Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Neurol Sci ; 40(3): 503-507, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30542963

RESUMEN

INTRODUCTION: The slow-channel congenital myasthenic syndrome (SCCMS) is a postsynaptic form of congenital myasthenic syndromes (CMSs), a clinically heterogeneous group of disorders caused by genetic defects leading to an abnormal signal transmission at the endplate. METHODS: We report clinical and molecular data of a multigenerational family in which the presentation of a progressive proximal-distal weakness with ocular involvement led to a number of different clinical diagnoses. RESULTS: A comprehensive genetic study which included whole-genome linkage analysis and whole-exome sequencing identified a heterozygous missense substitution (c.721C>T, p.L241F) in the ε subunit of the acetylcholine receptor (CHRNE) that was consistent with clinical weakness in all patients. DISCUSSION: SCCMS is characterized by a broad and heterogeneous clinical phenotype in which disease onset, symptoms, severity, and progression can be highly variable even between family members. The identification of a CHRNE mutation allowed to make the definitive diagnosis of CMS in this family and contributed to define the clinical spectrum of this disease.


Asunto(s)
Salud de la Familia , Mutación Missense/genética , Síndromes Miasténicos Congénitos/genética , Receptores Nicotínicos/genética , Adulto , Anciano , Electromiografía , Femenino , Ligamiento Genético , Humanos , Italia , Masculino , Persona de Mediana Edad , Músculo Esquelético/fisiopatología , Síndromes Miasténicos Congénitos/patología , Síndromes Miasténicos Congénitos/fisiopatología , Conducción Nerviosa/genética , Secuenciación del Exoma
2.
Biochim Biophys Acta ; 1860(6): 1247-55, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26968460

RESUMEN

OBJECTIVES: Deregulation of axonal transport in neurons is emerging as the major cause of many neurodegenerative diseases in human, such as Charcot-Marie-Tooth (CMT) neuropathy. However, little is known about how mitochondria move in vivo and whether cell culture systems truly represent what happens in living animals. Here we describe the generation of a new zebrafish transgenic line that specifically allows to study mitochondrial dynamics in motor neurons and its application to analyse mitochondrial movement in zebrafish models expressing CMT2A causing mutations. METHODS: The Tol2 transposon system was used to generate a transgenic zebrafish line expressing the photoconvertible fluorescent protein Kaede in mitochondria of motor neurons. Mitochondrial shape and movement were monitored by time-lapse confocal live imaging and measured by kymograph analysis. The effects of two well-known CMT causing mutations, L76P and R94Q substitutions in MFN2, were then investigated with the same methods. RESULTS: We generated the transgenic zebrafish Tg(hb9:MTS-Kaede) line with genetically labelled mitochondria in motor neurons. Kaede protein was correctly and stably targeted to mitochondrial matrix while retaining its photoconvertibility, thus qualifying this model for in vivo studies. Expression of the L76P and R94Q mutations reduced mitochondrial movement in axons and altered mitochondrial distribution in distinct ways. CONCLUSIONS AND GENERAL SIGNIFICANCE: These findings confirm previously published data obtained in cell cultures and strengthen the hypothesis of different mechanism of action of the two MFN2 mutations. Considering the number of neurodegenerative diseases associated to mitochondrial dynamics, the Tg(hb9:MTS-Kaede) zebrafish line is a promising model to study in vivo alterations of mitochondrial transport underlying human diseases.


Asunto(s)
Transporte Axonal/fisiología , Mitocondrias/fisiología , Animales , Animales Modificados Genéticamente , Enfermedad de Charcot-Marie-Tooth/genética , GTP Fosfohidrolasas/fisiología , Dinámicas Mitocondriales , Proteínas Mitocondriales/fisiología , Mutación , Pez Cebra
3.
Mol Cell Probes ; 27(3-4): 118-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23384994

RESUMEN

X-linked Charcot-Marie-Tooth disease (CMT Type X1, OMIM: 302800) represents a frequent cause of hereditary peripheral motor and sensory neuropathies and is associated with mutations in GJB1 encoding the gap junction beta 1 protein connexin 32 (Cx32). Studying an Argentinean family of Italian origin with seven affected males in three generations exhibiting clinical signs of CMT, eight obligate female carriers were identified genealogically. DNA sequencing of exon 2 and adjacent regions of the GJB1 gene in two symptomatic males whose respective maternal grandfathers, both affected, were brothers, revealed mutations in GJB1/Cx32. Surprisingly, each of the two affected patients had a different mutation in hemizygous state at the same nucleotide position: c.383C>T (p.S128L) and c.383C>A (p.S128X). In both cases, the identified mutation was present in heterozygous state in the corresponding maternal genomic DNA. Furthermore, X-chromosomal microsatellite analysis showed identical marker alleles in both patients. Together with the genealogical information, these molecular data imply that a primarily mutated allele mutated for a second time. In conclusion, two different mutations at the same nucleotide position in this Argentinean family represent a finding with a very low probability of occurrence.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Conexinas/genética , Alelos , Argentina , Enfermedad de Charcot-Marie-Tooth/etnología , Femenino , Haplotipos , Humanos , Masculino , Repeticiones de Microsatélite , Mutación , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Proteína beta1 de Unión Comunicante
4.
J Hum Genet ; 56(12): 869-72, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21993419

RESUMEN

The present study aims at investigating the association between common and rare variants of mitochondrial DNA (mtDNA), and increased risk of schizophrenia (SZ) and bipolar disorder (BPD) in a cohort of patients originating from the same Italian population. The distribution of the major European mtDNA haplogroups was determined in 89 patients and their frequencies did not significantly differ from those observed in the Italian population. Moreover, 27 patients with high probability of having inherited the disease from the maternal side were selected for whole mitochondrial genome sequencing to investigate the possible presence of causative point mutations. Overall, 213 known variants and 2 novel changes were identified, but none of them was predicted to have functional effects. Hence, none of the sequence changes we found in our sample could explain the maternal component of SZ and BPD predisposition.


Asunto(s)
Trastorno Bipolar/genética , ADN Mitocondrial , Genoma Mitocondrial , Esquizofrenia/genética , Estudios de Cohortes , Predisposición Genética a la Enfermedad , Humanos , Filogenia , Mutación Puntual
6.
BMC Med ; 7: 14, 2009 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-19351384

RESUMEN

BACKGROUND: Spinal muscular atrophy (SMA) is a neurodegenerative disorder associated with mutations of the survival motor neuron gene SMN and is characterized by muscle weakness and atrophy caused by degeneration of spinal motor neurons. SMN has a role in neurons but its deficiency may have a direct effect on muscle tissue. METHODS: We applied microarray and quantitative real-time PCR to study at transcriptional level the effects of a defective SMN gene in skeletal muscles affected by the two forms of SMA: the most severe type I and the mild type III. RESULTS: The two forms of SMA generated distinct expression signatures: the SMA III muscle transcriptome is close to that found under normal conditions, whereas in SMA I there is strong alteration of gene expression. Genes implicated in signal transduction were up-regulated in SMA III whereas those of energy metabolism and muscle contraction were consistently down-regulated in SMA I. The expression pattern of gene networks involved in atrophy signaling was completed by qRT-PCR, showing that specific pathways are involved, namely IGF/PI3K/Akt, TNF-alpha/p38 MAPK and Ras/ERK pathways. CONCLUSION: Our study suggests a different picture of atrophy pathways in each of the two forms of SMA. In particular, p38 may be the regulator of protein synthesis in SMA I. The SMA III profile appears as the result of the concurrent presence of atrophic and hypertrophic fibers. This more favorable condition might be due to the over-expression of MTOR that, given its role in the activation of protein synthesis, could lead to compensatory hypertrophy in SMA III muscle fibers.


Asunto(s)
Músculo Esquelético/metabolismo , Atrofia Muscular Espinal/genética , Proteínas Quinasas/metabolismo , Transcripción Genética , Apoptosis , Biopsia , Regulación hacia Abajo , Perfilación de la Expresión Génica , Genotipo , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/clasificación , Atrofia Muscular Espinal/patología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Proteínas del Complejo SMN/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora , Serina-Treonina Quinasas TOR , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Arch Neurol ; 63(5): 750-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16682546

RESUMEN

BACKGROUND: Hereditary spastic paraplegia (HSP) is a group of genetically heterogeneous disorders characterized by progressive spasticity of the lower limbs. Mutations in the SPG4 gene, which encodes spastin protein, are responsible for up to 45% of autosomal dominant cases. OBJECTIVE: To search for disease-causing mutations in a large series of Italian patients with HSP. DESIGN: Samples of DNA were analyzed by direct sequencing of all exons in SPG4. Samples from a subset of patients were also analyzed by direct sequencing of all exons in SPG3A, SPG6, SPG10, and SPG13. SETTING: Molecular testing facility in Italy. PATIENTS: Sixty unrelated Italian patients with pure (n = 50) and complicated (n = 10) HSP. MAIN OUTCOME MEASURES: Mutations in SPG4, SPG3A, SPG6, SPG10, and SPG13. RESULTS: We identified 12 different mutations, 8 of which were novel, in 13 patients. No mutations of any of the other HSP genes tested were found in 15 patients with sporadic pure HSP who did not have mutations in the SPG4 gene. CONCLUSIONS: The overall rate of mutation in the SPG4 gene within our sample was 22%, rising to 26% when only patients with pure HSP were considered. The negative result obtained in 15 patients without mutations in SPG4 in whom 4 other genes were analyzed (SPG3A, SPG6, SPG10, and SPG13) indicate that these genes are not frequently mutated in sporadic pure HSP.


Asunto(s)
Adenosina Trifosfatasas/genética , Mutación , Paraplejía Espástica Hereditaria/genética , Adulto , Anciano , Análisis Mutacional de ADN/métodos , Exones , Salud de la Familia , Femenino , Humanos , Italia , Masculino , Persona de Mediana Edad , Paraplejía Espástica Hereditaria/clasificación , Espastina
9.
J Mol Diagn ; 18(6): 912-922, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27637300

RESUMEN

Primary ciliary dyskinesia (PCD) is a rare genetic disorder that alters mucociliary clearance, with consequent chronic disease of upper and lower airways. Diagnosis of PCD is challenging, and genetic testing is hampered by the high heterogeneity of the disease, because autosomal recessive causative mutations were found in 34 different genes. In this study, we clinically and molecularly characterized a cohort of 51 Italian patients with clinical signs of PCD. A custom next-generation sequencing panel that enables the affordable and simultaneous screening of 24 PCD genes was developed for genetic analysis. After variant filtering and prioritization, the molecular diagnosis of PCD was achieved in 43% of the patients. Overall, 5 homozygous and 27 compound heterozygous mutations, 21 of which were never reported before, were identified in 11 PCD genes. The DNAH5 and DNAH11 genes were the most common cause of PCD in Italy, but some population specificities were identified. In addition, the number of unsolved cases and the identification of only a single mutation in six patients suggest further genetic heterogeneity and invoke the need of novel strategies to detect unconventional pathogenic DNA variants. Finally, despite the availability of mutation databases and in silico prediction tools helping the interpretation of variants in next-generation sequencing screenings, a comprehensive segregation analysis is required to establish the in trans inheritance and support the pathogenic role of mutations.


Asunto(s)
Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Síndrome de Kartagener/diagnóstico , Síndrome de Kartagener/genética , Adolescente , Adulto , Anciano , Dineínas Axonemales/genética , Biomarcadores , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Lactante , Italia , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Adulto Joven
10.
Neuromuscul Disord ; 15(12): 858-62, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16288874

RESUMEN

We report here clinical, electrophysiological, and molecular findings in a family affected with two inherited genetic diseases: limb girdle muscular dystrophy type 1B (LGMD1B) and hereditary neuropathy with liability to pressure palsies (HNPP). Members of the family carry a novel missense mutation in the LMNA gene and a nonsense mutation in the PMP22 gene. Interestingly, the double LMNA/PMP22 mutations carriers showed clinical features more severe than usually seen in HNPP, and electrophysiological findings suggesting an axonal loss in addition to a typical myelinopathy. This study provides further insights into the relevance of lamin A/C in muscle and nerve.


Asunto(s)
Salud de la Familia , Neuropatía Hereditaria Motora y Sensorial/genética , Lamina Tipo A/genética , Distrofia Muscular de Cinturas/genética , Mutación , Proteínas de la Mielina/genética , Anciano , Análisis Mutacional de ADN/métodos , Femenino , Neuropatía Hereditaria Motora y Sensorial/patología , Neuropatía Hereditaria Motora y Sensorial/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología , Distrofia Muscular de Cinturas/fisiopatología , Conducción Nerviosa/fisiología
11.
Am J Cardiol ; 116(8): 1245-51, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310507

RESUMEN

Dominant mutations in desmocollin-2 (DSC2) gene cause arrhythmogenic cardiomyopathy (ACM), a progressive heart muscle disease characterized by ventricular tachyarrhythmias, heart failure, and risk of juvenile sudden death. Recessive mutations are rare and are associated with a cardiac or cardiocutaneous phenotype. Here, we evaluated the impact of a homozygous founder DSC2 mutation on clinical expression of ACM. An exon-by-exon analysis of the DSC2 coding region was performed in 94 ACM index patients. The c.536A>G (p.D179G) mutation was identified in 5 patients (5.3%), 4 of which resulted to be homozygous carriers. The 5 subjects shared a conserved haplotype, strongly indicating a common founder. Genetic and clinical investigation of probands' families revealed that p.D179G homozygous carriers displayed severe forms of biventricular cardiomyopathy without hair or skin abnormalities. The only heterozygous proband, who carried an additional variant of unknown significance in αT-catenin gene, showed a mild form of ACM without left ventricular involvement. All heterozygous family members were clinically asymptomatic. In conclusion, this is the first homozygous founder mutation in DSC2 gene identified among Italian ACM probands. Our findings provide further evidence of the occurrence of recessive DSC2 mutations in patients with ACM predominantly presenting with biventricular forms of the disease.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Desmocolinas/genética , Mutación/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Exones/genética , Femenino , Efecto Fundador , Homocigoto , Humanos , Italia , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
12.
Neuromolecular Med ; 16(3): 540-50, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24819634

RESUMEN

Charcot-Marie-Tooth (CMT) diseases include a group of clinically heterogeneous inherited neuropathies subdivided into demyelinating (CMT1), axonal (CMT2) and intermediate CMT forms. CMTs are associated with different genes, although mutations in some of these genes may cause both clinical pictures. To date, more than 50 CMT genes have been identified, but more than half of the cases are due to mutations in MFN2, MPZ, GJB1 and PMP22. The aim of this study was to estimate the frequency of disease mutations of these four genes in the axonal form of CMT in order to evaluate their effectiveness in the molecular diagnosis of CMT2 patients. A cohort of 38 CMT2 Italian subjects was screened for mutations in the MFN2, MPZ and GJB1 genes by direct sequencing and for PMP22 rearrangements using the MLPA technique. Overall, we identified 15 mutations, 8 of which were novel: 11 mutations (28.9 %) were in the MFN2 gene, 2 (5.3 %) in MPZ and 2 (5.3 %) in PMP22. No mutations were found in GJB1. Two patients showed rearrangements in the PMP22 gene, which is commonly associated with CMT1 or HNPP phenotypes thus usually not tested in CMT2 patients. By including this gene in the analysis, we reached a molecular diagnosis rate of 39.5 %, which is one of the highest reported in the literature. Our findings confirm the MFN2 gene as the most common cause of CMT2 and suggest that PMP22 rearrangements should be considered in the molecular diagnosis of CMT2 patients.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Conexinas/genética , GTP Fosfohidrolasas/genética , Proteínas Mitocondriales/genética , Proteína P0 de la Mielina/genética , Proteínas de la Mielina/genética , Adolescente , Adulto , Edad de Inicio , Enfermedad de Charcot-Marie-Tooth/epidemiología , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Duplicación de Gen , Genes Dominantes , Genes Recesivos , Genotipo , Humanos , Lactante , Italia/epidemiología , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Fenotipo , Eliminación de Secuencia , Evaluación de Síntomas , Proteína beta1 de Unión Comunicante
13.
Neurobiol Aging ; 35(5): 1212.e7-1212.e10, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24325798

RESUMEN

Amyotrophic lateral sclerosis (ALS) is as an adult-onset neurodegenerative disorder involving both upper and lower motor neurons. About 5% of all cases exhibit signs of frontotemporal degeneration (FTD). We established the mutation frequency of C9ORF72, SOD1, TARDBP, and FUS genes in 307 patients with sporadic ALS, 46 patients with familial ALS (FALS), and 73 patients affected with FTD, all originating from the northeastern part of Italy. C9ORF72 pathogenic expansion was found on 22% of familial ALS, 5% of sporadic ALS, and 14% of FTD patients, resulting the most frequently genetic determinant in our cohort. Sequence analysis of ALS cohort identified 2 novel variants on SOD1 (p.Glu41Gly) and FUS (p.Gly496Glyfs*31). Interestingly, the single base deletion on FUS was observed in an homozygous state, suggesting a recessive pattern of inheritance. No point mutations were identified on FTD cohort. Although useful to direct genetic testing, this study results expand the current knowledge of ALS genetics.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Tasa de Mutación , Mutación , Proteína FUS de Unión a ARN/genética , Superóxido Dismutasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteína C9orf72 , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Femenino , Degeneración Lobar Frontotemporal/genética , Eliminación de Gen , Homocigoto , Humanos , Italia , Masculino , Persona de Mediana Edad , Proteínas/genética , Superóxido Dismutasa-1 , Adulto Joven
14.
Eur J Hum Genet ; 21(11): 1226-31, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23486541

RESUMEN

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a primary heart muscle disease characterized by progressive myocardial loss, with fibro-fatty replacement, and high frequency of ventricular arrhythmias that can lead to sudden cardiac death. ARVC is a genetically determined disorder, usually caused by point mutations in components of the cardiac desmosome. Conventional mutation screening of ARVC genes fails to detect causative mutations in about 50% of index cases, suggesting a further genetic heterogeneity. We performed a genome-wide linkage study and a copy number variations (CNVs) analysis, using high-density SNP arrays, in an ARVC family showing no mutations in any of the desmosomal genes. The CNVs analysis identified a heterozygous deletion of about 122 kb on chromosome 12p11.21, including the entire plakophilin-2 gene and shared by all affected family members. It was not listed on any of available public CNVs databases and was confirmed by quantitative real-time PCR. This is the first SNP array-based genome-wide study leading to the identification of a CNV segregating with the disease phenotype in an ARVC family. This result underscores the importance of performing additional analysis for possible genomic deletions/duplications in ARVC patients without point mutations in known disease genes.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Eliminación de Gen , Placofilinas/genética , Adulto , Anciano , Anciano de 80 o más Años , Displasia Ventricular Derecha Arritmogénica/diagnóstico por imagen , Cromosomas Humanos Par 12/genética , Variaciones en el Número de Copia de ADN , Familia , Femenino , Dosificación de Gen/genética , Ligamiento Genético , Humanos , Masculino , Persona de Mediana Edad , Linaje , Reacción en Cadena en Tiempo Real de la Polimerasa , Ultrasonografía , Adulto Joven
15.
Neuromuscul Disord ; 21(1): 58-67, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20951042

RESUMEN

The development of new animal models is a crucial step in determining the pathological mechanism underlying neurodegenerative diseases and is essential for the development of effective therapies. We have investigated the zebrafish (Danio rerio) as a new model to study CMT2A, a peripheral neuropathy characterized by the selective loss of motor neurons, caused by mutations of mitofusin 2 gene. Using a knock-down approach, we provide evidence that during embryonic development, mitofusin 2 loss of function is responsible of several morphological defects and motility impairment. Immunohistochemical investigations, revealing the presence of severe alterations in both motor neurons and muscles fibres, indicated the central role played by MFN2 in axonal and neuromuscular development. Finally, we demonstrated the ability of human MFN2 to balance the downregulation of endogenous mfn2 in zebrafish, further supporting the conserved function of the MFN2 gene. These results highlight the essential role of mitofusin 2 in the motor axon development and demonstrate the potential of zebrafish as a suitable and complementary platform for dissecting pathogenetic mechanisms of MFN2 mutations in vivo.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Discapacidades del Desarrollo/etiología , Regulación hacia Abajo/efectos de los fármacos , Enfermedades Neuromusculares/etiología , Oligodesoxirribonucleótidos Antisentido/efectos adversos , Metaloproteinasas Similares a Tolloid/genética , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Enfermedad de Charcot-Marie-Tooth/inducido químicamente , Enfermedad de Charcot-Marie-Tooth/complicaciones , Enfermedad de Charcot-Marie-Tooth/genética , Biología Computacional , Discapacidades del Desarrollo/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Embrión no Mamífero , GTP Fosfohidrolasas , Humanos , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Enfermedades Neuromusculares/genética , ARN Mensajero/metabolismo , Receptores Colinérgicos/metabolismo , Metaloproteinasas Similares a Tolloid/metabolismo , Tubulina (Proteína)/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
16.
J Child Neurol ; 26(1): 49-57, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21212451

RESUMEN

Mutations in the ganglioside-induced differentiation-associated protein 1 (GDAP1) gene may cause severe early-onset inherited neuropathies. Here, the authors report a clinical and neurophysiological follow-up of a Pakistani child with a very early-onset neuropathy carrying a novel homozygous mutation in the GDAP1gene. They discuss the relationship between the several forms of Charcot-Marie-Tooth disease presenting in the first months of life and focus on the literature of GDAP1-associated early-onset neuropathy. This case further expands on the clinical spectrum and the genetic heterogeneity of early-onset inherited neuropathy due to GDAP1 gene mutations.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Proteínas del Tejido Nervioso/genética , Niño , Preescolar , Homocigoto , Humanos , Lactante , Masculino , Mutación , Examen Neurológico , Polimorfismo de Nucleótido Simple
17.
J Neurol Sci ; 294(1-2): 124-6, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20447653

RESUMEN

X-linked hydrocephalus, MASA syndrome, X-linked complicated Spastic Paraplegia Type I and X-linked partial agenesis of the corpus callosum are the four rare diseases usually referred to L1 syndrome, caused by mutations in the L1CAM gene. By direct sequencing of L1CAM in 16 patients, we were able to identify seven mutations, five of which were never described before. Patients' phenotype evaluation revealed a correlation between the number of clinical features typical of L1 syndrome and the chance to find causative mutation. Our findings support that L1CAM mutations are associated with widely heterogeneous phenotypes, however the occurrence of several clinical features remains the best criterion for planning molecular testing both in familial and apparently sporadic cases.


Asunto(s)
Agenesia del Cuerpo Calloso , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Hidrocefalia/genética , Mutación , Molécula L1 de Adhesión de Célula Nerviosa/genética , Paraplejía Espástica Hereditaria/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/genética , Persona de Mediana Edad , Fenotipo , Síndrome , Pulgar/anomalías , Adulto Joven
18.
BMJ Case Rep ; 20092009.
Artículo en Inglés | MEDLINE | ID: mdl-21686698

RESUMEN

Charcot-Marie-Tooth (CMT) disease is the most common hereditary neuropathy. CMT falls into two main forms: the demyelinating CMT type 1 with decreased nerve conduction velocities and the axonal CMT type 2. CMT2 is further subtyped by linkage analysis into >10 loci, with eight genes identified.Recently, mutations in the mitochondrial fusion protein 2 (MFN2) gene were reported in families with CMT2A1 and additional mutations have been detected in other studies, bringing to 42 the total number of different MFN2 mutations described thus far.2(-)4In the current study, we report a novel MFN2 mutation shared by two apparently unrelated CMT2 families originating from the same area in Southern Italy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA