Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
HGG Adv ; 3(1): 100074, 2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35047859

RESUMO

Robinow syndrome (RS) is a genetically heterogeneous disorder with six genes that converge on the WNT/planar cell polarity (PCP) signaling pathway implicated (DVL1, DVL3, FZD2, NXN, ROR2, and WNT5A). RS is characterized by skeletal dysplasia and distinctive facial and physical characteristics. To further explore the genetic heterogeneity, paralog contribution, and phenotypic variability of RS, we investigated a cohort of 22 individuals clinically diagnosed with RS from 18 unrelated families. Pathogenic or likely pathogenic variants in genes associated with RS or RS phenocopies were identified in all 22 individuals, including the first variant to be reported in DVL2. We retrospectively collected medical records of 16 individuals from this cohort and extracted clinical descriptions from 52 previously published cases. We performed Human Phenotype Ontology (HPO) based quantitative phenotypic analyses to dissect allele-specific phenotypic differences. Individuals with FZD2 variants clustered into two groups with demonstrable phenotypic differences between those with missense and truncating alleles. Probands with biallelic NXN variants clustered together with the majority of probands carrying DVL1, DVL2, and DVL3 variants, demonstrating no phenotypic distinction between the NXN-autosomal recessive and dominant forms of RS. While phenotypically similar diseases on the RS differential matched through HPO analysis, clustering using phenotype similarity score placed RS-associated phenotypes in a unique cluster containing WNT5A, FZD2, and ROR2 apart from non-RS-associated paralogs. Through human phenotype analyses of this RS cohort and OMIM clinical synopses of Mendelian disease, this study begins to tease apart specific biologic roles for non-canonical WNT-pathway proteins.

2.
Front Mol Neurosci ; 13: 12, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116545

RESUMO

Multiple TREX mRNA export complex subunits (e.g., THOC1, THOC2, THOC5, THOC6, THOC7) have now been implicated in neurodevelopmental disorders (NDDs), neurodegeneration and cancer. We previously implicated missense and splicing-defective THOC2 variants in NDDs and a broad range of other clinical features. Here we report 10 individuals from nine families with rare missense THOC2 variants including the first case of a recurrent variant (p.Arg77Cys), and an additional individual with an intragenic THOC2 microdeletion (Del-Ex37-38). Ex vivo missense variant testing and patient-derived cell line data from current and published studies show 9 of the 14 missense THOC2 variants result in reduced protein stability. The splicing-defective and deletion variants result in a loss of small regions of the C-terminal THOC2 RNA binding domain (RBD). Interestingly, reduced stability of THOC2 variant proteins has a flow-on effect on the stability of the multi-protein TREX complex; specifically on the other NDD-associated THOC subunits. Our current, expanded cohort refines the core phenotype of THOC2 NDDs to language disorder and/or ID, with a variable severity, and disorders of growth. A subset of affected individuals' has severe-profound ID, persistent hypotonia and respiratory abnormalities. Further investigations to elucidate the pathophysiological basis for this severe phenotype are warranted.

3.
Nat Genet ; 48(7): 777-84, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27213289

RESUMO

We identified biallelic mutations in NANS, the gene encoding the synthase for N-acetylneuraminic acid (NeuNAc; sialic acid), in nine individuals with infantile-onset severe developmental delay and skeletal dysplasia. Patient body fluids showed an elevation in N-acetyl-D-mannosamine levels, and patient-derived fibroblasts had reduced NANS activity and were unable to incorporate sialic acid precursors into sialylated glycoproteins. Knockdown of nansa in zebrafish embryos resulted in abnormal skeletal development, and exogenously added sialic acid partially rescued the skeletal phenotype. Thus, NANS-mediated synthesis of sialic acid is required for early brain development and skeletal growth. Normal sialylation of plasma proteins was observed in spite of NANS deficiency. Exploration of endogenous synthesis, nutritional absorption, and rescue pathways for sialic acid in different tissues and developmental phases is warranted to design therapeutic strategies to counteract NANS deficiency and to shed light on sialic acid metabolism and its implications for human nutrition.


Assuntos
Doenças do Desenvolvimento Ósseo/patologia , Encéfalo/embriologia , Deficiências do Desenvolvimento/patologia , Mutação/genética , Oxo-Ácido-Liases/genética , Ácidos Siálicos/metabolismo , Peixe-Zebra/embriologia , Adulto , Idade de Início , Animais , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Pré-Escolar , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/metabolismo , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
4.
J Clin Invest ; 125(9): 3585-99, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26280580

RESUMO

The genetic disorder Kabuki syndrome (KS) is characterized by developmental delay and congenital anomalies. Dominant mutations in the chromatin regulators lysine (K)-specific methyltransferase 2D (KMT2D) (also known as MLL2) and lysine (K)-specific demethylase 6A (KDM6A) underlie the majority of cases. Although the functions of these chromatin-modifying proteins have been studied extensively, the physiological systems regulated by them are largely unknown. Using whole-exome sequencing, we identified a mutation in RAP1A that was converted to homozygosity as the result of uniparental isodisomy (UPD) in a patient with KS and a de novo, dominant mutation in RAP1B in a second individual with a KS-like phenotype. We elucidated a genetic and functional interaction between the respective KS-associated genes and their products in zebrafish models and patient cell lines. Specifically, we determined that dysfunction of known KS genes and the genes identified in this study results in aberrant MEK/ERK signaling as well as disruption of F-actin polymerization and cell intercalation. Moreover, these phenotypes could be rescued in zebrafish models by rebalancing MEK/ERK signaling via administration of small molecule inhibitors of MEK. Taken together, our studies suggest that the KS pathophysiology overlaps with the RASopathies and provide a potential direction for treatment design.


Assuntos
Anormalidades Múltiplas/genética , Exoma , Face/anormalidades , Doenças Hematológicas/genética , Sistema de Sinalização das MAP Quinases/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Mutação , Proteínas de Ligação a Telômeros/genética , Doenças Vestibulares/genética , Proteínas de Peixe-Zebra/genética , Anormalidades Múltiplas/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Bovinos , Criança , Pré-Escolar , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Doenças Hematológicas/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Humanos , Masculino , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratos , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo , Doenças Vestibulares/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
5.
Eur J Med Genet ; 58(9): 455-65, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26206081

RESUMO

INTRODUCTION: Oculo-auriculo-vertebral spectrum (OAVS OMIM 164210) is a craniofacial developmental disorder affecting the development of the structures derived from the 1st and the 2nd branchial arches during embryogenesis, with consequential maxillary, mandibular, and ear abnormalities. The phenotype in OAVS is variable and associated clinical features can involve the cardiac, renal, skeletal, and central nervous systems. Its aetiology is still poorly understood. METHODS: We have evaluated the clinical phenotypes of 51 previously unpublished patients with OAVS and their parents, and performed comparative genomic hybridization microarray studies to identify potential causative loci. RESULTS: Of all 51 patients, 16 (31%) had a family history of OAVS. Most had no relevant pre-natal history and only 5 (10%) cases had a history of environmental exposures that have previously been described as risk factors for OAVS. In 28 (55%) cases, the malformations were unilateral. When the involvement was bilateral, it was asymmetric. Ear abnormalities were present in 47 (92%) patients (unilateral in 24; and bilateral in 23). Hearing loss was common (85%), mostly conductive, but also sensorineural, or a combination of both. Hemifacial microsomia was present in 46 (90%) patients (17 also presented facial nerve palsy). Ocular anomalies were present in 15 (29%) patients. Vertebral anomalies were confirmed in 10 (20%) cases; 50% of those had additional heart, brain and/or other organ abnormalities. Brain abnormalities were present in 5 (10%) patients; developmental delay was more common among these patients. Limb abnormalities were found in 6 (12%) patients, and urogenital anomalies in 5 (10%). Array-CGH analysis identified 22q11 dosage anomalies in 10 out of 22 index cases screened. DISCUSSION: In this study we carried out in-depth phenotyping of OAVS in a large, multicentre cohort. Clinical characteristics are in line with those reported previously, however, we observed a higher incidence of hemifacial microsomia and lower incidence of ocular anomalies. Furthermore our data suggests that OAVS patients with vertebral anomalies or congenital heart defects have a higher frequency of additional brain, limb or other malformations. We had a higher rate of familial cases in our cohort in comparison with previous reports, possibly because these cases were referred preferentially to our genetic clinic where family members underwent examination. We propose that familial OAVS cases show phenotypic variability, hence, affected relatives might have been misclassified in previous reports. Moreover, in view of its phenotypic variability, OAVS is potentially a spectrum of conditions, which overlap with other conditions, such as mandibulofacial dysostosis. Array CGH in our cohort identified recurrent dosage anomalies on 22q11, which may contribute to, or increase the risk of OAVS. We hypothesize that although the 22q11 locus may harbour gene(s) or regulatory elements that play a role in the regulation of craniofacial symmetry and 1st and 2nd branchial arch development, OAVS is a heterogeneous condition and many cases have a multifactorial aetiology or are caused by mutations in as yet unidentified gene(s).


Assuntos
Deficiências do Desenvolvimento/genética , Síndrome de Goldenhar/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Encéfalo/anormalidades , Hibridização Genômica Comparativa , Orelha/anormalidades , Orelha/embriologia , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/genética , Feminino , Síndrome de Goldenhar/diagnóstico , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Hérnia Diafragmática/diagnóstico , Hérnia Diafragmática/genética , Humanos , Masculino , Coluna Vertebral/anormalidades
6.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24442880

RESUMO

OBJECTIVES: Leri's pleonosteosis (LP) is an autosomal dominant rheumatic condition characterised by flexion contractures of the interphalangeal joints, limited motion of multiple joints, and short broad metacarpals, metatarsals and phalanges. Scleroderma-like skin thickening can be seen in some individuals with LP. We undertook a study to characterise the phenotype of LP and identify its genetic basis. METHODS AND RESULTS: Whole-genome single-nucleotide polymorphism genotyping in two families with LP defined microduplications of chromosome 8q22.1 as the cause of this condition. Expression analysis of dermal fibroblasts from affected individuals showed overexpression of two genes, GDF6 and SDC2, within the duplicated region, leading to dysregulation of genes that encode proteins of the extracellular matrix and downstream players in the transforming growth factor (TGF)-ß pathway. Western blot analysis revealed markedly decreased inhibitory SMAD6 levels in patients with LP. Furthermore, in a cohort of 330 systemic sclerosis cases, we show that the minor allele of a missense SDC2 variant, p.Ser71Thr, could confer protection against disease (p<1×10(-5)). CONCLUSIONS: Our work identifies the genetic cause of LP in these two families, demonstrates the phenotypic range of the condition, implicates dysregulation of extracellular matrix homoeostasis genes in its pathogenesis, and highlights the link between TGF-ß/SMAD signalling, growth/differentiation factor 6 and syndecan-2. We propose that LP is an additional member of the growing 'TGF-ß-pathies' group of musculoskeletal disorders, which includes Myhre syndrome, acromicric dysplasia, geleophysic dysplasias, Weill-Marchesani syndromes and stiff skin syndrome. Identification of a systemic sclerosis-protective SDC2 variant lays the foundation for exploration of the role of syndecan-2 in systemic sclerosis in the future.


Assuntos
Cromossomos Humanos Par 8/genética , Duplicação Gênica , Fator 6 de Diferenciação de Crescimento/genética , Deformidades Congênitas da Mão/genética , Artropatias/congênito , Ossificação Heterotópica/genética , Escleroderma Sistêmico/genética , Sindecana-2/genética , Adulto , Idoso , Pré-Escolar , Matriz Extracelular/metabolismo , Fácies , Feminino , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Fator 6 de Diferenciação de Crescimento/metabolismo , Deformidades Congênitas da Mão/metabolismo , Deformidades Congênitas da Mão/fisiopatologia , Humanos , Lactente , Artropatias/genética , Artropatias/metabolismo , Artropatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/fisiopatologia , Fenótipo , Transdução de Sinais , Sindecana-2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
7.
Am J Med Genet A ; 155A(7): 1511-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21671394

RESUMO

Kabuki syndrome is a rare, multiple malformation disorder characterized by a distinctive facial appearance, cardiac anomalies, skeletal abnormalities, and mild to moderate intellectual disability. Simplex cases make up the vast majority of the reported cases with Kabuki syndrome, but parent-to-child transmission in more than a half-dozen instances indicates that it is an autosomal dominant disorder. We recently reported that Kabuki syndrome is caused by mutations in MLL2, a gene that encodes a Trithorax-group histone methyltransferase, a protein important in the epigenetic control of active chromatin states. Here, we report on the screening of 110 families with Kabuki syndrome. MLL2 mutations were found in 81/110 (74%) of families. In simplex cases for which DNA was available from both parents, 25 mutations were confirmed to be de novo, while a transmitted MLL2 mutation was found in two of three familial cases. The majority of variants found to cause Kabuki syndrome were novel nonsense or frameshift mutations that are predicted to result in haploinsufficiency. The clinical characteristics of MLL2 mutation-positive cases did not differ significantly from MLL2 mutation-negative cases with the exception that renal anomalies were more common in MLL2 mutation-positive cases. These results are important for understanding the phenotypic consequences of MLL2 mutations for individuals and their families as well as for providing a basis for the identification of additional genes for Kabuki syndrome.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ligação a DNA/genética , Doenças Hematológicas/genética , Mutação/genética , Proteínas de Neoplasias/genética , Doenças Vestibulares/genética , Anormalidades Múltiplas/diagnóstico , Alelos , Face/anormalidades , Ordem dos Genes , Testes Genéticos , Genótipo , Doenças Hematológicas/diagnóstico , Humanos , Fenótipo , Prognóstico , Doenças Vestibulares/diagnóstico
8.
Eur J Hum Genet ; 19(1): 18-22, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20717171

RESUMO

Severe congenital neutropenia type 4 (SCN4) is an autosomal recessive condition, which was defined recently with identification of the causative mutations in G6PC3. To date there are only three reports in the literature describing patients with SCN4 with mutations in the G6PC3 gene. We report four individuals with SCN4 who belong to a single large consanguineous kindred. We provide an overview of the non-haematological features of the condition with a focus on the adult phenotype, which has not been previously described in detail. We show that the superficial venous changes seen in SCN4 patients can develop into varicose veins and venous ulcers in adulthood. We review the range of congenital anomalies associated with SCN4. We demonstrate that secundum atrial septal defect, patent ductus arteriosus and valvular defects are the most frequent cardiac anomalies in SCN4. Drawing parallels with type 1 glycogen storage disease, we propose that poor growth of prenatal onset, mild-to-moderate learning disability, primary pulmonary hypertension, delayed or incomplete puberty, hypothyroidism and dysmorphism likely represent features of this syndrome. We also suggest monitoring for lipid anomalies, and kidney and liver function in affected patients. Delineation of the SCN4 phenotype may help in appropriate treatment and management and provide further insights into the pathogenesis of this multisystem disease.


Assuntos
Glucose-6-Fosfatase/genética , Mutação , Adolescente , Adulto , Pré-Escolar , Síndrome Congênita de Insuficiência da Medula Óssea , Consanguinidade , Deficiências do Desenvolvimento/genética , Éxons/genética , Feminino , Doença de Depósito de Glicogênio Tipo I/genética , Cardiopatias Congênitas/genética , Humanos , Deficiências da Aprendizagem/genética , Masculino , Neutropenia/congênito , Neutropenia/genética , Neutropenia/fisiopatologia , Linhagem , Fenótipo
9.
Clin Dysmorphol ; 20(1): 15-20, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21084979

RESUMO

Microdeletion of 17q21.31 causes a recurrent recognizable dysmorphic syndrome. A further four patients with 17q21.31 microdeletions are reported here in whom an earlier diagnosis of cardio-facio-cutaneous syndrome was suggested. These patients have significant similarities of facial gestalt to earlier reported 17q21.31 microdeletion patients, but a striking feature that has not been emphasized previously is the large number of naevi and other pigmentary skin abnormalities that may be present. These features, together with a coarse facial appearance, relative macrocephaly and significant learning disabilities, were what had led to the earlier diagnostic suggestion of cardio-facio-cutaneous syndrome in each of these four cases.


Assuntos
Pele/patologia , Adolescente , Adulto , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 17/genética , Diagnóstico Diferencial , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Fácies , Insuficiência de Crescimento/diagnóstico , Insuficiência de Crescimento/genética , Feminino , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Humanos , Recém-Nascido , Masculino , Síndrome de Smith-Magenis , Adulto Jovem
10.
Hum Mutat ; 31(10): 1142-54, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20672375

RESUMO

A range of phenotypes including Greig cephalopolysyndactyly and Pallister-Hall syndromes (GCPS, PHS) are caused by pathogenic mutation of the GLI3 gene. To characterize the clinical variability of GLI3 mutations, we present a subset of a cohort of 174 probands referred for GLI3 analysis. Eighty-one probands with typical GCPS or PHS were previously reported, and we report the remaining 93 probands here. This includes 19 probands (12 mutations) who fulfilled clinical criteria for GCPS or PHS, 48 probands (16 mutations) with features of GCPS or PHS but who did not meet the clinical criteria (sub-GCPS and sub-PHS), 21 probands (6 mutations) with features of PHS or GCPS and oral-facial-digital syndrome, and 5 probands (1 mutation) with nonsyndromic polydactyly. These data support previously identified genotype-phenotype correlations and demonstrate a more variable degree of severity than previously recognized. The finding of GLI3 mutations in patients with features of oral-facial-digital syndrome supports the observation that GLI3 interacts with cilia. We conclude that the phenotypic spectrum of GLI3 mutations is broader than that encompassed by the clinical diagnostic criteria, but the genotype-phenotype correlation persists. Individuals with features of either GCPS or PHS should be screened for mutations in GLI3 even if they do not fulfill clinical criteria.


Assuntos
Anormalidades Múltiplas/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação , Proteínas do Tecido Nervoso/genética , Síndrome de Pallister-Hall/patologia , Polidactilia/patologia , Sindactilia/patologia , Anormalidades Craniofaciais/genética , Genótipo , Humanos , Anormalidades da Boca/genética , Síndrome de Pallister-Hall/genética , Fenótipo , Polidactilia/genética , Sindactilia/genética , Proteína Gli3 com Dedos de Zinco
12.
Nat Genet ; 41(1): 95-100, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19079258

RESUMO

Abnormalities in WNT signaling are implicated in a broad range of developmental anomalies and also in tumorigenesis. Here we demonstrate that germline mutations in WTX (FAM123B), a gene that encodes a repressor of canonical WNT signaling, cause an X-linked sclerosing bone dysplasia, osteopathia striata congenita with cranial sclerosis (OSCS; MIM300373). This condition is typically characterized by increased bone density and craniofacial malformations in females and lethality in males. The mouse homolog of WTX is expressed in the fetal skeleton, and alternative splicing implicates plasma membrane localization of WTX as a factor associated with survival in males with OSCS. WTX has also been shown to be somatically inactivated in 11-29% of cases of Wilms tumor. Despite being germline for such mutations, individuals with OSCS are not predisposed to tumor development. The observed phenotypic discordance dependent upon whether a mutation is germline or occurs somatically suggests the existence of temporal or spatial constraints on the action of WTX during tumorigenesis.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Lesões Pré-Cancerosas/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal , Adolescente , Adulto , Processamento Alternativo/genética , Animais , Doenças do Desenvolvimento Ósseo/complicações , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Fenótipo , Mutação Puntual , Estrutura Terciária de Proteína , Esclerose , Proteínas Supressoras de Tumor/química , Tumor de Wilms/genética , Inativação do Cromossomo X/genética
13.
Per Med ; 5(5): 435-439, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29783442

RESUMO

It is now widely agreed that genetics is set to play a major role in healthcare; 'personalized' medicine, new approaches for treating cancer, and stem cell therapies are typical of the exciting areas of development, whilst large-scale epidemiological studies are uncovering genetic factors in pathways involved in complex disease. Nowgen, A Center for Genetics in Healthcare is a publicly funded organization examining the wider impact of genetics and genomics on the delivery of 21st century medicine. We recognize that the adoption of new technologies and treatments will be shaped by many factors, including public and patient beliefs and attitudes, the knowledge of professionals, and wider business approaches to these innovations. Our role is also to consider these developments within the UK National Health Service. We place ourselves at the center of the current discourse between these groups, setting out to perform applied research, inform and canvass opinion, disseminate knowledge, monitor developments and inform policy relating to the future of genetic medicine.

14.
Am J Hum Genet ; 80(6): 1162-70, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17503333

RESUMO

Carpenter syndrome is a pleiotropic disorder with autosomal recessive inheritance, the cardinal features of which include craniosynostosis, polysyndactyly, obesity, and cardiac defects. Using homozygosity mapping, we found linkage to chromosome 6p12.1-q12 and, in 15 independent families, identified five different mutations (four truncating and one missense) in RAB23, which encodes a member of the RAB guanosine triphosphatase (GTPase) family of vesicle transport proteins and acts as a negative regulator of hedgehog (HH) signaling. In 10 patients, the disease was caused by homozygosity for the same nonsense mutation, L145X, that resides on a common haplotype, indicative of a founder effect in patients of northern European descent. Surprisingly, nonsense mutations of Rab23 in open brain mice cause recessive embryonic lethality with neural-tube defects, suggesting a species difference in the requirement for RAB23 during early development. The discovery of RAB23 mutations in patients with Carpenter syndrome implicates HH signaling in cranial-suture biogenesis--an unexpected finding, given that craniosynostosis is not usually associated with mutations of other HH-pathway components--and provides a new molecular target for studies of obesity.


Assuntos
Acrocefalossindactilia/genética , Suturas Cranianas/crescimento & desenvolvimento , Proteínas Hedgehog/fisiologia , Mutação , Obesidade , Proteínas rab de Ligação ao GTP/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 6 , Genes Recessivos , Ligação Genética , Humanos , Transdução de Sinais , Síndrome
15.
Eur J Hum Genet ; 13(5): 547-55, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15756302

RESUMO

The skeletal dysplasias are a clinically and genetically heterogeneous group of conditions affecting the development of the osseous skeleton and fall into the category of rare genetic diseases in which the diagnosis can be difficult for the nonexpert. Two such diseases are pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED), which result in varying degrees of short stature, joint pain and stiffness and often resulting in early onset osteoarthritis. PSACH and some forms of MED result from mutations in the cartilage oligomeric matrix protein (COMP) gene and to aid the clinical diagnosis and counselling of patients with a suspected diagnosis of PSACH or MED, we developed an efficient and accurate molecular diagnostic service for the COMP gene. In a 36-month period, 100 families were screened for a mutation in COMP and we identified disease-causing mutations in 78% of PSACH families and 36% of MED families. Furthermore, in several of these families, the identification of a disease-causing mutation provided information that was immediately used to direct reproductive decision-making.


Assuntos
Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Mutação , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Adolescente , Proteína de Matriz Oligomérica de Cartilagem , Análise Mutacional de DNA , Feminino , Aconselhamento Genético/métodos , Testes Genéticos/métodos , Humanos , Lactente , Masculino , Proteínas Matrilinas , Mutação Puntual , Polimorfismo de Nucleotídeo Único
16.
Nat Genet ; 36(4): 411-6, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15004558

RESUMO

Lenz microphthalmia is inherited in an X-linked recessive pattern and comprises microphthalmia, mental retardation, and skeletal and other anomalies. Two loci associated with this syndrome, MAA (microphthalmia with associated anomalies) and MAA2, are situated respectively at Xq27-q28 (refs. 1,2) and Xp11.4-p21.2 (ref. 3). We identified a substitution, nt 254C-->T; P85L, in BCOR (encoding BCL-6-interacting corepressor, BCOR) in affected males from the family with Lenz syndrome previously used to identify the MAA2 locus. Oculofaciocardiodental syndrome (OFCD; OMIM 300166) is inherited in an X-linked dominant pattern with presumed male lethality and comprises microphthalmia, congenital cataracts, radiculomegaly, and cardiac and digital abnormalities. Given their phenotypic overlap, we proposed that OFCD and MAA2-associated Lenz microphthalmia were allelic, and we found different frameshift, deletion and nonsense mutations in BCOR in seven families affected with OFCD. Like wild-type BCOR, BCOR P85L and an OFCD-mutant form of BCOR can interact with BCL-6 and efficiently repress transcription. This indicates that these syndromes are likely to result from defects in alternative functions of BCOR, such as interactions with transcriptional partners other than BCL-6. We cloned the zebrafish (Danio rerio) ortholog of BCOR and found that knock-down of this ortholog caused developmental perturbations of the eye, skeleton and central nervous system consistent with the human syndromes, confirming that BCOR is a key transcriptional regulator during early embryogenesis.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades do Olho/genética , Cardiopatias Congênitas/genética , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Sequência de Aminoácidos , Animais , Mecanismo Genético de Compensação de Dose , Face/anormalidades , Humanos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/química , Proteínas Repressoras/química , Homologia de Sequência de Aminoácidos , Síndrome , Anormalidades Dentárias/genética , Cromossomo X , Peixe-Zebra
17.
J Am Soc Nephrol ; 14(3): 680-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12595504

RESUMO

Oral-facial-digital syndrome type 1 (OFD1) causes polycystic kidney disease (PKD) and malformations of the mouth, face and digits. Recently, a gene on Xp22, OFD1, was reported to be mutated in a limited set of OFD1 patients. This study describes mutation analysis in six further OFD1 families. Additionally, gene expression was sought in human development. In two OFD1 kindreds affected by PKD, a frameshift mutation and a splice-site mutation were detected. In four apparently sporadic cases, three frameshift and a missense mutation were found. Using RT-PCR of RNA from first-trimester normal human embryos, both alternative splice forms of mRNA (OFD1a and OFD1b) were found to be widely expressed in organogenesis. Northern blot detected OFD1 mRNA in metanephros, brain, tongue, and limb, all organs affected in the syndrome. A polyclonal antibody directed to a C-terminal OFD1a epitope detected a 120-kD protein in the metanephros and in human renal mesenchymal cell lines. In normal human embryos, OFD1a immunolocalized to the metanephric mesenchyme, oral mucosa, nasal and cranial cartilage, and brain. Moreover, using normal human renal mesenchymal cell lines, the immunoreactive protein colocalized with gamma-tubulin, suggesting that OFD1 is associated with the centrosome. First, it is concluded that OFD1 mutations would generally be predicted to result in unstable transcripts or nonfunctional proteins. Second, OFD1 is expressed in human organogenesis; on the basis of the metanephric expression pattern, the results suggest that OFD1 plays a role in differentiation of metanephric precursor cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Rim/fisiologia , Síndromes Orofaciodigitais/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Mutação da Fase de Leitura , Humanos , Rim/citologia , Mesoderma/citologia , Proteínas/genética , Sítios de Splice de RNA/genética , Transdução de Sinais/fisiologia , Transfecção
18.
Hum Genet ; 110(5): 422-8, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12073012

RESUMO

TDGF1 (CRIPTO) is an EGF-CFC family member and an obligate co-receptor involved in NODAL signaling, a developmental program implicated in midline, forebrain, and left-right axis development in model organisms. Previous studies of CFC1 (CRYPTIC), another member of the EGF-CFC family, demonstrated that normal function of this protein is required for proper laterality development in humans. Here we identify a mutation in the conserved CFC domain of TDGF1 in a patient with midline anomalies of the forebrain. The mutant protein is inactive in a zebrafish rescue assay, indicating a role for TDGF1 in human midline and forebrain development.


Assuntos
Fator de Crescimento Epidérmico , Holoprosencefalia/genética , Proteínas de Homeodomínio , Glicoproteínas de Membrana , Mutação/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Prosencéfalo/anormalidades , Fatores de Transcrição , Proteínas de Peixe-Zebra , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Pré-Escolar , Feminino , Proteínas Ligadas por GPI , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Peixe-Zebra/embriologia , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA