Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Med Genet ; 60(1): 99-105, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35264407

RESUMO

BACKGROUND: Copy number variants (CNVs) associated with late-onset medical conditions are rare but important secondary findings in chromosomal microarray analysis (CMA) performed during pregnancy. Here, we critically review the cases at two tertiary centres to assess the criteria which guide the disclosure of such findings and develop a disclosure decision tool (DDT) aimed at facilitating disclosure decision. Parental decisions on receiving CNVs associated with risks for late-onset conditions were also recorded. METHODS: Prenatal CMAs in Hadassah and Shaare Zedek Medical Centers from November 2013 to October 2021 were reviewed for CNVs associated with late-onset conditions. The DDT proposed uses a five-parameter scoring system, which considers the severity, median age of onset, penetrance, understanding of genotype-phenotype correlation and actionability of the finding. RESULTS: Out of 16 238 prenatal CMAs, 16 (0.1%) harboured CNVs associated with late-onset conditions, 15 of which were disclosed. Outcome information was available on 13 of the 16 pregnancies, all of which continued to delivery. CONCLUSIONS: Our suggested DDT will help clinicians to quantitatively weigh the variables associated with CNVs of this type and arrive at a well thought out clinical decision regarding disclosure. Although the prevalence of late-onset conditions as a major finding in the prenatal setup is low, it is expected to rise with the increasing use of non-invasive CMA testing and whole exome and genome sequencing.


Assuntos
Aberrações Cromossômicas , Diagnóstico Pré-Natal , Feminino , Humanos , Gravidez , Revelação , Variações do Número de Cópias de DNA/genética , Análise em Microsséries , Resultado da Gravidez
2.
J Assist Reprod Genet ; 32(6): 887-91, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25899990

RESUMO

PURPOSE: To determine the molecular basis of familial, autosomal-recessive, non-obstructive azoospermia in a consanguineous Iranian Jewish family. METHODS: We investigated the genetic cause of non-obstructive azoospermia in two affected siblings from a consanguineous family. Homozygosity mapping in the DNA samples of the patients and their normospermic brother was followed by exome analysis of one of the patients. Other family members were genotyped for the mutation by Sanger sequencing. The mutation effect was demonstrated by immunostaining of the patients' testicular tissue. RESULTS: The two patients were homozygous for a splice site mutation in SYCE1 which resulted in retention of intron three in the cDNA and premature stop codon. SYCE1 encodes a Synaptonemal Complex protein which plays an essential role during meiosis. Immunostaining of patient's testicular tissue with anti-Syce1 antibody revealed an undetectable level of Syce1. Histological examination of the patients' tissue disclosed immature-stages spermatocytes without mature forms, indicating maturation arrest. CONCLUSION: The significance of most synaptonemal complex proteins was previously demonstrated in a mutant mouse model. The present report underscores the importance of synaptonemal complex proteins in spermatogenenesis in humans. Our new approach, combining homozygosity mapping and exome sequencing, resulted in one of the first reports of an autosomal-recessive form of NOA.


Assuntos
Azoospermia/genética , Códon sem Sentido , Proteínas Nucleares/genética , Consanguinidade , Proteínas de Ligação a DNA , Homozigoto , Humanos , Masculino , Mutação , Proteínas Nucleares/química , Linhagem , Sítios de Splice de RNA/genética
3.
Am J Med Genet A ; 161A(12): 3115-20, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24254849

RESUMO

Truncus arteriosus accounts for approximately 1% of congenital heart defects and the cause of isolated non-syndromic truncus arteriosus is largely unknown. In order to identify the underlying molecular defect in a consanguineous family with recurrent tuncus arteriosus, homozygosity mapping followed by whole exome sequencing was performed. This resulted in the identification of a homozygous mutation, Arg1299Cys, in the PLXND1 gene. The mutation affected a highly conserved residue, segregated with the disease in the family and was absent from available SNP databases and ethnic matched controls. in silico comparative modeling revealed that the mutation resides in the N-terminal segment of the human plexin-D1 intracellular region which interacts with the catalytic GTPase-activating protein homology region. The mutation likely destabilizes the intracellular region, perturbing its anchoring and catalytic activity. The phenotype in human PLXND1 mutation is closely related to that of knockout mice for PLXND1, its co-receptor neuropilin-1 or its ligand SEMA3C. It is therefore suggested that SEMA3C signaling, propagated through the heterodimer receptor plexin-D1/neuropilin, is important for truncus arteriosus septation. Confirmation of this observation will require the identification of PLXND1 mutations in additional patients. Exome analysis is valuable for molecular investigation of single patients with congenital heart defects in whom chromosomal copy number variants have been excluded.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Estudos de Associação Genética , Cardiopatias Congênitas/genética , Tronco Arterial/fisiopatologia , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Pré-Escolar , Mapeamento Cromossômico , Exoma , Feminino , Cardiopatias Congênitas/fisiopatologia , Humanos , Lactente , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Glicoproteínas de Membrana , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Linhagem , Semaforinas/metabolismo , Análise de Sequência de DNA , Transdução de Sinais
4.
Blood ; 121(25): 5078-87, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23599270

RESUMO

Severe congenital neutropenia as well as primary myelofibrosis are rare in infancy. Elucidation of the underlying mechanism is important because it extends our understanding of the more common adult forms of these disorders. Using homozygosity mapping followed by exome sequencing, we identified a Thr224Asn mutation in the VPS45 gene in infants from consanguineous families who suffered from life-threatening neutropenia, which was refractory to granulocyte CSF, from defective platelet aggregation and myelofibrosis. The mutation segregated in the families, was not present in controls, affected a highly conserved codon, and apparently destabilized the Vps45 protein, which was reduced in the patients' leukocytes. Introduction of the corresponding mutation into yeast resulted in reduced cellular levels of Vps45 and also of the cognate syntaxin Tlg2, which is required for membrane traffic through the endosomal system. A defect in the endosomal-lysosomal pathway, the homologous system in humans, was suggested by the absence of lysosomes in the patients' fibroblasts and by the depletion of α granules in their platelets. Importantly, accelerated apoptosis was observed in the patients' neutrophils and bone marrow. This is the first report of a Vps45-related disease in humans, manifesting by neutropenia, thrombasthenia, myelofibrosis, and progressive bone marrow failure.


Assuntos
Mutação , Neutropenia/congênito , Mielofibrose Primária/genética , Proteínas de Transporte Vesicular/genética , Sequência de Bases , Western Blotting , Síndrome Congênita de Insuficiência da Medula Óssea , Consanguinidade , Feminino , Imunofluorescência , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Neutropenia/genética , Linhagem
5.
J Med Genet ; 50(2): 118-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23339110

RESUMO

BACKGROUND: Early infantile epileptic encephalopathies usually manifest as severely impaired cognitive and motor development and often result in a devastating permanent global developmental delay and intellectual disability. A large set of genes has been implicated in the aetiology of this heterogeneous group of disorders. Among these, the ion channelopathies play a prominent role. In this study, we investigated the genetic cause of infantile epilepsy in three affected siblings. METHODS AND RESULTS: Homozygosity mapping in DNA samples followed by exome analysis in one of the patients resulted in the identification of a homozygous mutation, p.L1040P, in the CACNA2D2 gene. This gene encodes the auxiliary α(2)δ2 subunit of high voltage gated calcium channels. The expression of the α(2)δ2-L1040P mutant instead of α(2)δ2 wild-type (WT) in Xenopus laevis oocytes was associated with a notable reduction of current density of both N (Ca(V)2.2) and L (Ca(V)1.2) type calcium channels. Western blot and confocal imaging analyses showed that the α(2)δ2-L1040P mutant was synthesised normally in oocyte but only the α(2)δ2-WT, and not the α(2)δ2-L1040P mutant, increased the expression of α(1B), the pore forming subunit of Ca(V)2.2, at the plasma membrane. The expression of α(2)δ2-WT with Ca(V)2.2 increased the surface expression of α(1B) 2.5-3 fold and accelerated current inactivation, whereas α(2)δ2-L1040P did not produce any of these effects. CONCLUSIONS: L1040P mutation in the CACNA2D2 gene is associated with dysfunction of α(2)δ2, resulting in reduced current density and slow inactivation in neuronal calcium channels. The prolonged calcium entry during depolarisation and changes in surface density of calcium channels caused by deficient α(2)δ2 could underlie the epileptic phenotype. This is the first report of an encephalopathy caused by mutation in the auxiliary α(2)δ subunit of high voltage gated calcium channels in humans, illustrating the importance of this subunit in normal physiology of the human brain.


Assuntos
Canais de Cálcio/genética , Canalopatias/genética , Mutação , Animais , Western Blotting , Canais de Cálcio/metabolismo , Canalopatias/metabolismo , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Oócitos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
6.
Blood ; 121(1): 129-35, 2013 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-23149847

RESUMO

CD59 deficiency is a common finding in RBCs and WBCs in patients with chronic hemolysis suffering from paroxysmal nocturnal hemoglobinuria in which the acquired mutation in the PIGA gene leads to membrane loss of glycosylphosphatidylinositol-anchored membrane proteins, including CD59. The objective of the present study was to elucidate the molecular basis of childhood familial chronic Coombs-negative hemolysis and relapsing polyneuropathy presenting as chronic inflammatory demyelinating polyradiculoneuropathy in infants of North-African Jewish origin from 4 unrelated families. A founder mutation was searched for using homozygosity mapping followed by exome sequencing. The expression of CD59, CD55, and CD14 was examined in blood cells by flow cytometry followed by Western blot of the CD59 protein. A homozygous missense mutation, p.Cys89Tyr in CD59, was identified in all patients. The mutation segregated with the disease in the families and had a carrier rate of 1:66 among Jewish subjects of North-African origin. The mutated protein was present in the patients' cells in reduced amounts and was undetectable on the membrane surface. Based on the results of the present study, we conclude that the Cys89Tyr mutation in CD59 is associated with a failure of proper localization of the CD59 protein in the cell surface. This mutation is manifested clinically in infancy by chronic hemolysis and relapsing peripheral demyelinating disease.


Assuntos
Anemia Hemolítica/genética , Antígenos CD59/genética , Hemoglobinúria/genética , Mutação de Sentido Incorreto , Mutação Puntual , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/genética , Idade de Início , Sequência de Aminoácidos , Anemia Hemolítica/sangue , Anemia Hemolítica/líquido cefalorraquidiano , Anemia Hemolítica/etnologia , Antígenos CD59/metabolismo , Pré-Escolar , Feminino , Efeito Fundador , Hemoglobinúria/sangue , Hemoglobinúria/líquido cefalorraquidiano , Hemoglobinúria/etnologia , Humanos , Lactente , Judeus/genética , Líbia/etnologia , Masculino , Proteínas de Membrana/análise , Dados de Sequência Molecular , Marrocos/etnologia , Linhagem , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/sangue , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/líquido cefalorraquidiano , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/etnologia , Transporte Proteico
7.
Epilepsia ; 54(2): e24-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23252400

RESUMO

West syndrome consists of infantile spasms, hypsarrhythmia, and developmental arrest. Most patients remain mentally retarded and many develop Lennox-Gastaut syndrome. Using homozygosity mapping followed by exome sequencing we identified an ST3GAL3 mutation in three infants with West syndrome. ST3GAL3 encodes a sialyltransferase involved in the biosynthesis of sialyl-Lewis epitopes on cell surface-expressed glycoproteins. The mutation affected an essential sialyl-motif and abolished enzymatic activity. Abnormalities in proteins involved in forebrain γ-aminobutyric acid (GABA)ergic synaptic growth and function were recently proposed to account for infantile spasms. Dysfunctional ST3GAL3 may thus result in perturbation of the posttranslational sialylation of proteins in these pathways.


Assuntos
Sialiltransferases/deficiência , Espasmos Infantis/genética , Adolescente , Idade de Início , Animais , Anticonvulsivantes/uso terapêutico , Células CHO , Criança , Pré-Escolar , Cricetinae , Cricetulus , Análise Mutacional de DNA , Eletroencefalografia , Epilepsia/tratamento farmacológico , Epilepsia/etiologia , Epitopos/genética , Éxons/genética , Feminino , Ligação Genética , Humanos , Lactente , Deficiência Intelectual/epidemiologia , Síndrome de Lennox-Gastaut , Masculino , Linhagem , Espasmos Infantis/epidemiologia , Ácido gama-Aminobutírico/fisiologia , beta-Galactosídeo alfa-2,3-Sialiltransferase
8.
PLoS One ; 7(5): e36458, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563501

RESUMO

Parkinson disease is caused by neuronal loss in the substantia nigra which manifests by abnormality of movement, muscle tone, and postural stability. Several genes have been implicated in the pathogenesis of Parkinson disease, but the underlying molecular basis is still unknown for ∼70% of the patients. Using homozygosity mapping and whole exome sequencing we identified a deleterious mutation in DNAJC6 in two patients with juvenile parkinsonism. The mutation was associated with abnormal transcripts and marked reduced DNAJC6 mRNA level. DNAJC6 encodes the HSP40 Auxilin, a protein which is selectively expressed in neurons and confers specificity to the ATPase activity of its partner Hcs70 in clathrin uncoating. In Auxilin null mice it was previously shown that the abnormally increased retention of assembled clathrin on vesicles and in empty cages leads to impaired synaptic vesicle recycling and perturbed clathrin mediated endocytosis. Endocytosis function, studied by transferring uptake, was normal in fibroblasts from our patients, likely because of the presence of another J-domain containing partner which co-chaperones Hsc70-mediated uncoating activity in non-neuronal cells. The present report underscores the importance of the endocytic/lysosomal pathway in the pathogenesis of Parkinson disease and other forms of parkinsonism.


Assuntos
Auxilinas/genética , Proteínas de Choque Térmico HSP40/genética , Mutação , Transtornos Parkinsonianos/genética , Adolescente , Auxilinas/metabolismo , Sequência de Bases , Clatrina/metabolismo , Análise Mutacional de DNA/métodos , Saúde da Família , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Masculino , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neurônios/metabolismo , Linhagem
9.
J Med Genet ; 49(4): 221-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22499339

RESUMO

BACKGROUND: Osteopetrosis is a life-threatening, rare disorder typically resulting from osteoclast dysfunction and infrequently from failure to commitment to osteoclast lineage. Patients commonly present in infancy with macrocephaly, feeding difficulties, evolving blindness and deafness, and bone marrow failure. In ∼70% of the patients there is a molecularly defined failure to maintain an acid pH at the osteoclast-bone interface (the ruffled border) which is necessary for the bone resorptive activity. METHODS AND RESULTS: In eight patients with infantile osteopetrosis which could be cured by bone marrow transplantation, the study identified by homozygosity mapping in distantly related consanguineous pedigrees a missense mutation in a highly conserved residue in the SNX10 gene. The mutation segregated with the disease in the families and was carried by one of 211 anonymous individuals of the same ethnicity. In the patients' osteoclasts, the mutant SNX10 protein was abnormally abundant and its distribution altered. The patients' osteoclasts were fewer and smaller than control cells, their resorptive capacity was markedly deranged, and the endosomal pathway was perturbed as evidenced by the distribution of internalised dextran. CONCLUSIONS: SNX10 was recently shown to interact with vacuolar type H(+)-ATPase (V-ATPase) which pumps protons at the osteoclast-bone interface. Mutations in TCIRG1, the gene encoding a subunit of the V-ATPase complex, account for the majority of cases of osteopetrosis. It is speculated that SNX10 is responsible for the vesicular sorting of V-ATPase from Golgi or for its targeting to the ruffled border. A mutation in SNX10 may therefore result in 'secondary V-ATPase deficiency' with a failure to acidify the resorption lacuna. Determination of the sequence of the SNX10 gene is warranted in molecularly undefined patients with recessive 'pure' osteopetrosis of infancy.


Assuntos
Mutação , Osteopetrose/genética , Nexinas de Classificação/genética , Sequência de Bases , Consanguinidade , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteopetrose/patologia , Linhagem , Polimorfismo de Nucleotídeo Único
10.
Am J Hum Genet ; 90(3): 518-23, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22405087

RESUMO

Degeneration of the cerebrum, cerebellum, and retina in infancy is part of the clinical spectrum of lysosomal storage disorders, mitochondrial respiratory chain defects, carbohydrate glycosylation defects, and infantile neuroaxonal dystrophy. We studied eight individuals from two unrelated families who presented at 2-6 months of age with truncal hypotonia and athetosis, seizure disorder, and ophthalmologic abnormalities. Their course was characterized by failure to acquire developmental milestones and culminated in profound psychomotor retardation and progressive visual loss, including optic nerve and retinal atrophy. Despite their debilitating state, the disease was compatible with survival of up to 18 years. Laboratory investigations were normal, but the oxidation of glutamate by muscle mitochondria was slightly reduced. Serial brain MRI displayed progressive, prominent cerebellar atrophy accompanied by thinning of the corpus callosum, dysmyelination, and frontal and temporal cortical atrophy. Homozygosity mapping followed by whole-exome sequencing disclosed a Ser112Arg mutation in ACO2, encoding mitochondrial aconitase, a component of the Krebs cycle. Specific aconitase activity in the individuals' lymphoblasts was severely reduced. Under restrictive conditions, the mutant human ACO2 failed to complement a yeast ACO1 deletion strain, whereas the wild-type human ACO2 succeeded, indicating that this mutation is pathogenic. Thus, a defect in mitochondrial aconitase is associated with an infantile neurodegenerative disorder affecting mainly the cerebellum and retina. In the absence of noninvasive biomarkers, determination of the ACO2 sequence or of aconitase activity in lymphoblasts are warranted in similarly affected individuals, based on clinical and neuroradiologic grounds.


Assuntos
Aconitato Hidratase/genética , Cerebelo/anormalidades , Mitocôndrias/enzimologia , Mutação , Doenças Neurodegenerativas/genética , Retina/anormalidades , Adolescente , Atrofia/enzimologia , Atrofia/genética , Cerebelo/enzimologia , Criança , Pré-Escolar , Exoma , Éxons , Feminino , Genótipo , Ácido Glutâmico/metabolismo , Heterozigoto , Homozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética/métodos , Masculino , Mitocôndrias/genética , Doenças Neurodegenerativas/enzimologia , Oxirredução , Polimorfismo de Nucleotídeo Único , Retina/enzimologia
11.
Mol Genet Metab ; 104(4): 517-20, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22019070

RESUMO

The identification of disease causing mutation in patients with neurodegenerative disorders originating from small, non-consanguineous families is challenging. Three siblings were found to have ventriculomegaly at early gestation; postnatally, there was no acquisition of developmental milestones, and the muscles of the children were dystrophic. Plasma and CSF lactate levels were normal, but the activities of mitochondrial complex I and IV were markedly decreased. Using linkage analysis in the family, followed by whole exome sequencing of a single patient, we identified a pathogenic mutation in the AIFM1 gene which segregated with the disease state and was absent in 86 anonymous controls. This is the second report of a mutation in the AIFM1 gene, extending the clinical spectrum to include prenatal ventriculomegaly and underscores the importance of AIF for complex I assembly. In summary, linkage analysis followed by exome sequencing of a single patient is a cost-effective approach for the identification of disease causing mutations in small non-consanguineous families.


Assuntos
Fator de Indução de Apoptose/genética , Exoma , Ligação Genética , Hidrocefalia/diagnóstico por imagem , Mutação de Sentido Incorreto , Sequência de Aminoácidos , Sequência de Bases , Sequência Conservada , Evolução Fatal , Feminino , Estudos de Associação Genética , Haplótipos , Humanos , Hidrocefalia/genética , Recém-Nascido , Masculino , Dados de Sequência Molecular , Gravidez , Análise de Sequência de DNA , Ultrassonografia Pré-Natal
12.
Am J Med Genet A ; 155A(11): 2801-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21990102

RESUMO

Diaphonospondylodysostosis (DSD) is a rare, recessively inherited, lethal skeletal dysplasia, characterized by severe spinal ossification, segmentation defects, and renal cystic dysplasia with nephrogenic rests. We hereby present three affected individuals: two children and a fetus from two unrelated East Jerusalem Arab-Muslim families. Whereas most fetuses die in utero or perinatally, one of the children survived to 15 months. Homozygosity mapping in the two families demonstrated a single common 3.87 Mb region on chromosome 7, ruling out previously known spondylocostal/spondylothoracic dysostosis loci. The 15 protein coding genes in the region were prioritized, and some were sequenced. A single, novel deleterious mutation, Q104X, was detected in the bone morphogenetic protein-binding endothelial regulator protein (BMPER) gene, recently reported to be mutated in other DSD patients [Funari et al., 2010]. The novel mutation we identified is an ancestral founder allele, as evidenced by a shared 440 SNP haplotype, and its frequency in the general Arab population is estimated to be <1:123. Our findings confirm loss of BMPER function as a cause of axial versus appendicular skeletal defects, and suggest that less deleterious mutations may be involved in milder axial skeleton abnormalities.


Assuntos
Proteínas de Transporte/genética , Cromossomos Humanos Par 17/genética , Mutação , Espondilose/genética , Árabes/genética , Mapeamento Cromossômico , Feminino , Efeito Fundador , Frequência do Gene , Aconselhamento Genético , Homozigoto , Humanos , Lactente , Recém-Nascido , Medição da Translucência Nucal , Linhagem , Polimorfismo de Nucleotídeo Único , Gravidez , Segundo Trimestre da Gravidez/genética , Radiografia , Espondilose/diagnóstico , Espondilose/diagnóstico por imagem
13.
Genome Res ; 21(5): 658-64, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21487076

RESUMO

Whole exome sequencing has become a pivotal methodology for rapid and cost-effective detection of pathogenic variations in Mendelian disorders. A major challenge of this approach is determining the causative mutation from a substantial number of bystander variations that do not play any role in the disease etiology. Current strategies to analyze variations have mainly relied on genetic and functional arguments such as mode of inheritance, conservation, and loss of function prediction. Here, we demonstrate that disease-network analysis provides an additional layer of information to stratify variations even in the presence of incomplete sequencing coverage, a known limitation of exome sequencing. We studied a case of Hereditary Spastic Paraparesis (HSP) in a single inbred Palestinian family. HSP is a group of neuropathological disorders that are characterized by abnormal gait and spasticity of the lower limbs. Forty-five loci have been associated with HSP and lesions in 20 genes have been documented to induce the disorder. We used whole exome sequencing and homozygosity mapping to create a list of possible candidates. After exhausting the genetic and functional arguments, we stratified the remaining candidates according to their similarity to the previously known disease genes. Our analysis implicated the causative mutation in the motor domain of KIF1A, a gene that has not yet associated with HSP, which functions in anterograde axonal transportation. Our strategy can be useful for a large class of disorders that are characterized by locus heterogeneity, particularly when studying disorders in single families.


Assuntos
Cinesinas/genética , Análise de Sequência de DNA/métodos , Paraplegia Espástica Hereditária/genética , Adolescente , Adulto , Sequência de Aminoácidos , Bases de Dados Genéticas , Éxons/genética , Genótipo , Homozigoto , Humanos , Masculino , Modelos Moleculares , Mutação , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Paraplegia Espástica Hereditária/patologia , Adulto Jovem
14.
Am J Hum Genet ; 87(5): 667-70, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20950787

RESUMO

Primary microcephaly of postnatal onset is a feature of many neurological disorders, mostly associated with mental retardation, seizures, and spasticity, and it typically carries a grave prognosis. Five infants from four unrelated families of Caucasus Jewish origin presented soon after birth with spasticity, epilepsy, and profound psychomotor retardation. Head circumference percentiles declined, and brain MRI disclosed marked cereberal and cerebellar atrophy with severe myelination defect. A search for a common homozygous region revealed a 2.28 Mb genomic segment on chromosome 11 that encompassed 16 protein-coding genes. A missense mutation in one of them, MED17, segregated with the disease state in the families and was carried by four of 79 anonymous Caucasus Jews. A corresponding mutation in the homologous S.cerevisiae gene SRB4 inactivated the protein, according to complementation assays. Screening of MED17 in additional patients with similar clinical and radiologic findings revealed four more patients, all homozygous for the p.L371P mutation and all originating from Caucasus Jewish families. We conclude that the p. L371P mutation in MED17 is a founder mutation in the Caucasus Jewish community and that homozygosity for this mutation is associated with infantile cerebral and cerebellar atrophy with poor myelination.


Assuntos
Atrofia/genética , Encéfalo/patologia , Cerebelo/patologia , Complexo Mediador/genética , Mutação , Cromossomos Humanos Par 11 , Efeito Fundador , Homozigoto , Humanos , Lactente , Judeus/genética , Microcefalia/genética , Doenças do Sistema Nervoso/genética
15.
Am J Hum Genet ; 86(1): 93-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20036350

RESUMO

Patients with Joubert syndrome 2 (JBTS2) suffer from a neurological disease manifested by psychomotor retardation, hypotonia, ataxia, nystagmus, and oculomotor apraxia and variably associated with dysmorphism, as well as retinal and renal involvement. Brain MRI results show cerebellar vermis hypoplasia and additional anomalies of the fourth ventricle, corpus callosum, and occipital cortex. The disease has previously been mapped to the centromeric region of chromosome 11. Using homozygosity mapping in 13 patients from eight Ashkenazi Jewish families, we identified a homozygous mutation, R12L, in the TMEM216 gene, in all affected individuals. Thirty individuals heterozygous for the mutation were detected among 2766 anonymous Ashkenazi Jews, indicating a carrier rate of 1:92. Given the small size of the TMEM216 gene relative to other JBTS genes, its sequence analysis is warranted in all JBTS patients, especially those who suffer from associated anomalies.


Assuntos
Mutação , Doenças do Sistema Nervoso/genética , Adolescente , Adulto , Alelos , Encéfalo/patologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Homozigoto , Humanos , Lactente , Judeus , Imageamento por Ressonância Magnética/métodos , Doenças do Sistema Nervoso/etnologia , Síndrome
16.
Genetics ; 179(4): 1989-99, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18689881

RESUMO

The Drosophila LIM-only (LMO) protein DLMO functions as a negative regulator of transcription during development of the fly wing. Here we report a novel role of DLMO as a positive regulator of transcription during the development of thoracic sensory bristles. We isolated new dlmo mutants, which lack some thoracic dorsocentral (DC) bristles. This phenotype is typical of malfunction of a thoracic multiprotein transcription complex, composed of CHIP, PANNIER (PNR), ACHAETE (AC), and DAUGHTERLESS (DA). Genetic interactions reveal that dlmo synergizes with pnr and ac to promote the development of thoracic DC bristles. Moreover, loss-of-function of dlmo reduces the expression of a reporter target gene of this complex in vivo. Using the GAL4-UAS system we also show that dlmo is spatially expressed where this complex is known to be active. Glutathione-S-transferase (GST)-pulldown assays showed that DLMO can physically bind CHIP and PNR through either of the two LIM domains of DLMO, suggesting that DLMO might function as part of this transcription complex in vivo. We propose that DLMO exerts its positive effect on DC bristle development by serving as a bridging molecule between components of the thoracic transcription complex.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila/crescimento & desenvolvimento , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Transcrição Gênica , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Tórax/embriologia , Tórax/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA