Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(13): 1107-1119, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38507070

RESUMEN

The dystrophin-glycoprotein complex (DGC) plays a crucial role in maintaining the structural integrity of the plasma membrane and the neuromuscular junction. In this study, we investigated the impact of the deficiency of α-dystrobrevin (αdbn), a component of the DGC, on the homeostasis of intracellular organelles, specifically mitochondria and the sarcoplasmic reticulum (SR). In αdbn deficient muscles, we observed a significant increase in the membrane-bound ATP synthase complex levels, a marker for mitochondria in oxidative muscle fiber types compared to wild-type. Furthermore, examination of muscle fibers deficient in αdbn using electron microscopy revealed profound alterations in the organization of mitochondria and the SR within certain myofibrils of muscle fibers. This included the formation of hyper-branched intermyofibrillar mitochondria with extended connections, an extensive network spanning several myofibrils, and a substantial increase in the number/density of subsarcolemmal mitochondria. Concurrently, in some cases, we observed significant structural alterations in mitochondria, such as cristae loss, fragmentation, swelling, and the formation of vacuoles and inclusions within the mitochondrial matrix cristae. Muscles deficient in αdbn also displayed notable alterations in the morphology of the SR, along with the formation of distinct anomalous concentric SR structures known as whorls. These whorls were prevalent in αdbn-deficient mice but were absent in wild-type muscles. These results suggest a crucial role of the DGC αdbn in regulating intracellular organelles, particularly mitochondria and the SR, within muscle cells. The remodeling of the SR and the formation of whorls may represent a novel mechanism of the unfolded protein response (UPR) in muscle cells.


Asunto(s)
Proteínas Asociadas a la Distrofina , Distrofina , Mitocondrias , Retículo Sarcoplasmático , Animales , Ratones , Distrofina/genética , Distrofina/metabolismo , Distrofina/deficiencia , Proteínas Asociadas a la Distrofina/genética , Proteínas Asociadas a la Distrofina/metabolismo , Glicoproteínas/metabolismo , Glicoproteínas/genética , Glicoproteínas/deficiencia , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Mitocondrias/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Miofibrillas/metabolismo , Miofibrillas/ultraestructura , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/ultraestructura
2.
BMC Musculoskelet Disord ; 25(1): 146, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365661

RESUMEN

BACKGROUND: Dysferlinopathy is a phenotypically heterogeneous group of hereditary diseases caused by mutations in the DYSF gene. Early contractures are considered rare, and rigid spine syndrome in dysferlinopathy has been previously reported only once. CASE PRESENTATION: We describe a 23-year-old patient with Miyoshi myopathy with a rigid spine and multiple contractures, a rare phenotypic variant. The disease first manifested when the patient was 13 years old, with fatigue of the gastrocnemius muscles and the development of pronounced contractures of the Achilles tendons, flexors of the fingers, and extensors of the toes, followed by the involvement of large joints and the spine. Magnetic resonance imaging revealed signs of connective tissue and fatty replacement of the posterior muscles of the thighs and lower legs. Edema was noted in the anterior and medial muscle groups of the thighs, lower legs, and the multifidus muscle of the back. Whole genome sequencing revealed previously described mutations in the DYSF gene in exon 39 (c.4282 C > T) and intron 51 (c.5785-824 C > T). An immunohistochemical analysis and Western blot showed the complete absence of dysferlin protein expression in the muscle fibers. CONCLUSIONS: This case expands the range of clinical and phenotypic correlations of dysferlinopathy and complements the diagnostic search for spine rigidity.


Asunto(s)
Contractura , Miopatías Distales , Atrofia Muscular , Distrofia Muscular de Cinturas , Humanos , Adolescente , Adulto Joven , Adulto , Proteínas de la Membrana/genética , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/diagnóstico por imagen , Distrofia Muscular de Cinturas/genética , Mutación , Contractura/etiología , Contractura/genética
3.
NMR Biomed ; : e4947, 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37021657

RESUMEN

MRI's T2 relaxation time is a valuable biomarker for neuromuscular disorders and muscle dystrophies. One of the hallmarks of these pathologies is the infiltration of adipose tissue and a loss of muscle volume. This leads to a mixture of two signal components, from fat and from water, to appear in each imaged voxel, each having a specific T2 relaxation time. In this proof-of-concept work, we present a technique that can separate the signals from water and from fat within each voxel, measure their separate T2 values, and calculate their relative fractions. The echo modulation curve (EMC) algorithm is a dictionary-based technique that offers accurate and reproducible mapping of T2 relaxation times. We present an extension of the EMC algorithm for estimating subvoxel fat and water fractions, alongside the T2 and proton-density values of each component. To facilitate data processing, calf and thigh anatomy were automatically segmented using a fully convolutional neural network and FSLeyes software. The preprocessing included creating two signal dictionaries, for water and for fat, using Bloch simulations of the prospective protocol. Postprocessing included voxelwise fitting for two components, by matching the experimental decay curve to a linear combination of the two simulated dictionaries. Subvoxel fat and water fractions and relaxation times were generated and used to calculate a new quantitative biomarker, termed viable muscle index, and reflecting disease severity. This biomarker indicates the fraction of remaining muscle out of the entire muscle region. The results were compared with those using the conventional Dixon technique, showing high agreement (R = 0.98, p < 0.001). It was concluded that the new extension of the EMC algorithm can be used to quantify abnormal fat infiltration as well as identify early inflammatory processes corresponding to elevation in the T2 value of the water (muscle) component. This new ability may improve the diagnostic accuracy of neuromuscular diseases, help stratification of patients according to disease severity, and offer an efficient tool for tracking disease progression.

4.
Int J Mol Sci ; 24(24)2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38139231

RESUMEN

Limb girdle muscular dystrophies (LGMDs) are a group of genetically inherited neuromuscular diseases with a very variable clinical presentation and overlapping traits. Over the last few years there has been an increasing interest in the use of non-invasive circulating biomarkers to monitor disease progression and to evaluate the efficacy of therapeutic approaches. Our aim was to identify the miRNA signature with potential value for LGMD patient screening and stratification. Using miRCURY LNA miRNA qPCR Serum/Plasma Panel, we analyzed 179 miRNAs from 16 patients, divided in four pools based on their genetic diagnosis, and from healthy controls. The miRNAs analysis showed a total of 107 dysregulated miRNAs in LGMD patients when compared to the healthy controls. After filtering via skeletal tissue expression and gene/pathways target analysis, the number of dysregulated miRNAs drastically reduced. Six selected miRNAs-let-7f-5p (in LGMDR1), miR-20a-5p (in LGMDR2), miR-130b-5p, miR-378a-5p (both in LGMDR3), miR-376c-3p and miR-382-5p (both in LGMDR4)-whose expression was significantly lower compared to controls in the different LGMD pools, were further investigated. The bioinformatic analysis of the target genes in each selected miRNA revealed ECM-receptor interaction and TGF-beta signaling as the most involved pathways. The correlation analysis showed a good correlation of let-7f-5p with fibrosis and with the cross sectional area of type I and type II fibers, while miR-130b-5p showed a good correlation with the age of onset of the disease. The receiver operating characteristic curves showed how single miRNAs were able to discriminate a specific group of LGMD patients and how the combination of six miRNAs was able to discriminate LGMD patients from controls.


Asunto(s)
MicroARNs , Distrofia Muscular de Cinturas , Humanos , MicroARNs/genética , Perfilación de la Expresión Génica , Biomarcadores , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/genética , Curva ROC
5.
Mol Genet Metab ; 136(3): 226-237, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35660068

RESUMEN

Popeye domain containing protein 1 (POPDC1) is a highly conserved transmembrane protein essential for striated muscle function and homeostasis. Pathogenic variants in the gene encoding POPDC1 (BVES, Blood vessel epicardial substance) are causative for limb-girdle muscular dystrophy (LGMDR25), associated with cardiac arrhythmia. We report on four affected children (age 7-19 years) from two consanguineous families with two novel pathogenic variants in BVES c.457C>T(p.Q153X) and c.578T>G (p.I193S). Detailed analyses were performed on muscle biopsies from an affected patient of each family including immunofluorescence, electron microscopy and proteomic profiling. Cardiac abnormalities were present in all patients and serum creatine kinase (CK) values were variably elevated despite lack of overt muscle weakness. Detailed histological analysis of skeletal muscle, however indicated a myopathy with reduced sarcolemmal expression of POPDC1 accompanied by altered sarcolemmal and sarcoplasmatic dysferlin and Xin/XIRP1 abundance. At the electron microscopic level, the muscle fiber membrane was focally disrupted. The proteomic signature showed statistically significant dysregulation of 191 proteins of which 173 were increased and 18 were decreased. Gene ontology-term analysis of affected biological processes revealed - among others - perturbation of muscle fibril assembly, myofilament sliding, and contraction as well as transition between fast and slow fibers. In conclusion, these findings demonstrate that the phenotype of LGMDR25 is highly variable and also includes younger children with conduction abnormalities, no apparent muscular problems, and only mildly elevated CK values. Biochemical studies suggest that BVES mutations causing loss of functional POPDC1 can impede striated muscle function by several mechanisms.


Asunto(s)
Proteínas Musculares , Distrofia Muscular de Cinturas , Moléculas de Adhesión Celular/genética , Proteínas de Unión al ADN/genética , Humanos , Proteínas de la Membrana/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Mutación , Proteínas Nucleares/genética , Proteómica
6.
Clin Genet ; 102(6): 530-536, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35932216

RESUMEN

Biallelic pathogenic variants in the genes encoding the dolichol-phosphate mannose synthase subunits (DPM) which produce mannosyl donors for glycosylphosphatidylinositols, N-glycan and protein O- and C-mannosylation, are rare causes of congenital disorders of glycosylation. Pathogenic variants in DPM1 and DPM2 are associated with muscle-eye-brain (MEB) disease, whereas DPM3 variants have mostly been reported in patients with isolated muscle disease-dystroglycanopathy. Thus far, only one affected individual with compound heterozygous DPM3 variants presenting with myopathy, mild intellectual disability, seizures, and nonspecific white matter abnormalities (WMA) around the lateral ventricles has been described. Here we present five affected individuals from four unrelated families with global developmental delay/intellectual disability ranging from mild to severe, microcephaly, seizures, WMA, muscle weakness and variable cardiomyopathy. Exome sequencing of the probands revealed an ultra-rare homozygous pathogenic missense DPM3 variant NM_018973.4:c.221A>G, p.(Tyr74Cys) which segregated with the phenotype in all families. Haplotype analysis indicated that the variant arose independently in three families. Functional analysis did not reveal any alteration in the N-glycosylation pathway caused by the variant; however, this does not exclude its pathogenicity in the function of the DPM complex and related cellular pathways. This report provides supporting evidence that, besides DPM1 and DPM2, defects in DPM3 can also lead to a muscle and brain phenotype.


Asunto(s)
Encefalopatías , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/patología , Homocigoto , Músculo Esquelético/patología , Encefalopatías/patología , Convulsiones/patología , Manosiltransferasas/genética , Proteínas de la Membrana/genética
7.
J Nutr ; 152(5): 1207-1219, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35102398

RESUMEN

BACKGROUND: Nutritional muscle dystrophy is associated with selenium (Se) deficiency; however, the underlying mechanism remains unclear. OBJECTIVES: This study aimed to understand the crosstalk among redox status, energy metabolism, and inflammation in nutritional muscle dystrophy induced by dietary Se deficiency. METHODS: Eighteen castrated male pigs (Yorkshire, 45 d old) were fed Se-deficient (Se-D; 0.007 mg Se/kg) or Se-adequate (Se-A; in the form of selenomethionine, 0.3 mg Se/kg) diets for 16 wk. The muscle Se concentrations; antioxidant capacity; and gene expression, transcriptome, global proteome, metabolome, and lipidome profiles were analyzed. The transcriptome, metabolome, and proteome profiles were analyzed with biostatistics, bioinformatics, and pathway enrichment analysis; other data were analyzed with Student's 2-sided t tests. RESULTS: The muscle Se content in the Se-D group was 96% lower than that in the Se-A group (P < 0.05). The activity of glutathione peroxidase (GPX) and thioredoxin reductase (TXNRD) in the Se-D group was 42%-69% lower than that in the Se-A group (P < 0.05). The mRNA levels of 10 selenoprotein genes were 25%-84% lower than those in the Se-A group (P < 0.05). Multi-omics analyses indicated that the levels of 1378 transcripts, 83 proteins, 22 metabolites, and 55 lipid molecules were significantly altered in response to Se deficiency. Se deficiency-induced redox imbalance led to muscle central carbon and lipid metabolism reprogramming, which enhanced the glycolysis pathway and decreased phospholipid synthesis. Inflammation and apoptosis were observed in response to Se deficiency-induced muscle oxidative stress, which may have been associated with extracellular matrix (ECM) remodeling, suppressed focal adhesion and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling, and activation of the NF-κB signaling pathway. CONCLUSIONS: These results contributed to understanding the crosstalk among redox, energy metabolism, and inflammation in Se deficiency-induced muscle dystrophy in pigs, and may provide intervention targets for muscle disease treatment.


Asunto(s)
Selenio , Animales , Antioxidantes/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Inflamación/metabolismo , Masculino , Músculos/metabolismo , Oxidación-Reducción , Fosfatidilinositol 3-Quinasas/metabolismo , Proteoma/metabolismo , Selenio/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Porcinos
8.
Muscle Nerve ; 66(2): 193-197, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35312090

RESUMEN

INTRODUCTION/AIMS: With current and anticipated disease-modifying treatments, including gene therapy, an early diagnosis for Duchenne muscular dystrophy (DMD) is crucial to assure maximum benefit. In 2009, a study from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet) showed an average diagnosis age of 5 years among males with DMD born from January 1, 1982 to December 31, 2000. Initiatives were implemented by the US Centers for Disease Control and Prevention (CDC) and patient organizations to reduce time to diagnosis. We conducted a follow-up study in a surveillance cohort born after January 1, 2000 to determine whether there has been an improvement in time to diagnosis. METHODS: We assessed the age of diagnosis among males with DMD born from January 1, 2000 to December 31, 2015 using data collected by six US MD STARnet surveillance sites (Colorado, Iowa, western New York State, the Piedmont region of North Carolina, South Carolina, and Utah). The analytic cohort included 221 males with definite or probable DMD diagnosis without a documented family history. We computed frequency count and percentage for categorical variables, and mean, median, and standard deviation (SD) for continuous variables. RESULTS: The mean [median] ages in years of diagnostic milestones were: first signs, 2.7 [2.0]; first creatine kinase (CK), 4.6 [4.6]; DNA/muscle biopsy testing, 4.9 [4.8]; and time from first signs to diagnostic confirmation, 2.2 [1.4]. DISCUSSION: The time interval between first signs of DMD and diagnosis remains unchanged at 2.2 years. This results in lost opportunities for timely genetic counseling, implementation of standards of care, initiation of glucocorticoids, and participation in clinical trials.


Asunto(s)
Distrofia Muscular de Duchenne , Preescolar , Estudios de Cohortes , Estudios de Seguimiento , Humanos , Masculino , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/epidemiología , Distrofia Muscular de Duchenne/genética , Vigilancia de la Población/métodos , Estudios Retrospectivos
9.
Muscle Nerve ; 66(4): 471-478, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35894554

RESUMEN

INTRODUCTION/AIM: The most common limb girdle muscular dystrophy (LGMD) worldwide is LGMD type R1 (LGMDR1). The aim of this study was to correlate the MRI findings with functional scores and to describe the whole-body MRI (WBMRI) pattern in a LGMDR1 Brazilian cohort. METHODS: LGMDR1 patients under follow-up in three centers were referred for the study. Clinical data were collected and a functional evaluation was performed, consisting of Gardner-Medwin and Walton (GMW) and Brooke scales. All patients underwent a WBMRI study (1.5T) with axial T1 and STIR images. Fifty-one muscles were semiquantitatively assessed regarding fatty infiltration and muscle edema. RESULTS: The study group consisted of 18 patients. The highest fatty infiltration scores involved the serratus anterior, biceps femoris long head, adductor magnus, and lumbar erector spinae. There was a latero-medial and caudo-cranial descending gradient of involvement of the paravertebral muscles, with erector spinae being significantly more affected than the transversospinalis muscles (p < 0.05). A striped appearance that has been dubbed the "pseudocollagen sign" was present in 72% of the patients. There was a positive correlation between the MRI score and GMW (Rho:0.83) and Brooke (Rho:0.53) scores. DISCUSSION: WBMRI in LGMDR1 allows a global patient evaluation including involvement of the paraspinal muscles, usually an underestimated feature in the clinical and imaging study of myopathies. Knowledge of the WBMRI pattern of LGMDR1 involvement can be useful in the diagnostic approach and in future studies to identify the best target muscles to serve as outcome measures in clinical trials.


Asunto(s)
Enfermedades Musculares , Distrofia Muscular de Cinturas , Humanos , Imagen por Resonancia Magnética/métodos , Músculo Esquelético/diagnóstico por imagen
10.
Bioorg Med Chem Lett ; 72: 128858, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35718104

RESUMEN

A new series of in vitro potent and highly selective histone methyl transferase enzyme G9a inhibitors was obtained. In particular, compound 2a, one the most potent G9a inhibitor identified, was endowed with >130-fold selectivity over GLP and excellent ligand efficiency. Therefore, it may represent a valuable tool compound to validate the role of highly selective G9a inhibitors in different pathological conditions. When 2a was characterized in vitro in cellular models of skeletal muscle differentiation, a relevant increase of myofibers' size and reduction of the fibroadipogenic infiltration were observed, further confirming the therapeutic potential of selective G9a inhibitors for the treatment of Duchenne muscle dystrophy.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Histonas , Inhibidores Enzimáticos/farmacología
11.
Int J Mol Sci ; 23(5)2022 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-35269765

RESUMEN

BMD is characterized by a marked heterogeneity of gene mutations resulting in many abnormal dystrophin proteins with different expression and residual functions. The smaller dystrophin molecules lacking a portion around exon 48 of the rod domain, named the D8 region, are related to milder phenotypes. The study aimed to determine which proteins might contribute to preserving muscle function in these patients. Patients were subdivided, based on the absence or presence of deletions in the D8 region, into two groups, BMD1 and BMD2. Muscle extracts were analyzed by 2-D DIGE, label-free LC-ESI-MS/MS, and Ingenuity pathway analysis (IPA). Increased levels of proteins typical of fast fibers and of proteins involved in the sarcomere reorganization characterize BMD2. IPA of proteomics datasets indicated in BMD2 prevalence of glycolysis and gluconeogenesis and a correct flux through the TCA cycle enabling them to maintain both metabolism and epithelial adherens junction. A 2-D DIGE analysis revealed an increase of acetylated proteoforms of moonlighting proteins aldolase, enolase, and glyceraldehyde-3-phosphate dehydrogenase that can target the nucleus promoting stem cell recruitment and muscle regeneration. In BMD2, immunoblotting indicated higher levels of myogenin and lower levels of PAX7 and SIRT1/2 associated with a set of proteins identified by proteomics as involved in muscle homeostasis maintenance.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Distrofina/genética , Distrofina/metabolismo , Exones/genética , Humanos , Músculos/metabolismo , Distrofia Muscular de Duchenne/genética , Fenotipo , Espectrometría de Masas en Tándem
12.
Mol Cell Neurosci ; 104: 103482, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32171922

RESUMEN

Dystrophin deficiency is associated with alterations in cell physiology. The functional consequences of dystrophin deficiency are particularly severe for muscle physiology, as observed in Duchenne muscle dystrophy (DMD). DMD is caused by the absence of a 427 kDa isoform of dystrophin. However, in addition to muscular dystrophy symptoms, DMD is frequently associated with memory and attention deficits and epilepsy. While this may be associated with a role for dystrophin in neuronal physiology, it is not clear what neuronal alterations are linked with DMD. Our work shows that CA1 pyramidal neurons from DBA/2J-mdx mice have increased afterhyperpolarization compared to WT controls. All the other electrotonic and electrogenic membrane properties were unaffected by this genotype. Finally, basal synaptic transmission, short-term and long-term synaptic plasticity at Schaffer collateral to CA1 glutamatergic synapses were unchanged between mdx and WT controls. These data show that the excitatory component of hippocampal activity is largely preserved in DBA/2J-mdx mice. Further studies, extending the investigation to the inhibitory GABAergic function, may provide a more complete picture of the functional, network alterations underlying impaired cognition in DMD. In addition, the investigation of changes in neuronal single conductance biophysical properties associated with this genotype, is required to identify the functional alterations associated with dystrophin deficiency and clarify its role in neuronal function.


Asunto(s)
Potenciales de Acción , Hipocampo/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Potenciales Sinápticos , Animales , Células Cultivadas , Ácido Glutámico/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética , Sinapsis/metabolismo , Sinapsis/fisiología , Ácido gamma-Aminobutírico/metabolismo
13.
Acta Neuropathol ; 139(3): 565-582, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31897643

RESUMEN

Protein O-glucosyltransferase 1 (POGLUT1) activity is critical for the Notch signaling pathway, being one of the main enzymes responsible for the glycosylation of the extracellular domain of Notch receptors. A biallelic mutation in the POGLUT1 gene has been reported in one family as the cause of an adult-onset limb-girdle muscular dystrophy (LGMD R21; OMIM# 617232). As the result of a collaborative international effort, we have identified the first cohort of 15 patients with LGMD R21, from nine unrelated families coming from different countries, providing a reliable phenotype-genotype and mechanistic insight. Patients carrying novel mutations in POGLUT1 all displayed a clinical picture of limb-girdle muscle weakness. However, the age at onset was broadened from adult to congenital and infantile onset. Moreover, we now report that the unique muscle imaging pattern of "inside-to-outside" fatty degeneration observed in the original cases is indeed a defining feature of POGLUT1 muscular dystrophy. Experiments on muscle biopsies from patients revealed a remarkable and consistent decrease in the level of the NOTCH1 intracellular domain, reduction of the pool of satellite cells (SC), and evidence of α-dystroglycan hypoglycosylation. In vitro biochemical and cell-based assays suggested a pathogenic role of the novel POGLUT1 mutations, leading to reduced enzymatic activity and/or protein stability. The association between the POGLUT1 variants and the muscular phenotype was established by in vivo experiments analyzing the indirect flight muscle development in transgenic Drosophila, showing that the human POGLUT1 mutations reduced its myogenic activity. In line with the well-known role of the Notch pathway in the homeostasis of SC and muscle regeneration, SC-derived myoblasts from patients' muscle samples showed decreased proliferation and facilitated differentiation. Together, these observations suggest that alterations in SC biology caused by reduced Notch1 signaling result in muscular dystrophy in LGMD R21 patients, likely with additional contribution from α-dystroglycan hypoglycosylation. This study settles the muscular clinical phenotype linked to POGLUT1 mutations and establishes the pathogenic mechanism underlying this muscle disorder. The description of a specific imaging pattern of fatty degeneration and muscle pathology with a decrease of α-dystroglycan glycosylation provides excellent tools which will help diagnose and follow up LGMD R21 patients.


Asunto(s)
Distroglicanos/metabolismo , Glucosiltransferasas/genética , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Femenino , Estudios de Asociación Genética , Glicosilación , Humanos , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Mutación , Linaje , Células Satélite del Músculo Esquelético/patología
14.
Muscle Nerve ; 61(5): 600-607, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32022288

RESUMEN

BACKGROUND: Muscle MRI is of increasing importance for neuromuscular patients to detect changes in muscle volume, fat-infiltration, and edema. We developed a method for semi-automated segmentation of muscle MRI datasets. METHODS: An active contour-evolution algorithm implemented within the ITK-SNAP software was used to segment T1-weighted MRI, and to quantify muscle volumes of neuromuscular patients (n = 65). RESULTS: Semi-automated compared with manual segmentation was shown to be accurate and time-efficient. Muscle volumes and ratios of thigh/lower leg volume were lower in myopathy patients than in controls (P < .0001; P < .05). We found a decrease of lower leg muscle volume in neuropathy patients compared with controls (P < .01), which correlated with clinical parameters. In myopathy patients, muscle volume showed a positive correlation with muscle strength (rleft = 0.79, pleft < .0001). Muscle volumes were independent of body mass index and age. CONCLUSIONS: Our method allows for exact and time-efficient quantification of muscle volumes with possible use as a biomarker in neuromuscular patients.


Asunto(s)
Imagen por Resonancia Magnética , Músculo Esquelético/diagnóstico por imagen , Enfermedades Musculares/diagnóstico por imagen , Enfermedades del Sistema Nervioso Periférico/diagnóstico por imagen , Programas Informáticos , Adulto , Anciano , Automatización , Estudios de Casos y Controles , Enfermedad de Charcot-Marie-Tooth/diagnóstico por imagen , Enfermedad de Charcot-Marie-Tooth/patología , Neuropatías Diabéticas/diagnóstico por imagen , Neuropatías Diabéticas/patología , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Enfermedades Musculares/patología , Distrofia Muscular de Cinturas/diagnóstico por imagen , Distrofia Muscular de Cinturas/patología , Miositis/diagnóstico por imagen , Miositis/patología , Miositis por Cuerpos de Inclusión/diagnóstico por imagen , Miositis por Cuerpos de Inclusión/patología , Tamaño de los Órganos , Enfermedades del Sistema Nervioso Periférico/patología , Polimiositis/diagnóstico por imagen , Polimiositis/patología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/diagnóstico por imagen , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/patología , Estudios Retrospectivos
15.
J Neuroeng Rehabil ; 17(1): 135, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33032627

RESUMEN

BACKGROUND: Physical activity is a recommended part of treatment for numerous neurological and neuromuscular disorders. Yet, many individuals with limited mobility are not able to meet the recommended activity levels. Lightweight, wearable robots like the Myosuit promise to facilitate functional ambulation and thereby physical activity. However, there is limited evidence of the safety and feasibility of training with such devices. METHODS: Twelve participants with diverse motor disorders and the ability to walk for at least 10 m were enrolled in this uncontrolled case series study. The study protocol included five training sessions with a net training time of 45 min each. Primary outcomes were the feasibility of engaging in training with the Myosuit, the occurrence of adverse events, and participant retention. As secondary outcomes, we analyzed the walking speed using the 10-m Walk Test (10MWT) and for three participants, walking endurance using the 2-min Walk Tests. RESULTS: Eight out of 12 participants completed the entire study protocol. Three participants withdrew from the study or were excluded for reasons unrelated to the study. One participant withdrew because of an unsafe feeling when walking with the Myosuit. No adverse events occurred during the study period for any of the participants and all scheduled trainings were completed. For five out of the eight participants that completed the full study, the walking speed when using the Myosuit was higher than to their baseline walking speed. CONCLUSIONS: Activity-based training with the Myosuit appears to be safe, feasible, and well-tolerated by individuals with diverse motor disorders.


Asunto(s)
Dispositivo Exoesqueleto , Trastornos Neurológicos de la Marcha/rehabilitación , Robótica/instrumentación , Adulto , Anciano , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Traumatismos de la Médula Espinal/fisiopatología , Velocidad al Caminar
16.
Am J Med Genet A ; 179(2): 317-321, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30561154

RESUMEN

Kyphoscoliotic Ehlers-Danlos syndrome associated with FKBP14 (FKBP14-kEDS) is an ultrarare autosomal recessive disorder reported in less than 30 individuals so far. In its original description, emphasis was put on the mild muscle involvement. Further reports confirm that FKBP14-kEDS is distinguishable from primary muscle disorders by the lack of progressive muscle disease. We report a 15-year-old girl with FKBP14-kEDS as a result of the recurrent c.362dupC variant, who also showed severe involvement of the lower limb muscles. She never attained autonomous walking and presented significant lower limb weakness. Lower limb magnetic resonance imaging showed a pattern of multiple muscle involvement. Further musculoskeletal assessment revealed significant bone mass density reduction of the spine, unilateral congenital hip dysplasia, and occipitoatlantoaxial instability. This patient points out the existence of a wider phenotypic spectrum of FKBP14-kEDS to include early onset muscle disease.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Isomerasa de Peptidilprolil/genética , Adolescente , Síndrome de Ehlers-Danlos/diagnóstico por imagen , Síndrome de Ehlers-Danlos/fisiopatología , Femenino , Homocigoto , Humanos , Imagen por Resonancia Magnética , Mutación/genética , Fenotipo
17.
BMC Neurol ; 19(1): 32, 2019 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808312

RESUMEN

BACKGROUND: Collagen VI-related myopathies are a spectrum of muscular diseases with features of muscle weakness and atrophy, multiple contractures of joints, distal hyperextensibility, severe respiratory dysfunction and cutaneous alterations, attributable to mutations in the COL6A1, COL6A2, and COL6A3 genes. However, no case of collagen VI mutations with hematuria has been reported. We report a 14-year-old boy who had both Bethlem myopathy and recurrent hematuria and who carried a known de novo COL6A1 missense mutation c.877G > A (p.G293R). CASE PRESENTATION: The patient was a 14-year-old boy presenting with muscle weakness from 3 years of age without any family history. Six months before admission, he developed recurrent gross hematuria, three bouts in total, with the presence of blood clots in the urine. Next-generation sequencing of his whole-exome was performed. The result of sequencing revealed a de novo heterozygous G-to-A nucleotide substitution at position 877 in exon 10 of the COL6A1 gene. After treatment, the hematuria healed, but the muscle weakness failed to improve. CONCLUSIONS: Hematuria in Bethlem myopathy can be caused by COL6 mutations, which may be related to the aberrant connection between collagen VI and collagen IV.


Asunto(s)
Colágeno Tipo VI/genética , Contractura/genética , Distrofias Musculares/congénito , Adolescente , Contractura/complicaciones , Contractura/diagnóstico , Contractura/patología , Hematuria/etiología , Hematuria/genética , Humanos , Masculino , Distrofias Musculares/complicaciones , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Distrofias Musculares/patología , Mutación , Recurrencia
18.
Am J Physiol Cell Physiol ; 314(4): C415-C427, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351412

RESUMEN

Fibrosis is a common feature of several chronic diseases and is characterized by exacerbated accumulation of ECM. An understanding of the cellular and molecular mechanisms involved in the development of this condition is crucial for designing efficient treatments for those pathologies. Connective tissue growth factor (CTGF/CCN2) is a pleiotropic protein with strong profibrotic activity. In this report, we present experimental evidence showing that ECM stimulates the synthesis of CTGF in response to lysophosphatidic acid (LPA).The integrin/focal adhesion kinase (FAK) signaling pathway mediates this effect, since CTGF expression is abolished by the use of the Arg-Gly-Asp-Ser peptide and also by an inhibitor of FAK autophosphorylation at tyrosine 397. Cilengitide, a specific inhibitor of αv integrins, inhibits the expression of CTGF mediated by LPA or transforming growth factor ß1. We show that ECM obtained from decellularized myofibroblast cultures or derived from activated fibroblasts from muscles of the Duchenne muscular dystrophy mouse model ( mdx) induces the expression of CTGF. This effect is dependent on FAK phosphorylation in response to its activation by integrin. We also found that the fibrotic ECM inhibits skeletal muscle differentiation. This novel regulatory mechanism of CTGF expression could be acting as a positive profibrotic feedback between the ECM and CTGF, revealing a novel concept in the control of fibrosis under chronic damage.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Integrina alfaV/metabolismo , Lisofosfolípidos/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/enzimología , Mioblastos/efectos de los fármacos , Animales , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Matriz Extracelular/enzimología , Matriz Extracelular/patología , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Integrina alfaV/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Mioblastos/enzimología , Mioblastos/patología , Fosforilación , Transducción de Señal/efectos de los fármacos
19.
Int J Med Sci ; 13(3): 206-19, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26941581

RESUMEN

BACKGROUND: Diets enriched with n-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to exert a positive impact on muscle diseases. Flaxseed is one of the richest sources of n-3 PUFA acid α-linolenic acid (ALA). The aim of this study was to assess the effects of flaxseed and ALA in models of skeletal muscle degeneration characterized by high levels of Tumor Necrosis Factor-α (TNF). METHODS: The in vivo studies were carried out on dystrophic hamsters affected by muscle damage associated with high TNF plasma levels and fed with a long-term 30% flaxseed-supplemented diet. Differentiating C2C12 myoblasts treated with TNF and challenged with ALA represented the in vitro model. Skeletal muscle morphology was scrutinized by applying the Principal Component Analysis statistical method. Apoptosis, inflammation and myogenesis were analyzed by immunofluorescence. Finally, an in silico analysis was carried out to predict the possible pathways underlying the effects of n-3 PUFAs. RESULTS: The flaxseed-enriched diet protected the dystrophic muscle from apoptosis and preserved muscle myogenesis by increasing the myogenin and alpha myosin heavy chain. Moreover, it restored the normal expression pattern of caveolin-3 thereby allowing protein retention at the sarcolemma. ALA reduced TNF-induced apoptosis in differentiating myoblasts and prevented the TNF-induced inhibition of myogenesis, as demonstrated by the increased expression of myogenin, myosin heavy chain and caveolin-3, while promoting myotube fusion. The in silico investigation revealed that FAK pathways may play a central role in the protective effects of ALA on myogenesis. CONCLUSIONS: These findings indicate that flaxseed may exert potent beneficial effects by preserving skeletal muscle regeneration and homeostasis partly through an ALA-mediated action. Thus, dietary flaxseed and ALA may serve as a useful strategy for treating patients with muscle dystrophies.


Asunto(s)
Lino , Músculo Esquelético/fisiología , Regeneración/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Cricetinae , Suplementos Dietéticos , Ácidos Grasos Omega-3/farmacología , Masculino , Mesocricetus , Ratones , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/dietoterapia , Distrofia Muscular Animal/fisiopatología , Mioblastos Esqueléticos/efectos de los fármacos , Regeneración/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Ácido alfa-Linolénico/farmacología
20.
Stem Cells ; 32(4): 874-85, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24170326

RESUMEN

Duchenne muscular dystrophy (DMD) is an hereditary disease characterized by loss of muscle fibers and their progressive substitution by fat and fibrous tissue. Mesenchymal fibro-adipogenic progenitors (FAPs) expressing the platelet-derived growth factor receptor alpha (PDGFRα) are an important source of fibrosis and adipogenesis in dystrophic skeletal muscle. Among the therapies suggested for dystrophy are those based on nitric oxide (NO) donating drugs, the administration of which slows disease progression. NO has been shown to act by enhancing the regenerative potential of the diseased muscle. Whether it acts also by inhibiting fibrosis and adipogenesis was not known. Here, we show in vitro that NO regulates FAP fate through inhibition of their differentiation into adipocytes. In mdx mice, an animal model of DMD, treatment with the NO donating drug molsidomine reduced the number of PDGFRα(+) cells as well as the deposition of both skeletal muscle fat and connective tissues. Inhibition of adipogenesis was due to NO-induced increased expression of miR-27b leading to downregulation of peroxisome proliferator-activated receptors gamma (Pparγ1) expression in a pathway independent of cGMP generation. These findings reveal an additional effect of NO in dystrophic muscle that conceivably synergizes with its known effects on regeneration improvement and explain why NO-based therapies appear effective in the treatment of muscular dystrophy.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Óxido Nítrico/metabolismo , Adipocitos/patología , Animales , GMP Cíclico , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , PPAR gamma/genética , PPAR gamma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA