Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Eur Heart J ; 37(23): 1826-34, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-26537620

RESUMO

AIMS: Increased left ventricular wall thickness (LVWT) is a common finding in cardiology. It is not known how often hereditary transthyretin-related familial amyloid cardiomyopathy (mTTR-FAC) is responsible for LVWT. Several therapeutic modalities for mTTR-FAC are currently in clinical trials; thus, it is important to establish the prevalence of TTR mutations (mTTR) and the clinical characteristics of the patients with mTTR-FAC. METHODS AND RESULTS: In a prospective multicentre, cross-sectional study, the TTR gene was sequenced in 298 consecutive patients diagnosed with increased LVWT in primary cardiology clinics in France. Among the included patients, median (25-75th percentiles) age was 62 [50;74]; 74% were men; 23% were of African origin; and 36% were in NYHA Class III-IV. Median LVWT was 18 (16-21) mm. Seventeen (5.7%; 95% confidence interval [CI]: [3.4;9.0]) patients had mTTR of whom 15 (5.0%; 95% CI [2.9;8.2]) had mTTR-FAC. The most frequent mutations were V142I (n = 8), V50M (n = 2), and I127V (n = 2). All mTTR-FAC patients were older than 63 years with a median age of 74 [69;79]. Of the 15 patients with mTTR-FAC, 8 were of African descent while 7 were of European descent. In the African descendants, mTTR-FAC median age was 74 [72;79] vs. 55 [46;65] years in non-mTTR-FAC (P < 0.001). In an adjusted multivariate model, African origin, neuropathy, carpal tunnel syndrome, electrocardiogram (ECG) low voltage, and late gadolinium enhancement (LGE) at cardiac-magnetic resonance imaging were all independently associated with mTTR-FAC. CONCLUSION: Five per cent of patients diagnosed with hypertrophic cardiomyopathy have mTTR-FAC. Mutated transthyretin genetic screening is warranted in elderly subjects with increased LVWT, particularly, those of African descent with neuropathy, carpal tunnel syndrome, ECG low voltage, or LGE.


Assuntos
Neuropatias Amiloides Familiares/patologia , Cardiomiopatia Hipertrófica/patologia , Idoso , Idoso de 80 Anos ou mais , Amiloide/genética , Neuropatias Amiloides Familiares/epidemiologia , Neuropatias Amiloides Familiares/genética , Cardiomiopatia Hipertrófica/epidemiologia , Cardiomiopatia Hipertrófica/genética , Estudos Transversais , Feminino , França/epidemiologia , Insuficiência Cardíaca/epidemiologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Ventrículos do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Pré-Albumina/genética , Prevalência , Estudos Prospectivos
2.
Hum Mutat ; 35(7): 805-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24633926

RESUMO

Cystic fibrosis may be revealed by nasal polyposis (NP) starting early in life. We performed cystic fibrosis transmembrane conductance regulator (CFTR) DNA and mRNA analyses in the family of a 12-year-old boy presenting with NP and a normal sweat test. Routine DNA analysis only showed the heterozygous c.2551C>T (p.Arg851*) mutation in the child and the father. mRNA analysis showed partial exon skipping due to c.2551C>T and a significant increase in total CFTR mRNA in the patient and the mother, which was attributable to the heterozygous c. -2954G>A variant in the distant promoter region, as demonstrated by in vitro luciferase assays. The 5' rapid amplification of cDNA ends analysis showed the presence of a novel transcript, where the canonical exon 1 was replaced by an alternative exon called 1a-Long. This case report could represent the first description of a CFTR-related disorder associated with the presence of a 5' alternative, probably nonfunctional transcript, similar to those of fetal origin.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Mutação , Pólipos Nasais/genética , Isoformas de RNA , RNA Mensageiro/genética , Regiões 5' não Traduzidas , Processamento Alternativo , Criança , Análise Mutacional de DNA , Ordem dos Genes , Humanos , Masculino , Pólipos Nasais/diagnóstico , Linhagem
3.
Eur J Hum Genet ; 22(4): 471-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24129437

RESUMO

Obesity is a common but highly, clinically, and genetically heterogeneous disease. Deletion of the terminal region of the short arm of chromosome 2 is rare and has been reported in about 13 patients in the literature often associated with a Prader-Willi-like phenotype. We report on five unrelated patients with 2p25 deletion of paternal origin presenting with early-onset obesity, hyperphagia, intellectual deficiency, and behavioural difficulties. Among these patients, three had de novo pure 2pter deletions, one presented with a paternal derivative der(2)t(2;15)(p25.3;q26) with deletion in the 2pter region and the last patient presented with an interstitial 2p25 deletion. The size of the deletions was characterized by SNP array or array-CGH and was confirmed by fluorescence in situ hybridization (FISH) studies. Four patients shared a 2p25.3 deletion with a minimal critical region estimated at 1.97 Mb and encompassing seven genes, namely SH3HYL1, ACP1, TMEMI8, SNTG2, TPO, PXDN, and MYT1L genes. The fifth patient had a smaller interstitial deletion encompassing the TPO, PXDN, and MYT1L genes. Paternal origin of the deletion was determined by genotyping using microsatellite markers. Analysis of the genes encompassed in the deleted region led us to speculate that the ACP1, TMEM18, and/or MYT1L genes might be involved in early-onset obesity. In addition, intellectual deficiency and behavioural troubles can be explained by the heterozygous loss of the SNTG2 and MYT1L genes. Finally, we discuss the parent-of-origin of the deletion.


Assuntos
Cromossomos Humanos Par 2/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Obesidade/genética , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas/genética , Deleção de Sequência , Fatores de Transcrição/genética , Criança , Pré-Escolar , Hibridização Genômica Comparativa , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Hibridização in Situ Fluorescente , Masculino , Obesidade/complicações , Obesidade/diagnóstico , Fenótipo , Polimorfismo de Nucleotídeo Único , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/genética , Análise de Sequência de DNA
4.
Am J Hum Genet ; 92(5): 707-24, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23643381

RESUMO

Transcription factor SOX10 plays a role in the maintenance of progenitor cell multipotency, lineage specification, and cell differentiation and is a major actor in the development of the neural crest. It has been implicated in Waardenburg syndrome (WS), a rare disorder characterized by the association between pigmentation abnormalities and deafness, but SOX10 mutations cause a variable phenotype that spreads over the initial limits of the syndrome definition. On the basis of recent findings of olfactory-bulb agenesis in WS individuals, we suspected SOX10 was also involved in Kallmann syndrome (KS). KS is defined by the association between anosmia and hypogonadotropic hypogonadism due to incomplete migration of neuroendocrine gonadotropin-releasing hormone (GnRH) cells along the olfactory, vomeronasal, and terminal nerves. Mutations in any of the nine genes identified to date account for only 30% of the KS cases. KS can be either isolated or associated with a variety of other symptoms, including deafness. This study reports SOX10 loss-of-function mutations in approximately one-third of KS individuals with deafness, indicating a substantial involvement in this clinical condition. Study of SOX10-null mutant mice revealed a developmental role of SOX10 in a subpopulation of glial cells called olfactory ensheathing cells. These mice indeed showed an almost complete absence of these cells along the olfactory nerve pathway, as well as defasciculation and misrouting of the nerve fibers, impaired migration of GnRH cells, and disorganization of the olfactory nerve layer of the olfactory bulbs.


Assuntos
Surdez/genética , Predisposição Genética para Doença/genética , Síndrome de Kallmann/genética , Neuroglia/patologia , Condutos Olfatórios/patologia , Fatores de Transcrição SOXE/genética , Animais , Análise Mutacional de DNA , Surdez/patologia , Feminino , França , Galactosídeos , Células HeLa , Humanos , Indóis , Síndrome de Kallmann/patologia , Masculino , Camundongos , Mutação/genética , Plasmídeos/genética
5.
Hum Mol Genet ; 22(12): 2387-99, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23427148

RESUMO

Goldberg-Shprintzen syndrome (GOSHS, MIM #609460) is an autosomal recessive disorder of intellectual disability, specific facial gestalt and Hirschsprung's disease (HSCR). In 2005, homozygosity mapping in a large consanguineous family identified KIAA1279 as the disease-causing gene. KIAA1279 encodes KIF-binding protein (KBP), whose function is incompletely understood. Studies have identified either the mitochondria or the cytoskeleton as the site of KBP localization and interactions. To better delineate the KIAA1279-related clinical spectrum and the molecular mechanisms involved in GOSHS, we studied five new patients from three different families. The homozygous KIAA1279 mutations in these patients (p.Arg90X, p.Ser200X or p.Arg202IlefsX2) led to nonsense-mediated mRNA decay and loss of KBP function. Despite the absence of functional KBP, respiratory chain complex activity in patient fibroblasts was normal. KBP did not co-localize with mitochondria in control human fibroblasts, but interacted with the actin and tubulin cytoskeleton. KBP expression directly affected neurite growth in a neuron-like cell line (human neuroblastoma SH-SY5Y), in keeping with the central (polymicrogyria) and enteric (HSCR) neuronal developmental defects seen in GOSHS patients. The KBP interactions with actin filaments and microtubules (MTs) demonstrated in our study constitute the first evidence that an actin MT cross-link protein is involved in neuronal development in humans.


Assuntos
Anormalidades Craniofaciais/metabolismo , Doença de Hirschsprung/metabolismo , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Actinas/genética , Actinas/metabolismo , Adolescente , Adulto , Criança , Anormalidades Craniofaciais/genética , Feminino , França , Doença de Hirschsprung/genética , Humanos , Lactente , Iraque , Masculino , Microtúbulos/genética , Mutação , Proteínas do Tecido Nervoso/genética , Linhagem , Ligação Proteica , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , População Branca/genética
6.
Hum Mutat ; 34(6): 873-81, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23420618

RESUMO

With the increased number of identified nucleotide sequence variations in genes, the current challenge is to classify them as disease causing or neutral. These variants of unknown clinical significance can alter multiple processes, from gene transcription to RNA splicing or protein function. Using an approach combining several in silico tools, we identified some exons presenting weaker splicing motifs than other exons in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) gene. These exons exhibit higher rates of basal skipping than exons harboring no identifiable weak splicing signals using minigene assays. We then screened 19 described mutations in three different exons, and identified exon-skipping substitutions. These substitutions induced higher skipping levels in exons having one or more weak splicing motifs. Indeed, this level remained under 2% for exons with strong splicing motifs and could reach 40% for exons having at least one weak motif. Further analysis revealed a functional exon splicing enhancer within exon 3 that was associated with the SR protein SF2/ASF and whose disruption induced exon skipping. Exon skipping was confirmed in vivo in two nasal epithelial cell brushing samples. Our approach, which point out exons with some splicing signals weaknesses, will help spot splicing mutations of clinical relevance.


Assuntos
Processamento Alternativo , Biologia Computacional , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Éxons , Biologia Computacional/métodos , Simulação por Computador , Humanos , Modelos Biológicos , Mutação de Sentido Incorreto , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico , Fatores de Processamento de Serina-Arginina , Transcrição Gênica
7.
Hum Mutat ; 34(2): 287-91, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23065710

RESUMO

The correction of premature termination codons (PTCs) by agents that promote readthrough represents a promising emerging tool for the treatment of many genetic diseases. The efficiency of the treatment, however, varies depending on the stop codon itself and the amount of correctible transcripts related to the efficiency of nonsense-mediated decay. In the current study, a screen by in vitro minigene assay of all six PTCs described in exon 15 of the CFTR gene demonstrated alternative splicing to differing degrees for five of them. Of the five, PTC mutations c.2537G>A (p.Trp846*(UAG) ) and c.2551C>T (p.Arg851*) cause the greatest proportion of transcripts lacking exon 15; both mutations altering exonic splicing regulatory elements. In order to increase the amount of full-length transcripts, different pharmacological treatments were performed showing both negative and positive effects on exon inclusion for the same mutation. Therefore, the total amount of transcripts together with the splicing profile should be assessed to anticipate and improve efficacy of readthrough therapy.


Assuntos
Processamento Alternativo/genética , Códon sem Sentido/genética , Éxons , Fases de Leitura/genética , Brônquios/citologia , Brônquios/metabolismo , Linhagem Celular , Fibrose Cística/genética , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
8.
Hum Reprod ; 27(5): 1460-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22416012

RESUMO

BACKGROUND: Kallmann syndrome (KS) is a genetic disorder associating pubertal failure with congenitally absent or impaired sense of smell. KS is related to defective neuronal development affecting both the migration of olfactory nerve endings and GnRH neurons. The discovery of several genetic mutations responsible for KS led to the identification of signaling pathways involved in these processes, but the mutations so far identified account for only 30% of cases of KS. Here, we attempted to identify new genes responsible for KS by using a pan-genomic approach. METHODS: From a cohort of 120 KS patients, we selected 48 propositi with no mutations in known KS genes. They were analyzed by comparative genomic hybridization array, using Agilent 105K oligonucleotide chips with a mean resolution of 50 kb. RESULTS: One propositus was found to have a heterozygous deletion of 213 kb at locus 7q21.11, confirmed by real-time qPCR, deleting 11 of the 17 SEMA3A exons. This deletion cosegregated in the propositus' family with the KS phenotype, that was transmitted in autosomal dominant fashion and was not associated with other neurological or non-neurological clinical disorders. SEMA3A codes for semaphorin 3A, a protein that interacts with neuropilins. Mice lacking semaphorin 3A expression have been showed to have a Kallmann-like phenotype. CONCLUSIONS: SEMA3A is therefore a new gene whose loss-of-function is involved in KS. These findings validate the specific role of semaphorin 3A in the development of the olfactory system and in neuronal control of puberty in humans.


Assuntos
Deleção de Genes , Síndrome de Kallmann/genética , Semaforina-3A/genética , Feminino , Humanos , Masculino , Linhagem , Fenótipo , Puberdade/genética , Puberdade/fisiologia , Semaforina-3A/fisiologia , Olfato/genética , Olfato/fisiologia
9.
J Cyst Fibros ; 10(6): 479-82, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21783433

RESUMO

BACKGROUND: The identification by CFTR mRNA studies of a new deep-intronic splicing mutation, c.870-1113_1110delGAAT, in one patient of our series with mild CF symptoms and in three CF patients of an Italian study, led us to evaluate the mutation frequency and phenotype/genotype correlations. METHODS: 266 patients with CF and related disorders and having at least one undetected mutation, were tested at the gDNA level in three French reference laboratories. RESULTS: In total, the mutation was found in 13 unrelated patients (5% of those already carrying a mutation) plus 4 siblings, including one homozygote and 12 heterozygotes having a severe CF mutation. The sweat test was positive in 10/14 documented cases, the diagnosis was delayed after 20 years in 9/15 and pancreatic insufficiency was present in 5/16. CONCLUSION: c.870-1113_1110delGAAT should be considered as CF-causing with phenotype variability and overall delayed diagnosis. Its frequency highlights the potential of mRNA studies.


Assuntos
Fibrose Cística/genética , Íntrons/genética , Mutação , RNA Mensageiro/genética , Adolescente , Adulto , Criança , Pré-Escolar , Fibrose Cística/diagnóstico , Feminino , Humanos , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Adulto Jovem
10.
PLoS One ; 6(3): e18334, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21483833

RESUMO

The CFTR (cystic fibrosis transmembrane conductance regulator) protein is a large polytopic protein whose biogenesis is inefficient. To better understand the regulation of CFTR processing and trafficking, we conducted a genetic screen that identified COMMD1 as a new CFTR partner. COMMD1 is a protein associated with multiple cellular pathways, including the regulation of hepatic copper excretion, sodium uptake through interaction with ENaC (epithelial sodium channel) and NF-kappaB signaling. In this study, we show that COMMD1 interacts with CFTR in cells expressing both proteins endogenously. This interaction promotes CFTR cell surface expression as assessed by biotinylation experiments in heterologously expressing cells through regulation of CFTR ubiquitination. In summary, our data demonstrate that CFTR is protected from ubiquitination by COMMD1, which sustains CFTR expression at the plasma membrane. Thus, increasing COMMD1 expression may provide an approach to simultaneously inhibit ENaC absorption and enhance CFTR trafficking, two major issues in cystic fibrosis.


Assuntos
Proteínas de Transporte/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte Proteico/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Biotinilação , Proteínas de Transporte/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células HT29 , Células HeLa , Humanos , Imunoprecipitação , Ligação Proteica/genética , Ligação Proteica/fisiologia , Transporte Proteico/genética , RNA Interferente Pequeno , Ubiquitinação/genética , Ubiquitinação/fisiologia
11.
Hum Genet ; 129(4): 387-96, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21184098

RESUMO

Fetal bowel anomalies may reveal cystic fibrosis (CF) and the search for CF transmembrane conductance regulator (CFTR) gene mutations is part of the diagnostic investigations in such pregnancies, according to European recommendations. We report on our 18-year experience to document comprehensive CFTR genotypes and correlations with ultrasound patterns in a series of 694 cases of fetal bowel anomalies. CFTR gene analysis was performed in a multistep process, including search for frequent mutations in the parents and subsequent in-depth search for rare mutations, depending on the context. Ultrasound patterns were correlated with the genotypes. Cases were distinguished according to whether they had been referred directly to our laboratory or after an initial testing in another laboratory. A total of 30 CF fetuses and 8 cases compatible with CFTR-related disorders were identified. CFTR rearrangements were found in 5/30 CF fetuses. 21.2% of fetuses carrying a frequent mutation had a second rare mutation, indicative of CF. The frequency of CF among fetuses with no frequent mutation was 0.43%. Correlation with ultrasound patterns revealed a significant frequency of multiple bowel anomalies in CF fetuses. The results emphasize the need to search for rearrangements in the diagnosis strategy of fetal bowel anomalies. The diagnostic value of ultrasound patterns combining hyperechogenic bowel, loop dilatation and/or non-visualized gallbladder reveals a need to revise current strategies and to offer extensive CFTR gene testing when the triad is diagnosed, even when no frequent mutation is found in the first-step analysis.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Intestino Ecogênico/genética , Mutação , Fibrose Cística/diagnóstico , Fibrose Cística/diagnóstico por imagem , Análise Mutacional de DNA , Intestino Ecogênico/diagnóstico , Intestino Ecogênico/diagnóstico por imagem , Feminino , Frequência do Gene , Genótipo , Humanos , Recém-Nascido , Masculino , Fenótipo , Gravidez , Medição de Risco , Fatores de Risco , Ultrassonografia Pré-Natal
12.
PLoS Genet ; 6(10)2010 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-20949073

RESUMO

Approximately 30% of alleles causing genetic disorders generate premature termination codons (PTCs), which are usually associated with severe phenotypes. However, bypassing the deleterious stop codon can lead to a mild disease outcome. Splicing at NAGNAG tandem splice sites has been reported to result in insertion or deletion (indel) of three nucleotides. We identified such a mechanism as the origin of the mild to asymptomatic phenotype observed in cystic fibrosis patients homozygous for the E831X mutation (2623G>T) in the CFTR gene. Analyses performed on nasal epithelial cell mRNA detected three distinct isoforms, a considerably more complex situation than expected for a single nucleotide substitution. Structure-function studies and in silico analyses provided the first experimental evidence of an indel of a stop codon by alternative splicing at a NAGNAG acceptor site. In addition to contributing to proteome plasticity, alternative splicing at a NAGNAG tandem site can thus remove a disease-causing UAG stop codon. This molecular study reveals a naturally occurring mechanism where the effect of either modifier genes or epigenetic factors could be suspected. This finding is of importance for genetic counseling as well as for deciding appropriate therapeutic strategies.


Assuntos
Processamento Alternativo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Mutação de Sentido Incorreto , Sítios de Splice de RNA/genética , Sequência de Bases , Fibrose Cística/genética , Fibrose Cística/patologia , Análise Mutacional de DNA , Feminino , Genótipo , Células HEK293 , Células HeLa , Humanos , Mutação INDEL , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Linhagem , Fenótipo , Transfecção
13.
Eur J Hum Genet ; 18(10): 1166-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20512161

RESUMO

Grade III fetal bowel hyperechogenicity and/or loop dilatation observed at the second trimester of pregnancy can be due to several disease conditions, including cystic fibrosis (CF). Screening for frequent CF mutations is performed as a first step and, in certain situations, such as when a frequent CF mutation is found in the fetus, the increased risk of CF justifies an in-depth study of the second allele. To determine the contribution of large CFTR gene rearrangements in such cases, detected using a semiquantitative fluorescent multiplex PCR (QFM-PCR) assay, we collated data on 669 referrals related to suspicion of CF in fetuses from 1998 to 2009. Deletions were found in 5/70 cases in which QFM-PCR was applied, dele19, dele22_23, dele2_6b, dele14b_15 and dele6a_6b, of which the last three remain undescribed. In 3/5 cases, hyperechogenicity was associated with dilatation and/or gallbladder anomalies. Of the total cases of CF recognized in the subgroup of first-hand referrals, deletions represent 16.7% of CF alleles. Our study thus strengthens the need to consider large CFTR gene rearrangements in the diagnosis strategy of fetal bowel anomalies, in particular in the presence of multiple anomalies.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/diagnóstico , Fibrose Cística/genética , Intestino Ecogênico/genética , Rearranjo Gênico , Testes Genéticos , Diagnóstico Pré-Natal , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/genética , Aberrações Cromossômicas , Fibrose Cística/embriologia , Análise Mutacional de DNA , Feminino , Doenças Fetais/diagnóstico , Doenças Fetais/genética , Vesícula Biliar/anormalidades , Humanos , Recém-Nascido , Intestinos/embriologia , Mutação Puntual , Reação em Cadeia da Polimerase , Gravidez , Segundo Trimestre da Gravidez , Deleção de Sequência
14.
Eur J Hum Genet ; 17(10): 1325-35, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19367324

RESUMO

Oculofaciocardiodental (OFCD) and Lenz microphthalmia syndromes form part of a spectrum of X-linked microphthalmia disorders characterized by ocular, dental, cardiac and skeletal anomalies and mental retardation. The two syndromes are allelic, caused by mutations in the BCL-6 corepressor gene (BCOR). To extend the series of phenotypes associated with pathogenic mutations in BCOR, we sequenced the BCOR gene in patients with (1) OFCD syndrome, (2) putative X-linked ('Lenz') microphthalmia syndrome, (3) isolated ocular defects and (4) laterality phenotypes. We present a new cohort of females with OFCD syndrome and null mutations in BCOR, supporting the hypothesis that BCOR is the sole molecular cause of this syndrome. We identify for the first time mosaic BCOR mutations in two females with OFCD syndrome and one apparently asymptomatic female. We present a female diagnosed with isolated ocular defects and identify minor features of OFCD syndrome, suggesting that OFCD syndrome may be mild and underdiagnosed. We have sequenced a cohort of males diagnosed with putative X-linked microphthalmia and found a mutation, p.P85L, in a single case, suggesting that BCOR mutations are not a major cause of X-linked microphthalmia in males. The absence of BCOR mutations in a panel of patients with non-specific laterality defects suggests that mutations in BCOR are not a major cause of isolated heart and laterality defects. Phenotypic analysis of OFCD and Lenz microphthalmia syndromes shows that in addition to the standard diagnostic criteria of congenital cataract, microphthalmia and radiculomegaly, patients should be examined for skeletal defects, particularly radioulnar synostosis, and cardiac/laterality defects.


Assuntos
Anormalidades do Olho/genética , Cardiopatias/genética , Deficiência Intelectual/genética , Microftalmia/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Adolescente , Adulto , Idoso , Alelos , Animais , Criança , Pré-Escolar , Estudos de Coortes , Anormalidades do Olho/complicações , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/genética , Cardiopatias/complicações , Humanos , Recém-Nascido , Deficiência Intelectual/complicações , Masculino , Microftalmia/complicações , Pessoa de Meia-Idade , Síndrome
15.
Eur J Med Genet ; 51(6): 598-607, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18822396

RESUMO

Interstitial 2q36 deletion is a rare event. We report on a patient with a de novo del(2)(q36.2q36.3) interstitial deletion of the long arm of chromosome 2 diagnosed by classical banding. The phenotype comprised facial dysmorphism, enlarged kidneys with multiple renal cysts, abnormal minora labia, asymmetric lower limbs with dysplastic patella, and severe mental retardation. By physical mapping, using array-comparative genomic hybridisation (CGH) confirmed by Fluorescent In Situ Hybridisation (FISH), the breakpoints of the deletion were mapped and the size of the deletions was measured: 5.61+/-0.19Mb. A skin biopsy was analysed using electronic microscopy showing an alteration of the structure and organisation of the dermal and peri-neuronal basement membrane. The relation between the phenotype and the deletion of both COL4A4 and COL4A3 genes, located in 2q36.3 loci, as well as the disruption of TRIP12 were discussed.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 2 , Cistos/genética , Deficiência Intelectual/genética , Nefropatias/genética , Pré-Escolar , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Hibridização de Ácido Nucleico
16.
Clin Chem ; 54(9): 1564-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18755906

RESUMO

BACKGROUND: Precise genotyping of the intron 8 poly(TG) and poly(T) tracts of the cystic fibrosis transmembrane conductance regulator (CFTR) gene is of clinical relevance in CFTR pathology. The (TG)(m) locus influences the penetrance of the (T)(5) allele, which may be associated with male infertility by congenital bilateral absence of the vas deferens (CBAVD) or other CFTR-related disorders (CFTR-RD), in particular in the context of (TG)(12) and (TG)(13). Simple and accurate genotyping of both loci should thus be routinely offered in laboratories. METHODS: We designed a new single test method relying on multiplex allele-specific fluorescent PCR: (T)(5)-, (T)(7)-, and (T)(9)-specific primers, labeled with different fluorophores, in combination with a common primer. Each fluorescent PCR product was identified on a capillary sequencer by its fluorescence color, specific for (T)(n), and size, indicative of the (TG) length. We first validated the assay in 2 different laboratories on 52 DNA samples with already known genotypes. We then evaluated the method prospectively, compared with sequencing, on 62 samples from healthy individuals and 108 samples from patients with CBAVD or other CFTR-RDs. RESULTS: We observed a 100% match in both validation steps. Results found in CBAVD and CFTR-RD patients are in keeping with data in the literature. CONCLUSIONS: The assay proved to be simple, rapid, and accurate for single-test (TG)(m)(T)(n) genotyping and suited for analysis in clinical laboratories.


Assuntos
Alelos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Íntrons/genética , Reação em Cadeia da Polimerase/métodos , Espectrometria de Fluorescência/métodos , Haplótipos , Humanos
17.
Haematologica ; 93(7): 1072-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18492694

RESUMO

Thirty-six unrelated cases with erythrocytosis of unknown origin were investigated. Exons 5-8 of the erythropoietin receptor gene (EPOR), the von Hippel-Lindau gene, and the prolyl hydroxylase domain protein 2 gene (PHD2) were screened by direct DNA sequencing. The Janus kinase 2 mutation, JAK2 (Val617Phe), was screened by allele specific PCR. In this study, three new mutations of EPOR causing deletions in exon 8 were found: the first led directly to a stop codon [g.5957_5958delTT (p.Phe424X)], the second to a stop codon after one residue [g.5828_5829delCC (p.Pro381GlnfsX1)] and the third to a stop codon following a frameshift sequence of 23 residues [g.5971delC (p.Leu429TrpfsX23)]. One patient had a previously reported EPOR mutation [g.6146A>G (p.Asn487Ser)] and another, a silent one (g.5799G>A). All were heterozygotes. In addition, 2 patients were positive for JAK2 (Val617Phe), and 2 reported elsewhere, were mutated in the PHD2 gene [c.606delG (p.Met202IlefsX71).


Assuntos
Regulação da Expressão Gênica , Mutação , Policitemia/diagnóstico , Policitemia/genética , Receptores da Eritropoetina/genética , Adolescente , Adulto , Idoso , Criança , Feminino , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Masculino , Pessoa de Meia-Idade , Pró-Colágeno-Prolina Dioxigenase/genética , Receptores da Eritropoetina/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
18.
Biochim Biophys Acta ; 1783(6): 1189-99, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18267124

RESUMO

Cystic fibrosis is mainly caused by mutations that interfere with the biosynthetic folding of the cystic fibrosis transmembrane conductance regulator (CFTR) protein. The aim of this study was to find cellular proteins interacting with CFTR and regulating its processing. We have used a genetic screen in yeast to identify such proteins and identified CSN5 that interacted with the third cytoplasmic loop of CFTR. CSN5 is the 5th component of the COP9 signalosome, a complex of eight subunits that shares significant homologies to the lid subcomplex of the 26S proteasome and controls the stability of many proteins. The present study shows that CSN5 associates with the core-glycosylated form of CFTR and suggests that this association targets misfolded CFTR to the degradative pathway. Identifying CSN5 as a new component of the degradative pathway is an important step towards the goal of unraveling the sorting between misfolded and correctly folded CFTR proteins.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeo Hidrolases/metabolismo , Dobramento de Proteína , Western Blotting , Complexo do Signalossomo COP9 , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Glicosilação , Células HeLa , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Peptídeo Hidrolases/genética , Ligação Proteica , Proteínas Tirosina Quinases/antagonistas & inibidores , Saccharomyces cerevisiae , Estilbenos/farmacologia , Frações Subcelulares , Técnicas do Sistema de Duplo-Híbrido
19.
J Cyst Fibros ; 7(5): 440-3, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18243066

RESUMO

BACKGROUND: The epidemiology of cystic fibrosis (CF) is poorly known in North African populations, in particular in Morocco and the CF carrier frequency in the general Moroccan population has never been evaluated. METHODS: To estimate the prevalence of CF mutations in Morocco, blood samples from 150 healthy Moroccans were tested for frequent CFTR mutations and the intron 8 polyT variant. RESULTS: Two subjects were heterozygous for F508del and eight others for the (T)5 variant. CONCLUSION: These findings indicate that the Moroccan population is at risk for CF and CFTR-related disorders. CF prevalence could be in the range of that found in European populations. Wider studies are necessary to identify the clinical pattern and accurately determine the prevalence and molecular basis of CF in Morocco.


Assuntos
Fibrose Cística/epidemiologia , Adolescente , Adulto , Portador Sadio , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Marrocos/epidemiologia , Mutação , Prevalência
20.
Hum Reprod ; 22(5): 1285-91, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17329263

RESUMO

BACKGROUND: Mutations in the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) gene have been widely detected in infertile men with congenital bilateral absence of the vas deferens (CBAVD). Despite extensive analysis of the CFTR gene using varied screening methods, a number of cases remain unsolved and could be attributable to the presence of large gene rearrangements, as recently shown for CF patients. METHODS: We carried out a complete CFTR gene study in a group of 222 CBAVD patients with strict diagnosis criteria and without renal anomaly, and searched for rearrangements using a semi-quantitative assay in a subgroup of 61 patients. RESULTS: The overall mutation detection rate was 87.8%, and 82% of patients carried two mutations. Ten out of the 99 different mutations accounted for 74.6% of identified alleles. Four large rearrangements were found in patients who already carried a mild mutation: two known partial deletions (exons 17a to 18 and 22 to 23), a complete deletion and a new partial duplication (exons 11 to 13). The rearrangements accounted for 7% of the previously unknown alleles and 1% of all identified alleles. CONCLUSIONS: Screening for rearrangements should be part of comprehensive CFTR gene studies in CBAVD patients and may have impacts on genetic counselling for the patients and their families.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Rearranjo Gênico/genética , Aconselhamento Genético , Ducto Deferente/anormalidades , França , Deleção de Genes , Genótipo , Humanos , Masculino , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA