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











Base de dados
Intervalo de ano de publicação
1.
Neuropediatrics ; 51(1): 72-75, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31627234

RESUMO

Muscular dystrophy-dystroglycanopathies (MDDG) are a group of genetically heterogeneous autosomal recessive disorders characterized by hypoglycosylation of α-dystroglycan. Here, we report on two female patients from a consanguineous Lebanese family that presented in early infancy with generalized muscle hypotonia and primary microcephaly. Brain magnetic resonance imaging (MRI) showed different degrees of hypoplasia of the cerebellar vermis and hypoplasia of corpus callosum. Muscle biopsy analyses revealed a muscular dystrophy with reduced expression of α-dystroglycan and merosin in immunoblot analyses. Homozygosity mapping failed to elucidate the causal mutation due to the accepted notion that, in consanguineous families, homozygote mutations cause disease. However, by applying whole exome sequencing, we identified a novel compound heterozygous POMT1 mutation that segregates with the phenotype and is in line with the clinical presentation. This underscores that a less expected compound heterozygous instead of homozygous mutation in a consanguineous marriage results in a recessive disorder and highlights the growing role of next generation sequencing in neuromuscular disorder diagnostics.


Assuntos
Deficiências do Desenvolvimento/etiologia , Manosiltransferases/genética , Microcefalia/etiologia , Distrofias Musculares/congênito , Distrofias Musculares/genética , Criança , Consanguinidade , Evolução Fatal , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Distrofias Musculares/complicações , Linhagem , Síndrome de Wolff-Parkinson-White/genética
2.
Hum Mutat ; 38(6): 621-636, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28236339

RESUMO

Intellectual disability (ID) is the hallmark of an extremely heterogeneous group of disorders that comprises a wide variety of syndromic and non-syndromic phenotypes. Here, we report on mutations in two aminoacyl-tRNA synthetases that are associated with ID in two unrelated Iranian families. In the first family, we identified a homozygous missense mutation (c.514G>A, p.Asp172Asn) in the cytoplasmic seryl-tRNA synthetase (SARS) gene. The mutation affects the enzymatic core domain of the protein and impairs its enzymatic activity, probably leading to reduced cytoplasmic tRNASer concentrations. The mutant protein was predicted to be unstable, which could be substantiated by investigating ectopic mutant SARS in transfected HEK293T cells. In the second family, we found a compound heterozygous genotype of the mitochondrial tryptophanyl-tRNA synthetase (WARS2) gene, comprising a nonsense mutation (c.325delA, p.Ser109Alafs*15), which very likely entails nonsense-mediated mRNA decay and a missense mutation (c.37T>G, p.Trp13Gly). The latter affects the mitochondrial localization signal of WARS2, causing protein mislocalization. Including AIMP1, which we have recently implicated in the etiology of ID, three genes with a role in tRNA-aminoacylation are now associated with this condition. We therefore suggest that the functional integrity of tRNAs in general is an important factor in the development and maintenance of human cognitive functions.


Assuntos
Aminoacil-tRNA Sintetases/genética , Deficiência Intelectual/genética , Degradação do RNAm Mediada por Códon sem Sentido/genética , Adolescente , Adulto , Criança , Citocinas/genética , Feminino , Células HEK293 , Homozigoto , Humanos , Deficiência Intelectual/patologia , Irã (Geográfico) , Masculino , Mutação de Sentido Incorreto/genética , Proteínas de Neoplasias/genética , Linhagem , Proteínas de Ligação a RNA/genética
3.
Eur J Hum Genet ; 24(3): 392-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26173967

RESUMO

AIMP1/p43 is a multifunctional non-catalytic component of the multisynthetase complex. The complex consists of nine catalytic and three non-catalytic proteins, which catalyze the ligation of amino acids to their cognate tRNA isoacceptors for use in protein translation. To date, two allelic variants in the AIMP1 gene have been reported as the underlying cause of autosomal recessive primary neurodegenerative disorder. Here, we present two consanguineous families from Pakistan and Iran, presenting with moderate to severe intellectual disability, global developmental delay, and speech impairment without neurodegeneration. By the combination of homozygosity mapping and next generation sequencing, we identified two homozygous missense variants, p.(Gly299Arg) and p.(Val176Gly), in the gene AIMP1 that co-segregated with the phenotype in the respective families. Molecular modeling of the variants revealed deleterious effects on the protein structure that are predicted to result in reduced AIMP1 function. Our findings indicate that the clinical spectrum for AIMP1 defects is broader than witnessed so far.


Assuntos
Citocinas/genética , Genes Recessivos , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Mutação de Sentido Incorreto/genética , Proteínas de Neoplasias/genética , Degeneração Neural/complicações , Degeneração Neural/genética , Proteínas de Ligação a RNA/genética , Adulto , Sequência de Aminoácidos , Criança , Simulação por Computador , Citocinas/química , Exoma/genética , Família , Feminino , Haplótipos/genética , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/química , Linhagem , Estrutura Secundária de Proteína , Proteínas de Ligação a RNA/química , Reprodutibilidade dos Testes , Adulto Jovem
4.
Hum Mol Genet ; 24(20): 5697-710, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26206890

RESUMO

Histamine (HA) acts as a neurotransmitter in the brain, which participates in the regulation of many biological processes including inflammation, gastric acid secretion and neuromodulation. The enzyme histamine N-methyltransferase (HNMT) inactivates HA by transferring a methyl group from S-adenosyl-l-methionine to HA, and is the only well-known pathway for termination of neurotransmission actions of HA in mammalian central nervous system. We performed autozygosity mapping followed by targeted exome sequencing and identified two homozygous HNMT alterations, p.Gly60Asp and p.Leu208Pro, in patients affected with nonsyndromic autosomal recessive intellectual disability from two unrelated consanguineous families of Turkish and Kurdish ancestry, respectively. We verified the complete absence of a functional HNMT in patients using in vitro toxicology assay. Using mutant and wild-type DNA constructs as well as in silico protein modeling, we confirmed that p.Gly60Asp disrupts the enzymatic activity of the protein, and that p.Leu208Pro results in reduced protein stability, resulting in decreased HA inactivation. Our results highlight the importance of inclusion of HNMT for genetic testing of individuals presenting with intellectual disability.


Assuntos
Genes Recessivos , Histamina N-Metiltransferase/genética , Deficiência Intelectual/genética , Mutação de Sentido Incorreto , Adolescente , Adulto , Sequência de Aminoácidos , Domínio Catalítico , Criança , Pré-Escolar , Simulação por Computador , Análise Mutacional de DNA , Exoma , Feminino , Histamina N-Metiltransferase/metabolismo , Humanos , Lactente , Deficiência Intelectual/enzimologia , Iraque , Masculino , Dados de Sequência Molecular , Linhagem , Alinhamento de Sequência , Turquia , População Branca/genética
5.
Arch Iran Med ; 18(3): 179-84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25773692

RESUMO

BACKGROUND: Knowledge of the genes responsible for intellectual disability, particularly autosomal recessive forms, is rapidly expanding. Increasing numbers of the gene show great heterogeneity and supports the hypothesis that human genome may contain over 2000 causative genes with a critical role in brain development. METHODS: Since 2004, we have applied genome-wide SNP genotyping and next-generation sequencing in large consanguineous Iranian families with intellectual disability, to identify the genes harboring disease-causing mutations. The current study paved the way for identification of responsible genes in two unrelated Iranian families. RESULTS: We found two novel nonsense mutations, p.C77* and p.Q115*, in the calpain catalytic domain of CAPN10, which is a cysteine protease known to be involved in pathogenesis of noninsulin-dependent diabetes mellitus. Another different mutation in this gene (p.S138_R139ins5) has previously been reported in an Iranian family. All of these patients have common clinical features in spite of specific brain structural abnormalities on MRI. CONCLUSIONS: Different mutations in CAPN10 have already been found in three independent Iranian families. These results have strongly supported the possible role of CAPN10 in human brain development. Altogether, we proposed CAPN10 as a promising candidate gene for intellectual disability, which should be considered in diagnostic gene panels.


Assuntos
Calpaína/genética , Códon sem Sentido , Exoma , Deficiência Intelectual/genética , Adolescente , Adulto , Encéfalo/patologia , Calpaína/fisiologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imageamento por Ressonância Magnética , Masculino , Polimorfismo de Nucleotídeo Único
6.
Hum Mutat ; 36(1): 106-17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25385192

RESUMO

Variants in cullin 4B (CUL4B) are a known cause of syndromic X-linked intellectual disability. Here, we describe an additional 25 patients from 11 families with variants in CUL4B. We identified nine different novel variants in these families and confirmed the pathogenicity of all nontruncating variants. Neuroimaging data, available for 15 patients, showed the presence of cerebral malformations in ten patients. The cerebral anomalies comprised malformations of cortical development (MCD), ventriculomegaly, and diminished white matter volume. The phenotypic heterogeneity of the cerebral malformations might result from the involvement of CUL-4B in various cellular pathways essential for normal brain development. Accordingly, we show that CUL-4B interacts with WDR62, a protein in which variants were previously identified in patients with microcephaly and a wide range of MCD. This interaction might contribute to the development of cerebral malformations in patients with variants in CUL4B.


Assuntos
Encéfalo/patologia , Proteínas Culina/genética , Proteínas Culina/metabolismo , Malformações do Desenvolvimento Cortical/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteínas do Tecido Nervoso/metabolismo , Adolescente , Adulto , Proteínas de Ciclo Celular , Células Cultivadas , Criança , Pré-Escolar , Estudos de Associação Genética , Células HEK293 , Humanos , Lactente , Masculino , Malformações do Desenvolvimento Cortical/metabolismo , Malformações do Desenvolvimento Cortical/patologia , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Pessoa de Meia-Idade , Linhagem , Análise de Sequência de DNA , Adulto Jovem
7.
Hum Mol Genet ; 23(23): 6163-76, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24986922

RESUMO

Genome instability, epigenetic remodelling and structural chromosomal rearrangements are hallmarks of cancer. However, the coordinated epigenetic effects of constitutional chromosomal rearrangements that disrupt genes associated with congenital neurodevelopmental diseases are poorly understood. To understand the genetic-epigenetic interplay at breakpoints of chromosomal translocations disrupting CG-rich loci, we quantified epigenetic modifications at DLGAP4 (SAPAP4), a key post-synaptic density 95 (PSD95) associated gene, truncated by the chromosome translocation t(8;20)(p12;q11.23), co-segregating with cerebellar ataxia in a five-generation family. We report significant epigenetic remodelling of the DLGAP4 locus triggered by the t(8;20)(p12;q11.23) translocation and leading to dysregulation of DLGAP4 expression in affected carriers. Disruption of DLGAP4 results in monoallelic hypermethylation of the truncated DLGAP4 promoter CpG island. This induced hypermethylation is maintained in somatic cells of carriers across several generations in a t(8;20) dependent-manner however, is erased in the germ cells of the translocation carriers. Subsequently, chromatin remodelling of the locus-perturbed monoallelic expression of DLGAP4 mRNAs and non-coding RNAs in haploid cells having the translocation. Our results provide new mechanistic insight into the way a balanced chromosomal rearrangement associated with a neurodevelopmental disorder perturbs allele-specific epigenetic mechanisms at breakpoints leading to the deregulation of the truncated locus.


Assuntos
Ataxia Cerebelar/genética , Montagem e Desmontagem da Cromatina , Epigênese Genética , Proteínas do Tecido Nervoso/genética , Cromossomos Humanos Par 8/genética , Ilhas de CpG , Metilação de DNA , Feminino , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Proteínas do Tecido Nervoso/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Associadas SAP90-PSD95 , Translocação Genética
8.
J Clin Invest ; 123(5): 2244-56, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23563313

RESUMO

NF-κB is a master regulator of inflammation and has been implicated in the pathogenesis of immune disorders and cancer. Its regulation involves a variety of steps, including the controlled degradation of inhibitory IκB proteins. In addition, the inactivation of DNA-bound NF-κB is essential for its regulation. This step requires a factor known as copper metabolism Murr1 domain-containing 1 (COMMD1), the prototype member of a conserved gene family. While COMMD proteins have been linked to the ubiquitination pathway, little else is known about other family members. Here we demonstrate that all COMMD proteins bind to CCDC22, a factor recently implicated in X-linked intellectual disability (XLID). We showed that an XLID-associated CCDC22 mutation decreased CCDC22 protein expression and impaired its binding to COMMD proteins. Moreover, some affected individuals displayed ectodermal dysplasia, a congenital condition that can result from developmental NF-κB blockade. Indeed, patient-derived cells demonstrated impaired NF-κB activation due to decreased IκB ubiquitination and degradation. In addition, we found that COMMD8 acted in conjunction with CCDC22 to direct the degradation of IκB proteins. Taken together, our results indicate that CCDC22 participates in NF-κB activation and that its deficiency leads to decreased IκB turnover in humans, highlighting an important regulatory component of this pathway.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , NF-kappa B/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cromossomos Humanos X , Displasia Ectodérmica/metabolismo , Ligação Genética , Células HEK293 , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Inflamação , Microscopia de Fluorescência , Mutação , Inibidor de NF-kappaB alfa , Neoplasias/metabolismo , Estrutura Terciária de Proteína , Ubiquitina/metabolismo
9.
J Neurochem ; 123(4): 589-601, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22891703

RESUMO

Many extracellular factors sensitize nociceptors. Often they act simultaneously and/or sequentially on nociceptive neurons. We investigated if stimulation of the protein kinase C epsilon (PKCε) signaling pathway influences the signaling of a subsequent sensitizing stimulus. Central in activation of PKCs is their transient translocation to cellular membranes. We found in cultured nociceptive neurons that only a first stimulation of the PKCε signaling pathway resulted in PKCε translocation. We identified a novel inhibitory cascade to branch off upstream of PKCε, but downstream of Epac via IP3-induced calcium release. This signaling branch actively inhibited subsequent translocation and even attenuated ongoing translocation. A second 'sensitizing' stimulus was rerouted from the sensitizing to the inhibitory branch of the signaling cascade. Central for the rerouting was cytoplasmic calcium increase and CaMKII activation. Accordingly, in behavioral experiments, activation of calcium stores switched sensitizing substances into desensitizing substances in a CaMKII-dependent manner. This mechanism was also observed by in vivo C-fiber electrophysiology corroborating the peripheral location of the switch. Thus, we conclude that the net effect of signaling in nociceptors is defined by the context of the individual cell's signaling history.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Neurônios/metabolismo , Nociceptores/fisiologia , Limiar da Dor/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Análise de Variância , Animais , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Gânglios Espinais/citologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Inositol 1,4,5-Trifosfato/farmacologia , Isoproterenol/farmacologia , Masculino , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Neurônios/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Rianodina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Canais de Cátion TRPV/metabolismo , Tionucleotídeos/farmacologia , Uridina Trifosfato/farmacologia
10.
Am J Hum Genet ; 91(1): 56-72, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22770980

RESUMO

Potocki-Shaffer syndrome (PSS) is a contiguous gene disorder due to the interstitial deletion of band p11.2 of chromosome 11 and is characterized by multiple exostoses, parietal foramina, intellectual disability (ID), and craniofacial anomalies (CFAs). Despite the identification of individual genes responsible for multiple exostoses and parietal foramina in PSS, the identity of the gene(s) associated with the ID and CFA phenotypes has remained elusive. Through characterization of independent subjects with balanced translocations and supportive comparative deletion mapping of PSS subjects, we have uncovered evidence that the ID and CFA phenotypes are both caused by haploinsufficiency of a single gene, PHF21A, at 11p11.2. PHF21A encodes a plant homeodomain finger protein whose murine and zebrafish orthologs are both expressed in a manner consistent with a function in neurofacial and craniofacial development, and suppression of the latter led to both craniofacial abnormalities and neuronal apoptosis. Along with lysine-specific demethylase 1 (LSD1), PHF21A, also known as BHC80, is a component of the BRAF-histone deacetylase complex that represses target-gene transcription. In lymphoblastoid cell lines from two translocation subjects in whom PHF21A was directly disrupted by the respective breakpoints, we observed derepression of the neuronal gene SCN3A and reduced LSD1 occupancy at the SCN3A promoter, supporting a direct functional consequence of PHF21A haploinsufficiency on transcriptional regulation. Our finding that disruption of PHF21A by translocations in the PSS region is associated with ID adds to the growing list of ID-associated genes that emphasize the critical role of transcriptional regulation and chromatin remodeling in normal brain development and cognitive function.


Assuntos
Transtornos Cromossômicos/genética , Cromossomos Humanos Par 11 , Anormalidades Craniofaciais/genética , Histona Desacetilases/genética , Deficiência Intelectual/genética , Translocação Genética , Adolescente , Adulto , Animais , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Exostose Múltipla Hereditária , Feminino , Genótipo , Haploinsuficiência , Humanos , Recém-Nascido , Masculino , Canal de Sódio Disparado por Voltagem NAV1.3 , Canais de Sódio/genética , Peixe-Zebra
11.
Am J Med Genet A ; 155A(8): 1976-80, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21739581

RESUMO

The genetic basis of autosomal recessive mental retardation (ARMR) is extremely heterogeneous, and there is reason to suspect that the number of underlying gene defects may well go beyond 1,000. To date, however, only less than 10 genes have been implicated in non-specific/non-syndromic ARMR (NS-ARMR). As part of an ongoing systematic study aiming to identify further ARMR genes, we investigated a consanguineous family with three patients with NS-ARMR. By linkage analysis and subsequent mutation screening we identified a novel nonsense mutation (c.163C > T [p.Q55X]) in the second exon of the TUSC3 gene. This is the third MR causing defect in TUSC3 to be described and the second independent mutation in this gene in a cohort of more than 200 ARMR families from the Iranian population. This argues for a more prominent role of TUSC3 in the etiology of this genetically heterogeneous disorder as compared to most of the other so far identified ARMR genes.


Assuntos
Códon sem Sentido , Consanguinidade , Genes Recessivos , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Humanos Par 8/genética , Análise Mutacional de DNA , Feminino , Ligação Genética , Haplótipos , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
12.
Am J Hum Genet ; 87(4): 465-79, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20887964

RESUMO

By defining the chromosomal breakpoint of a balanced t(10;12) translocation from a subject with Kallmann syndrome and scanning genes in its vicinity in unrelated hypogonadal subjects, we have identified WDR11 as a gene involved in human puberty. We found six patients with a total of five different heterozygous WDR11 missense mutations, including three alterations (A435T, R448Q, and H690Q) in WD domains important for ß propeller formation and protein-protein interaction. In addition, we discovered that WDR11 interacts with EMX1, a homeodomain transcription factor involved in the development of olfactory neurons, and that missense alterations reduce or abolish this interaction. Our findings suggest that impaired pubertal development in these patients results from a deficiency of productive WDR11 protein interaction.


Assuntos
Cromossomos Humanos Par 10/genética , Proteínas de Homeodomínio/genética , Hipogonadismo/genética , Síndrome de Kallmann/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas/genética , Puberdade/genética , Fatores de Transcrição/genética , Translocação Genética/genética , Adolescente , Animais , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Hibridização in Situ Fluorescente , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Análise em Microsséries , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido , Peixe-Zebra
13.
Am J Hum Genet ; 86(6): 949-56, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20493458

RESUMO

Cranioectodermal dysplasia (CED) is a disorder characterized by craniofacial, skeletal, and ectodermal abnormalities. Most cases reported to date are sporadic, but a few familial cases support an autosomal-recessive inheritance pattern. Aiming at the elucidation of the genetic basis of CED, we collected 13 patients with CED symptoms from 12 independent families. In one family with consanguineous parents two siblings were affected, permitting linkage analysis and homozygosity mapping. This revealed a single region of homozygosity with a significant LOD score (3.57) on chromosome 3q21-3q24. By sequencing candidate genes from this interval we found a homozygous missense mutation in the IFT122 (WDR10) gene that cosegregated with the disease. Examination of IFT122 in our patient cohort revealed one additional homozygous missense change in the patient from a second consanguineous family. In addition, we found compound heterozygosity for a donor splice-site change and a missense change in one sporadic patient. All mutations were absent in 340 control chromosomes. Because IFT122 plays an important role in the assembly and maintenance of eukaryotic cilia, we investigated patient fibroblasts and found significantly reduced frequency and length of primary cilia as compared to controls. Furthermore, we transiently knocked down ift122 in zebrafish embryos and observed the typical phenotype found in other models of ciliopathies. Because not all of our patients harbored mutations in IFT122, CED seems to be genetically heterogeneous. Still, by identifying CED as a ciliary disorder, our study suggests that the causative mutations in the unresolved cases most likely affect primary cilia function too.


Assuntos
Anormalidades Craniofaciais/genética , Displasia Ectodérmica/genética , Proteínas/genética , Proteínas Adaptadoras de Transdução de Sinal , Criança , Pré-Escolar , Transtornos da Motilidade Ciliar/genética , Proteínas do Citoesqueleto , Feminino , Humanos , Lactente , Masculino , Mutação
14.
PLoS One ; 5(2): e9242, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20169082

RESUMO

Mutations in the human gene MCPH1 cause primary microcephaly associated with a unique cellular phenotype with premature chromosome condensation (PCC) in early G2 phase and delayed decondensation post-mitosis (PCC syndrome). The gene encodes the BRCT-domain containing protein microcephalin/BRIT1. Apart from its role in the regulation of chromosome condensation, the protein is involved in the cellular response to DNA damage. We report here on the first mouse model of impaired Mcph1-function. The model was established based on an embryonic stem cell line from BayGenomics (RR0608) containing a gene trap in intron 12 of the Mcph1 gene deleting the C-terminal BRCT-domain of the protein. Although residual wild type allele can be detected by quantitative real-time PCR cell cultures generated from mouse tissues bearing the homozygous gene trap mutation display the cellular phenotype of misregulated chromosome condensation that is characteristic for the human disorder, confirming defective Mcph1 function due to the gene trap mutation. While surprisingly the DNA damage response (formation of repair foci, chromosomal breakage, and G2/M checkpoint function after irradiation) appears to be largely normal in cell cultures derived from Mcph1(gt/gt) mice, the overall survival rates of the Mcph1(gt/gt) animals are significantly reduced compared to wild type and heterozygous mice. However, we could not detect clear signs of premature malignant disease development due to the perturbed Mcph1 function. Moreover, the animals show no obvious physical phenotype and no reduced fertility. Body and brain size are within the range of wild type controls. Gene expression on RNA and protein level did not reveal any specific pattern of differentially regulated genes. To the best of our knowledge this represents the first mammalian transgenic model displaying a defect in mitotic chromosome condensation and is also the first mouse model for impaired Mcph1-function.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Quebra Cromossômica , Cromossomos de Mamíferos/genética , Dano ao DNA , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Proteínas do Citoesqueleto , Eletroforese em Gel Bidimensional , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Modelos Animais , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Análise de Sobrevida
15.
Am J Hum Genet ; 82(5): 1158-64, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18452889

RESUMO

Recent studies have shown that autosomal recessive mental retardation (ARMR) is extremely heterogeneous, and there is reason to believe that the number of underlying gene defects goes into the thousands. To date, however, only four genes have been implicated in nonsyndromic ARMR (NS-ARMR): PRSS12 (neurotrypsin), CRBN (cereblon), CC2D1A, and GRIK2. As part of an ongoing systematic study aiming to identify ARMR genes, we investigated a large consanguineous family comprising seven patients with nonsyndromic ARMR in four sibships. Genome-wide SNP typing enabled us to map the relevant genetic defect to a 4.6 Mbp interval on chromosome 8. Haplotype analyses and copy-number studies led to the identification of a homozygous deletion partly removing TUSC3 (N33) in all patients. All obligate carriers of this family were heterozygous, but none of 192 unrelated healthy individuals from the same population carried this deletion. We excluded other disease-causing mutations in the coding regions of all genes within the linkage interval by sequencing; moreover, we verified the complete absence of a functional TUSC3 transcript in all patients through RT-PCR. TUSC3 is thought to encode a subunit of the endoplasmic reticulum-bound oligosaccharyltransferase complex that catalyzes a pivotal step in the protein N-glycosylation process. Our data suggest that in contrast to other genetic defects of glycosylation, inactivation of TUSC3 causes nonsyndromic MR, a conclusion that is supported by a separate report in this issue of AJHG. TUSC3 is only the fifth gene implicated in NS-ARMR and the first for which mutations have been reported in more than one family.


Assuntos
Ligação Genética , Genoma Humano , Hexosiltransferases/genética , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Criança , Feminino , Glicosilação , Homozigoto , Humanos , Masculino , Linhagem , Polimorfismo de Nucleotídeo Único
16.
Am J Hum Genet ; 82(2): 432-43, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18252223

RESUMO

Submicroscopic copy-number imbalances contribute significantly to the genetic etiology of human disease. Here, we report a novel microduplication hot spot at Xp11.22 identified in six unrelated families with predominantly nonsyndromic XLMR. All duplications segregate with the disease, including the large families MRX17 and MRX31. The minimal, commonly duplicated region contains three genes: RIBC1, HSD17B10, and HUWE1. RIBC1 could be excluded on the basis of its absence of expression in the brain and because it escapes X inactivation in females. For the other genes, expression array and quantitative PCR analysis in patient cell lines compared to controls showed a significant upregulation of HSD17B10 and HUWE1 as well as several important genes in their molecular pathways. Loss-of-function mutations of HSD17B10 have previously been associated with progressive neurological disease and XLMR. The E3 ubiquitin ligase HUWE1 has been implicated in TP53-associated regulation of the neuronal cell cycle. Here, we also report segregating sequence changes of highly conserved residues in HUWE1 in three XLMR families; these changes are possibly associated with the phenotype. Our findings demonstrate that an increased gene dosage of HSD17B10, HUWE1, or both contribute to the etiology of XLMR and suggest that point mutations in HUWE1 are associated with this disease too.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/genética , Cromossomos Humanos X/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Ubiquitina-Proteína Ligases/genética , Sequência de Bases , Western Blotting , Análise Mutacional de DNA , DNA Complementar/genética , Dosagem de Genes/genética , Duplicação Gênica , Humanos , Hibridização in Situ Fluorescente , Análise em Microsséries , Dados de Sequência Molecular , Mutação/genética , Linhagem , Proteínas Supressoras de Tumor
17.
Hum Genet ; 121(5): 539-47, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17333282

RESUMO

Using high resolution X chromosome array-CGH we identified an interstitial microdeletion at Xp11.23 in three brothers with moderate to severe mental retardation (MR) without dysmorphic features. The extent of the deletion was subsequently delineated to about 50 kb by regular PCR and included only the SLC38A5 and FTSJ1 genes. The loss of the FTSJ1 MR gene in males is expected to result in the observed phenotype but the contribution of the deletion of the solute carrier SLC38A5 gene is less clear. Their mother also carries the deletion and completely inactivates the aberrant X chromosome. Interestingly, the distal breakpoint is situated within a 200 kb SSX repeat region that appears to stimulate recombination since subtle copy number changes often occur at this location and it is frequently involved in translocations in tumours. Since this apparent SSX unstable structure is flanked proximally by FTSJ1 and PQBP1, subtle deletions or duplications at this location would be expected to cause MR, as in our family. So far, we have screened a cohort of 300 patients but did not find additional aberrations at the FTSJ1 locus indicating that the frequency is likely to be low.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Deleção Cromossômica , Cromossomos Humanos X , Deficiência Intelectual/genética , Metiltransferases/genética , Proteínas Nucleares/genética , Feminino , Humanos , Masculino , Reação em Cadeia da Polimerase , Aberrações dos Cromossomos Sexuais , Irmãos , Inativação do Cromossomo X
18.
Am J Med Genet A ; 140(5): 496-502, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16470790

RESUMO

We report a 2(3/12)-year-old boy with a constitutional interstitial deletion of 5q,46,XY,del(5)(q23.3q31.2) de novo. Clinical manifestations in this patient included failure to thrive, psychomotor retardation, mild facial dysmorphic features, and long and slender fingers and toes. The precise location and extent (9.5 Mb) of the deletion was determined by fluorescence in situ hybridization (FISH) using 19 YAC and BAC clones. Comparison of the present patient with six other patients with deletions of chromosomal bands 5q22-5q31 allowed further delineation of a constitutional del5q22q31 syndrome. The main features of this syndrome are psychomotor retardation, failure to thrive, hypotonia, hypoplastic muscles, cleft or high arched palate, low-set and dysplastic ears, flat nasal bridge, downslanting palpebral fissures, hypertelorism, anteverted nostrils, and micro- and/or retrognathia.


Assuntos
Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Anormalidades Múltiplas/patologia , Pré-Escolar , Bandeamento Cromossômico , Deficiências do Desenvolvimento/patologia , Orelha/anormalidades , Face/anormalidades , Insuficiência de Crescimento/patologia , Transtornos de Alimentação na Infância/patologia , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Fenótipo
19.
Dev Biol ; 272(1): 53-65, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15242790

RESUMO

Choroideremia (CHM) is a hereditary eye disease caused by mutations in the X-linked CHM gene. Disruption of the Chm gene in mice resulted in prenatal death of Chm-/Y males and Chm-/Chm+ females that had inherited the mutation from their mothers. Male chimeras and Chm+/Chm- females with paternal transmission of the mutation were viable and had photoreceptor degeneration reminiscent of human choroideremia. Here, we show that Chm-/Y males and Chm-/Chm+ females were retarded at e7.5 and died before e11.5 due to multiple defects of the extra-embryonic tissues. Mutant embryos exhibited deficiency of diploid trophoblasts associated with overabundance of giant cells. In yolk sac and placenta, severe defects in vasculogenesis were obvious. Chm-/Y males exhibited more pronounced phenotypes than Chm-/Chm+ females. The lethal genotypes could be rescued by tetraploid aggregation. Chm-/Chm+ females, but not Chm-/Y males, could also be rescued when their Chm+/Chm- mothers were mated with Mus spretus males. Backcross analysis suggested that the viability of interspecies hybrid Chm-/Chm+ females may be due to expression from the Chm allele on the M. spretus X-chromosome rather than a modifier effect. Our results demonstrate that Chm is essential for diploid trophoblast development and plays a role in the vascularization in placenta and yolk sac.


Assuntos
Alquil e Aril Transferases/genética , Vasos Sanguíneos/embriologia , Embrião de Mamíferos/irrigação sanguínea , Regulação da Expressão Gênica no Desenvolvimento , Trofoblastos/fisiologia , Alquil e Aril Transferases/metabolismo , Animais , Vasos Sanguíneos/fisiologia , Cruzamento , Olho/patologia , Feminino , Morte Fetal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Placenta/patologia , Poliploidia , Gravidez , Cromossomo X , Cromossomo Y , Saco Vitelino/irrigação sanguínea , Saco Vitelino/patologia
20.
Am J Hum Genet ; 75(2): 305-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15162322

RESUMO

Nonsyndromic X-linked mental retardation (NSXLMR) is a very heterogeneous condition, and most of the underlying gene defects are still unknown. Recently, we have shown that approximately 30% of these genes cluster on the proximal Xp, which prompted us to perform systematic mutation screening in brain-expressed genes from this region. Here, we report on a novel NSXLMR gene, FTSJ1, which harbors mutations in three unrelated families--one with a splicing defect, one with a nonsense mutation, and one with a deletion of one nucleotide. In two families, subsequent expression studies showed complete absence or significant reduction of mutant FTSJ1 transcripts. FTSJ1 protein is a homolog of Escherichia coli RNA methyltransferase FtsJ/RrmJ and may play a role in the regulation of translation. Further studies aim to elucidate the function of human FTSJ1 and its role during brain development.


Assuntos
Cromossomos Humanos X , Deficiência Intelectual Ligada ao Cromossomo X/genética , Metiltransferases/genética , Proteínas Nucleares/genética , Adulto , Northern Blotting , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Linhagem , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA