. Fractionation experiments revealed a significant decrease of R448H from the myofibrillar fraction, likely due to the mutant's inability to bind titin. Our data suggest that R448H and D705G mutations affect both CAPN3s anchorage to titin and its stability. These studies reveal a novel mechanism by which mutations that spare enzymatic activity can still lead to calpainopathy.


Asunto(s)
Calpaína/genética , Distrofia Muscular de Cinturas/genética , Miofibrillas/metabolismo , Animales , Western Blotting , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Mutación , Mutación Missense , Miofibrillas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda