Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 61(4): 369-377, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-37935568

RESUMEN

BACKGROUND: Titinopathies are caused by mutations in the titin gene (TTN). Titin is the largest known human protein; its gene has the longest coding phase with 364 exons. Titinopathies are very complex neuromuscular pathologies due to the variable age of onset of symptoms, the great diversity of pathological and muscular impairment patterns (cardiac, skeletal muscle or mixed) and both autosomal dominant and recessive modes of transmission. Until now, only few CNVs in TTN have been reported without clear genotype-phenotype associations. METHODS: Our study includes eight families with dominant titinopathies. We performed next-generation sequencing or comparative genomic hybridisation array analyses and found CNVs in the TTN gene. We characterised these CNVs by RNA sequencing (RNAseq) analyses in six patients' muscles and performed genotype-phenotype inheritance association study by combining the clinical and biological data of these eight families. RESULTS: Seven deletion-type CNVs in the TTN gene were identified among these families. Genotype and RNAseq results showed that five deletions do not alter the reading frame and one is out-of-reading frame. The main phenotype identified was distal myopathy associated with contractures. The analysis of morphological, clinical and genetic data and imaging let us draw new genotype-phenotype associations of titinopathies. CONCLUSION: Identifying TTN CNVs will further increase diagnostic sensitivity in these complex neuromuscular pathologies. Our cohort of patients enabled us to identify new deletion-type CNVs in the TTN gene, with unexpected autosomal dominant transmission. This is valuable in establishing new genotype-phenotype associations of titinopathies, mainly distal myopathy in most of the patients.


Asunto(s)
Miopatías Distales , Humanos , Conectina/genética , Miopatías Distales/genética , Variaciones en el Número de Copia de ADN/genética , Músculo Esquelético/patología , Mutación/genética , Fenotipo
2.
Am J Hum Genet ; 108(5): 840-856, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33861953

RESUMEN

JAG2 encodes the Notch ligand Jagged2. The conserved Notch signaling pathway contributes to the development and homeostasis of multiple tissues, including skeletal muscle. We studied an international cohort of 23 individuals with genetically unsolved muscular dystrophy from 13 unrelated families. Whole-exome sequencing identified rare homozygous or compound heterozygous JAG2 variants in all 13 families. The identified bi-allelic variants include 10 missense variants that disrupt highly conserved amino acids, a nonsense variant, two frameshift variants, an in-frame deletion, and a microdeletion encompassing JAG2. Onset of muscle weakness occurred from infancy to young adulthood. Serum creatine kinase (CK) levels were normal or mildly elevated. Muscle histology was primarily dystrophic. MRI of the lower extremities revealed a distinct, slightly asymmetric pattern of muscle involvement with cores of preserved and affected muscles in quadriceps and tibialis anterior, in some cases resembling patterns seen in POGLUT1-associated muscular dystrophy. Transcriptome analysis of muscle tissue from two participants suggested misregulation of genes involved in myogenesis, including PAX7. In complementary studies, Jag2 downregulation in murine myoblasts led to downregulation of multiple components of the Notch pathway, including Megf10. Investigations in Drosophila suggested an interaction between Serrate and Drpr, the fly orthologs of JAG1/JAG2 and MEGF10, respectively. In silico analysis predicted that many Jagged2 missense variants are associated with structural changes and protein misfolding. In summary, we describe a muscular dystrophy associated with pathogenic variants in JAG2 and evidence suggests a disease mechanism related to Notch pathway dysfunction.


Asunto(s)
Proteína Jagged-2/genética , Distrofias Musculares/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Línea Celular , Niño , Preescolar , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Glucosiltransferasas/genética , Haplotipos/genética , Humanos , Proteína Jagged-1/genética , Proteína Jagged-2/química , Proteína Jagged-2/deficiencia , Proteína Jagged-2/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Persona de Mediana Edad , Modelos Moleculares , Músculos/metabolismo , Músculos/patología , Distrofias Musculares/patología , Mioblastos/metabolismo , Mioblastos/patología , Linaje , Fenotipo , Receptores Notch/metabolismo , Transducción de Señal , Secuenciación del Exoma , Adulto Joven
3.
Brain ; 144(8): 2427-2442, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-33792664

RESUMEN

Marinesco-Sjögren syndrome is a rare human disorder caused by biallelic mutations in SIL1 characterized by cataracts in infancy, myopathy and ataxia, symptoms which are also associated with a novel disorder caused by mutations in INPP5K. While these phenotypic similarities may suggest commonalties at a molecular level, an overlapping pathomechanism has not been established yet. In this study, we present six new INPP5K patients and expand the current mutational and phenotypical spectrum of the disease showing the clinical overlap between Marinesco-Sjögren syndrome and the INPP5K phenotype. We applied unbiased proteomic profiling on cells derived from Marinesco-Sjögren syndrome and INPP5K patients and identified alterations in d-3-PHGDH as a common molecular feature. d-3-PHGDH modulates the production of l-serine and mutations in this enzyme were previously associated with a neurological phenotype, which clinically overlaps with Marinesco-Sjögren syndrome and INPP5K disease. As l-serine administration represents a promising therapeutic strategy for d-3-PHGDH patients, we tested the effect of l-serine in generated sil1, phgdh and inpp5k a+b zebrafish models, which showed an improvement in their neuronal phenotype. Thus, our study defines a core phenotypical feature underpinning a key common molecular mechanism in three rare diseases and reveals a common and novel therapeutic target for these patients.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Inositol Polifosfato 5-Fosfatasas/genética , Mutación , Fenotipo , Fosfoglicerato-Deshidrogenasa/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Proteómica , Degeneraciones Espinocerebelosas/patología , Pez Cebra
4.
Ann Neurol ; 88(2): 332-347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32403198

RESUMEN

OBJECTIVE: A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS: We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS: A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION: The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.


Asunto(s)
Dimetilaliltranstransferasa/genética , Farnesiltransferasa/genética , Geraniltranstransferasa/genética , Pérdida Auditiva/genética , Distrofias Musculares/genética , Mutación/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Pérdida Auditiva/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofias Musculares/diagnóstico por imagen , Linaje , Insuficiencia Ovárica Primaria/diagnóstico por imagen , Estructura Secundaria de Proteína , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos , Adulto Joven
5.
Am J Hum Genet ; 99(3): 753-761, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27569547

RESUMEN

The neuromuscular junction (NMJ) is one of the best-studied cholinergic synapses. Inherited defects of peripheral neurotransmission result in congenital myasthenic syndromes (CMSs), a clinically and genetically heterogeneous group of rare diseases with fluctuating fatigable muscle weakness as the clinical hallmark. Whole-exome sequencing and Sanger sequencing in six unrelated families identified compound heterozygous and homozygous mutations in SLC5A7 encoding the presynaptic sodium-dependent high-affinity choline transporter 1 (CHT), which is known to be mutated in one dominant form of distal motor neuronopathy (DHMN7A). We identified 11 recessive mutations in SLC5A7 that were associated with a spectrum of severe muscle weakness ranging from a lethal antenatal form of arthrogryposis and severe hypotonia to a neonatal form of CMS with episodic apnea and a favorable prognosis when well managed at the clinical level. As expected given the critical role of CHT for multisystemic cholinergic neurotransmission, autonomic dysfunctions were reported in the antenatal form and cognitive impairment was noticed in half of the persons with the neonatal form. The missense mutations induced a near complete loss of function of CHT activity in cell models. At the human NMJ, a delay in synaptic maturation and an altered maintenance were observed in the antenatal and neonatal forms, respectively. Increased synaptic expression of butyrylcholinesterase was also observed, exposing the dysfunction of cholinergic metabolism when CHT is deficient in vivo. This work broadens the clinical spectrum of human diseases resulting from reduced CHT activity and highlights the complexity of cholinergic metabolism at the synapse.


Asunto(s)
Apnea/genética , Mutación/genética , Miastenia Gravis/genética , Terminales Presinápticos/metabolismo , Simportadores/genética , Simportadores/metabolismo , Adolescente , Apnea/complicaciones , Apnea/metabolismo , Apnea/patología , Artrogriposis/complicaciones , Artrogriposis/genética , Butirilcolinesterasa/metabolismo , Niño , Preescolar , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Análisis Mutacional de ADN , Exoma/genética , Femenino , Genes Recesivos/genética , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Hipotonía Muscular/genética , Debilidad Muscular/complicaciones , Debilidad Muscular/genética , Debilidad Muscular/patología , Mutación Missense/genética , Miastenia Gravis/complicaciones , Miastenia Gravis/metabolismo , Miastenia Gravis/patología , Unión Neuromuscular/enzimología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Terminales Presinápticos/patología , Simportadores/deficiencia , Transmisión Sináptica
7.
J Neurol Neurosurg Psychiatry ; 89(5): 506-512, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29175898

RESUMEN

BACKGROUND: Mutations in the gene coding for protein O-mannosyl-transferase 2 (POMT2) are known to cause severe congenital muscular dystrophy, and recently, mutations in POMT2 have also been linked to a milder limb-girdle muscular dystrophy (LGMD) phenotype, named LGMD type 2N (LGMD2N). Only four cases have been reported so far.ClinicalTrials.gov ID: NCT02759302 METHODS: We report 12 new cases of LGMD2N, aged 18-63 years. Muscle involvement was assessed by MRI, muscle strength testing and muscle biopsy analysis. Other clinical features were also recorded. RESULTS: Presenting symptoms were difficulties in walking, pain during exercise, delayed motor milestones and learning disabilities at school. All had some degree of cognitive impairment. Brain MRIs were abnormal in 3 of 10 patients, showing ventricular enlargement in one, periventricular hyperintensities in another and frontal atrophy of the left hemisphere in a third patient. Most affected muscle groups were hip and knee flexors and extensors on strength testing. On MRI, most affected muscles were hamstrings followed by paraspinal and gluteal muscles. The 12 patients in our cohort carried 11 alleles with known mutations, whereas 11 novel mutations accounted for the remaining 13 alleles. CONCLUSION: We describe the first cohort of patients with LGMD2N and show that unlike other LGMD types, all patients had cognitive impairment. Primary muscle involvement was found in hamstring, paraspinal and gluteal muscles on MRI, which correlated well with reduced muscle strength in hip and knee flexors and extensors. The study expands the mutational spectrum for LGMD2N, with the description of 11 novel POMT2 mutations in the association with LGMD2N. CLINICAL TRIAL REGISTRATION: NCT02759302.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Manosiltransferasas/genética , Distrofia Muscular de Cinturas/genética , Adolescente , Adulto , Alelos , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Debilidad Muscular/complicaciones , Debilidad Muscular/fisiopatología , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/patología , Distrofia Muscular de Cinturas/fisiopatología , Mutación , Neuroimagen , Adulto Joven
8.
J Neuromuscul Dis ; 10(1): 125-133, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36373293

RESUMEN

We report three siblings from a non-consanguineous family presenting with contractural limb-girdle phenotype with intrafamilial variability. Muscle MRI showed posterior thigh and quadriceps involvement with a sandwich-like sign. Whole-exome sequencing identified two compound heterozygous missense TTN variants and one heterozygous LAMA2 variant. Brain MRI performed because of concentration difficulties in one of the siblings evidenced white-matter abnormalities, subsequently found in the others. The genetic analysis was re-oriented, revealing a novel pathogenic intronic LAMA2 variant which confirmed the LAMA2-RD diagnosis. This work highlights the importance of a thorough clinical phenotyping and the importance of brain imaging, in order to orientate and interpret the genetic analysis.


Asunto(s)
Distrofia Muscular de Cinturas , Distrofias Musculares , Humanos , Distrofia Muscular de Cinturas/diagnóstico por imagen , Distrofia Muscular de Cinturas/genética , Distrofias Musculares/diagnóstico por imagen , Distrofias Musculares/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Pruebas Genéticas , Neuroimagen
10.
Nat Commun ; 13(1): 2306, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35484142

RESUMEN

Missense variants in RNA-binding proteins (RBPs) underlie a spectrum of disease phenotypes, including amyotrophic lateral sclerosis, frontotemporal dementia, and inclusion body myopathy. Here, we present ten independent families with a severe, progressive muscular dystrophy, reminiscent of oculopharyngeal muscular dystrophy (OPMD) but of much earlier onset, caused by heterozygous frameshift variants in the RBP hnRNPA2/B1. All disease-causing frameshift mutations abolish the native stop codon and extend the reading frame, creating novel transcripts that escape nonsense-mediated decay and are translated to produce hnRNPA2/B1 protein with the same neomorphic C-terminal sequence. In contrast to previously reported disease-causing missense variants in HNRNPA2B1, these frameshift variants do not increase the propensity of hnRNPA2 protein to fibrillize. Rather, the frameshift variants have reduced affinity for the nuclear import receptor karyopherin ß2, resulting in cytoplasmic accumulation of hnRNPA2 protein in cells and in animal models that recapitulate the human pathology. Thus, we expand the phenotypes associated with HNRNPA2B1 to include an early-onset form of OPMD caused by frameshift variants that alter its nucleocytoplasmic transport dynamics.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Distrofia Muscular Oculofaríngea , Esclerosis Amiotrófica Lateral/genética , Animales , Mutación del Sistema de Lectura , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Heterocigoto , Humanos , Distrofia Muscular Oculofaríngea/genética
11.
Eur J Med Genet ; 63(12): 104063, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32947049

RESUMEN

OBJECTIVE: To perform genotype-phenotype, clinical and molecular analysis in a large 3-generation family with autosomal dominant congenital spinal muscular atrophy. METHODS: Using a combined genetic approach including whole genome scanning, next generation sequencing-based multigene panel, whole genome sequencing, and targeted variant Sanger sequencing, we studied the proband and multiple affected individuals of this family who presented bilateral proximal lower limb muscle weakness and atrophy. RESULTS: We identified a novel heterozygous variant, c.1826T > C; p.Ile609Thr, in the DYNC1H1 gene localized within the common haplotype in the 14q32.3 chromosomal region which cosegregated with disease in this large family. Within the family, affected individuals were found to have a wide array of clinical variability. Although some individuals presented the typical lower motor neuron phenotype with areflexia and denervation, others presented with muscle weakness and atrophy, hyperreflexia, and absence of denervation suggesting a predominant upper motor neuron disease. In addition, some affected individuals presented with an intermediate phenotype characterized by hyperreflexia and denervation, expressing a combination of lower and upper motor neuron defects. CONCLUSION: Our study demonstrates the wide clinical variability associated with a single disease causing variant in DYNC1H1 gene and this variant demonstrated a high penetrance within this large family.


Asunto(s)
Dineínas Citoplasmáticas/genética , Atrofia Muscular Espinal/genética , Mutación Missense , Adolescente , Adulto , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Extremidad Inferior/fisiopatología , Masculino , Persona de Mediana Edad , Neuronas Motoras/fisiología , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Linaje , Fenotipo , Reflejo , Extremidad Superior/fisiopatología
12.
Neurol Genet ; 5(2): e321, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31119192

RESUMEN

OBJECTIVE: To study the genetic and phenotypic spectrum of patients harboring recessive mutations in BVES. METHODS: We performed whole-exome sequencing in a multicenter cohort of 1929 patients with a suspected hereditary myopathy, showing unexplained limb-girdle muscular weakness and/or elevated creatine kinase levels. Immunohistochemistry and mRNA experiments on patients' skeletal muscle tissue were performed to study the pathogenicity of identified loss-of-function (LOF) variants in BVES. RESULTS: We identified 4 individuals from 3 families harboring homozygous LOF variants in BVES, the gene that encodes for Popeye domain containing protein 1 (POPDC1). Patients showed skeletal muscle involvement and cardiac conduction abnormalities of varying nature and severity, but all exhibited at least subclinical signs of both skeletal muscle and cardiac disease. All identified mutations lead to a partial or complete loss of function of BVES through nonsense-mediated decay or through functional changes to the POPDC1 protein. CONCLUSIONS: We report the identification of homozygous LOF mutations in BVES, causal in a young adult-onset myopathy with concomitant cardiac conduction disorders in the absence of structural heart disease. These findings underline the role of POPDC1, and by extension, other members of this protein family, in striated muscle physiology and disease. This disorder appears to have a low prevalence, although it is probably underdiagnosed because of its striking phenotypic variability and often subtle yet clinically relevant manifestations, particularly concerning the cardiac conduction abnormalities.

13.
Mol Ther Nucleic Acids ; 10: 376-386, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499949

RESUMEN

We assessed the potential of Lmna-mRNA repair by spliceosome-mediated RNA trans-splicing as a therapeutic approach for LMNA-related congenital muscular dystrophy. This gene therapy strategy leads to reduction of mutated transcript expression for the benefit of corresponding wild-type (WT) transcripts. We developed 5'-RNA pre-trans-splicing molecules containing the first five exons of Lmna and targeting intron 5 of Lmna pre-mRNA. Among nine pre-trans-splicing molecules, differing in the targeted sequence in intron 5 and tested in C2C12 myoblasts, three induced trans-splicing events on endogenous Lmna mRNA and confirmed at protein level. Further analyses performed in primary myotubes derived from an LMNA-related congenital muscular dystrophy (L-CMD) mouse model led to a partial rescue of the mutant phenotype. Finally, we tested this approach in vivo using adeno-associated virus (AAV) delivery in newborn mice and showed that trans-splicing events occurred in WT mice 50 days after AAV delivery, although at a low rate. Altogether, while these results provide the first evidence for reprogramming LMNA mRNA in vitro, strategies to improve the rate of trans-splicing events still need to be developed for efficient application of this therapeutic approach in vivo.

14.
J Neuropathol Exp Neurol ; 77(12): 1101-1114, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30365001

RESUMEN

Titin-related myopathies are heterogeneous clinical conditions associated with mutations in TTN. To define their histopathologic boundaries and try to overcome the difficulty in assessing the pathogenic role of TTN variants, we performed a thorough morphological skeletal muscle analysis including light and electron microscopy in 23 patients with different clinical phenotypes presenting pathogenic autosomal dominant or autosomal recessive (AR) mutations located in different TTN domains. We identified a consistent pattern characterized by diverse defects in oxidative staining with prominent nuclear internalization in congenital phenotypes (AR-CM) (n = 10), ± necrotic/regenerative fibers, associated with endomysial fibrosis and rimmed vacuoles (RVs) in AR early-onset Emery-Dreifuss-like (AR-ED) (n = 4) and AR adult-onset distal myopathies (n = 4), and cytoplasmic bodies (CBs) as predominant finding in hereditary myopathy with early respiratory failure (HMERF) patients (n = 5). Ultrastructurally, the most significant abnormalities, particularly in AR-CM, were multiple narrow core lesions and/or clear small areas of disorganizations affecting one or a few sarcomeres with M-band and sometimes A-band disruption and loss of thick filaments. CBs were noted in some AR-CM and associated with RVs in HMERF and some AR-ED cases. As a whole, we described recognizable histopathological patterns and structural alterations that could point toward considering the pathogenicity of TTN mutations.


Asunto(s)
Conectina/genética , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Sarcómeros/genética , Sarcómeros/patología , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Músculo Esquelético/ultraestructura , Estudios Retrospectivos , Adulto Joven
15.
Neurol Genet ; 3(6): e208, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29264399

RESUMEN

OBJECTIVE: To describe the variability of muscle symptoms in patients carrying mutations in the GYG1 gene, encoding glycogenin-1, an enzyme involved in the biosynthesis of glycogen, and to discuss genotype-phenotype relations. METHODS: We describe 9 patients from 5 families in whom muscle biopsies showed vacuoles with an abnormal accumulation of glycogen in muscle fibers, partially α-amylase resistant suggesting polyglucosan bodies. The patients had either progressive early-onset limb-girdle weakness or late-onset distal or scapuloperoneal muscle affection as shown by muscle imaging. No clear definite cardiac disease was found. Histologic and protein analysis investigations were performed on muscle. RESULTS: Genetic analyses by direct or exome sequencing of the GYG1 gene revealed 6 different GYG1 mutations. Four of the mutations were novel. They were compound heterozygous in 3 families and homozygous in 2. Protein analysis revealed either the absence of glycogenin-1 or reduced glycogenin-1 expression with impaired glucosylation. CONCLUSIONS: Our report extends the genetic and clinical spectrum of glycogenin-1-related myopathies to include scapuloperoneal and distal affection with glycogen accumulation.

16.
J Neurol ; 253(6): 714-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16511635

RESUMEN

Hereditary spastic paraplegias are genetically and clinically heterogeneous. Twenty-six loci have been identified to date. SPG27 was recently mapped to chromosome 10 in a single family with autosomal recessive hereditary spastic paraplegia (AR-HSP) and a pure phenotype. We describe a Tunisian family with a complicated form of AR-HSP also linked to SPG27. The parents are first cousins and 3 out of their 4 children manifest early onset progressive spastic paraparesis associated with sensorimotor polyneuropathy. In addition, the eldest girl had facial dysmorphism and short stature (-3SD). Two of the three patients were mentally retarded, and one of these also had cerebellar signs. Their ages at onset were 2, 5 and 7 years. A genome-wide scan suggested linkage to SPG27 on the long arm of chromosome 10 with a multipoint lod score of 2.54. In addition, a recombination detected in this family by haplotype reconstruction reduced the SPG27 locus from 25 to 19.6 cM. This is the first clinical description of a complicated form of spastic paraplegia, characterized by great phenotypic variability among the sibs, associated with the SPG27 locus.


Asunto(s)
Corteza Cerebral/patología , Cromosomas Humanos Par 10 , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/patología , Adulto , Mapeo Cromosómico/métodos , Salud de la Familia , Femenino , Humanos , Escala de Lod , Imagen por Resonancia Magnética , Masculino , Fenotipo
17.
J Neuromuscul Dis ; 3(4): 497-510, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27911330

RESUMEN

BACKGROUND: Emery-Dreifuss muscular dystrophy (EDMD) is associated with mutations in EMD and LMNA genes, encoding for the nuclear envelope proteins emerin and lamin A/C, indicating that EDMD is a nuclear envelope disease. We recently reported mutations in FHL1 gene in X-linked EDMD. FHL1 encodes FHL1A, and the two minor isoforms FHL1B and FHL1C. So far, none have been described at the nuclear envelope. OBJECTIVE: To gain insight into the pathophysiology of EDMD, we focused our attention on the poorly characterized FHL1B isoform. METHODS: The amount and the localisation of FHL1B were evaluated in control and diseased human primary myoblasts using immunofluorescence and western blotting. RESULTS: We found that in addition to a cytoplasmic localization, this isoform strongly accumulated at the nuclear envelope of primary human myoblasts, like but independently of lamin A/C and emerin. During myoblast differentiation, we observed a major reduction of FHL1B protein expression, especially in the nucleus. Interestingly, we found elevated FHL1B expression level in myoblasts from an FHL1-related EDMD patient where the FHL1 mutation only affects FHL1A, as well as in myoblasts from an LMNA-related EDMD patient. CONCLUSIONS: Altogether, the specific localization of FHL1B and its modulation in disease-patient's myoblasts confirmed FHL1-related EDMD as a nuclear envelope disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Lamina Tipo A/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , Western Blotting , Estudios de Casos y Controles , Técnica del Anticuerpo Fluorescente , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mutación , Lámina Nuclear , Isoformas de Proteínas
18.
J Neuromuscul Dis ; 2(3): 229-240, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-27858741

RESUMEN

BACKGROUND: Laminin α2 deficient congenital muscular dystrophy, caused by mutations in the LAMA2 gene, is characterized by early muscle weakness associated with abnormal white matter signal on cerebral MRI. OBJECTIVE: To report on 4 patients with LAMA2 gene mutations whose original clinical features complicated the diagnosis strategy. METHODS: Clinical, electrophysiological, muscle imaging and histopathological data were retrospectively collected from all patients. DNA samples were analysed by next-generation sequencing or direct gene sequencing. Laminin α2 was analysed by western-blot and immunohistochemistry. RESULTS: The four patients achieved independent walking. All had proximal muscle weakness with scapular winging and prominent joint contractures without peripheral neuropathy. During follow-up, two patients suffered from refractory epilepsy associated with brain leukoencephalopathy in one, polymicrogyria and lissencephaly without white matter changes in the other. In two patients, the distribution of fatty infiltration resembles that of collagen-VI related myopathies. Dilated cardiomyopathy contstartabstractwith conduction defects, suggestive of Emery-Dreifuss myopathy, emerged in two of them within the 4th decade. Molecular diagnosis remained elusive for many years. Finally, targeted capture-DNA sequencing unveiled the involvement of the LAMA2 gene in two families, and led us to further identify LAMA2 mutations in the remaining family using Sanger sequencing. CONCLUSIONS: This report extends the clinical and radiological features of partial Laminin α2 deficiency since patients showed atypical manifestations including dilated cardiomyopathy with conduction defects in 2, epilepsy in 2, one of whom also had sole cortical brain abnormalities. Importantly, clinical findings and muscle imaging initially pointed to collagen-VI related disorders and Emery-Dreifuss muscular dystrophy.

19.
Neurology ; 85(24): 2126-35, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26581302

RESUMEN

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


Asunto(s)
Cardiomiopatías , Conectina/genética , Mutación del Sistema de Lectura/genética , Distrofia Muscular de Emery-Dreifuss/diagnóstico , Distrofia Muscular de Emery-Dreifuss/genética , Fenotipo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
20.
Eur J Hum Genet ; 23(7): 929-34, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25351777

RESUMEN

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


Asunto(s)
Hibridación Genómica Comparativa/métodos , Eliminación de Gen , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Distrofia Muscular de Cinturas/genética , Factores de Transcripción/genética , Adulto , Secuencia de Bases , Análisis Mutacional de ADN/métodos , Salud de la Familia , Femenino , Humanos , Masculino , Distrofia Muscular de Cinturas/patología , Linaje , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA