Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Am J Med Genet A ; 191(7): 1849-1857, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37081310

RESUMO

Partial deletions at chromosome 7q11.23 are causative for the autosomal-dominant Williams-Beuren syndrome (WBS), whereas the partial duplication of this region leads to the 7q11.23 duplication syndrome. Both syndromes are highly penetrant and occur with a frequency of 1:7500-10,000 (WBS) and 1:13,000-20,000 (7q11.23 duplication syndrome). They are associated with multiple organ defects, intellectual disability, and typical facial dysmorphisms showing broad phenotypic variability. The 7q11.23 region is susceptible to chromosomal rearrangements due to flanking segmental duplications and regions of long repetitive DNA segments. Here, we report on a family with two children affected by WBS and clinically unaffected parents. Interestingly, metaphase fluorescence in situ hybridization (FISH) revealed a deletion on 7q11.23 in the father. Intensive genetic testing, using interphase FISH, whole genome sequencing and optical genome mapping led to the confirmation of a 1.5 Mb deletion at one 7q11.23 allele and the identification of a reciprocal 1.8 Mb duplication at the other allele. This finding is highly important regarding genetic counseling in this family. The father is a silent carrier for two syndromic disorders, thus his risk to transmit a disease-causing allele is 100%. To the best of our knowledge we, here, report on the first case in which the phenotype of a microdeletion/microduplication syndrome was compensated by its reciprocal counterpart.


Assuntos
Síndrome de Williams , Humanos , Hibridização in Situ Fluorescente , Síndrome de Williams/genética , Testes Genéticos , Fenótipo , Aberrações Cromossômicas , Cromossomos Humanos Par 7/genética , Deleção Cromossômica
2.
Hum Mutat ; 42(7): 862-876, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33942433

RESUMO

Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme involved in over 400 cellular reactions. During embryogenesis, mammals synthesize NAD de novo from dietary l -tryptophan via the kynurenine pathway. Biallelic, inactivating variants in three genes encoding enzymes of this biosynthesis pathway (KYNU, HAAO, and NADSYN1) disrupt NAD synthesis and have been identified in patients with multiple malformations of the heart, kidney, vertebrae, and limbs; these patients have Congenital NAD Deficiency Disorder HAAO and four families with biallelic variants in KYNU. These patients present similarly with multiple malformations of the heart, kidney, vertebrae, and limbs, of variable severity. We show that each variant identified in these patients results in loss-of-function, revealed by a significant reduction in NAD levels via yeast genetic complementation assays. For the first time, missense mutations are identified as a cause of malformation and shown to disrupt enzyme function. These missense and frameshift variants cause moderate to severe NAD deficiency in yeast, analogous to insufficient synthesized NAD in patients. We hereby expand the genotypic and corresponding phenotypic spectrum of Congenital NAD Deficiency Disorder.


Assuntos
NAD , Coluna Vertebral , Animais , Genótipo , Humanos , Mamíferos , Mutação de Sentido Incorreto , Coluna Vertebral/anormalidades
3.
J Hum Genet ; 65(2): 193-197, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31767933

RESUMO

Biallelic pathogenic variants in POC1A are ultra rare. They have been reported in 13 families as causing either Short stature, Onychodysplasia, Facial dysmorphism, and hypoTrichosis (SOFT) syndrome, or a milder partially overlapping phenotype, variant POC1A-related syndrome. This pleiotropic effect is likely precipitated by the variant's location and respective affected protein domain. Here, we describe seven patients from two consanguineous Omani families with classic SOFT syndrome and a novel homozygous POC1A variant (c.64G>T; p.(Val22Phe)), which is the first one described for the alternative exon 2. This result refines the POC1A mutational spectrum relevant for exertion of the described pleiotropic effect. Furthermore, six of our patients experienced recurrent mild to severe respiratory difficulties that have not been previously reported for SOFT syndrome and may be an underdiagnosed or a genotype-specific complication that warrants attention in future studies. Thus, our study unravels new aspects of the genotype-phenotype correlation suggested by previous reports.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ciclo Celular/genética , Anormalidades Craniofaciais/genética , Proteínas do Citoesqueleto/genética , Nanismo/genética , Estudos de Associação Genética , Hipotricose/genética , Atrofia Muscular/genética , Processamento Alternativo , Criança , Pré-Escolar , Consanguinidade , Éxons/genética , Feminino , Genótipo , Humanos , Masculino , Mutação , Fenótipo
4.
J Hum Genet ; 64(5): 409-419, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30816285

RESUMO

Autosomal Dominant Polycystic Kidney Disease (ADPKD) accounts for 2.6% of the patients with chronic kidney disease in India. ADPKD is caused by pathogenic variants in either PKD1 or PKD2 gene. There is no comprehensive genetic data from Indian subcontinent. We aimed to identify the pathogenic variants in the heterogeneous Indian population. PKD1 and PKD2 variants were identified by direct gene sequencing and/or multiplex ligation-dependent probe amplification (MLPA) in 125 unrelated patients of ADPKD. The pathogenic potential of the variants was evaluated computationally and were classified according to ACMG guidelines. Overall 300 variants were observed in PKD1 and PKD2 genes, of which 141 (47%) have been reported previously as benign. The remaining 159 variants were categorized into different classes based on their pathogenicity. Pathogenic variants were observed in 105 (84%) of 125 patients, of which 99 (94.3%) were linked to PKD1 gene and 6 (6.1%) to PKD2 gene. Of 159 variants, 97 were novel variants, of which 43 (44.33%) were pathogenic, and 10 (10.31%) were of uncertain significance. Our data demonstrate the diverse genotypic makeup of single gene disorders in India as compared to the West. These data would be valuable in counseling and further identification of probable donors among the relatives of patients with ADPKD.


Assuntos
Ligação Genética , Variação Genética , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Índia , Masculino , Pessoa de Meia-Idade
5.
BMC Med Genet ; 19(1): 85, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29801475

RESUMO

BACKGROUND: Dyskeratosis congenita (DC) is a rare genetic disorder of bone marrow failure inherited in an X-linked, autosomal dominant or autosomal recessive pattern. It has a wide array of clinical features and patients may be cared for by many medical sub specialties. The typical clinical features consist of lacy reticular skin pigmentation, nail dystrophy and oral leukoplakia. As the disease advances, patients may develop progressive bone marrow failure, pulmonary fibrosis, oesophageal stenosis, urethral stenosis, liver cirrhosis as well as haematological and solid malignancies. Several genes have been implicated in the pathogenesis of dyskeratosis congenita, with the dyskerin pseudouridine synthase 1 (DKC1) gene mutations being the X-linked recessive gene. CASE PRESENTATION: Herein, we report a 31-year-old male with history of recurrent febrile episodes who was found to have reticulate skin pigmentation interspersed with hypopigmented macules involving the face, neck and extremities, hyperkeratosis of palms and soles, nail dystrophy, leukoplakia of the tongue, premature graying of hair, watery eyes and dental caries. Several of his male relatives, including two maternal uncles and three maternal cousins were affected with a similar type of disease condition. Pedigree analysis suggested a possible X-linked pattern of inheritance. Genetic testing in the proband showed a novel hemizygous, non-synonymous likely pathogenic variant [NM_001363.4: c.1054A > G: p.Thr352Ala] in the PUA domain of the DKC1 gene. Quantitative polymerase chain reaction for relative telomere length measurements performed in the proband showed that he had very short telomeres [0.38, compared to a control median of 0.71 (range 0.44-1.19)], which is consistent with the DC diagnosis. Co-segregation analysis of the novel mutation and telomere length measurements in the extended family members could not be performed as they were unwilling to provide consent for testing. CONCLUSIONS: The novel variant detected in the DKC1 gene adds further to the existing scientific literature on the genotype-phenotype correlation of DC, and has important implications for the clinical and molecular characterization of the disease.


Assuntos
Proteínas de Ciclo Celular/genética , Disceratose Congênita/genética , Hemizigoto , Proteínas Nucleares/genética , Mutação Puntual , Adulto , Proteínas de Ciclo Celular/química , Humanos , Masculino , Proteínas Nucleares/química , Linhagem , Domínios Proteicos , Análise de Sequência de DNA , Homeostase do Telômero
6.
Neuropediatrics ; 49(6): 373-378, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30114719

RESUMO

Neonatal-onset movement disorders, especially in combination with seizures, are rare and often related to mitochondrial disorders. 3-methylglutaconic aciduria (3-MGA-uria) is a marker for mitochondrial dysfunction. In particular, consistently elevated urinary excretion of 3-methylglutaconic acid is the hallmark of a small but growing group of inborn errors of metabolism (IEM) due to defective phospholipid remodeling or mitochondrial membrane-associated disorders (mutations in TAZ, SERAC1, OPA3, CLPB, DNAJC19, TMEM70, TIMM50). Exome/genome sequencing is a powerful tool for the diagnosis of the clinically and genetically heterogeneous mitochondrial disorders. Here, we report 11 individuals, of whom 2 are previously unpublished, with biallelic variants in high temperature requirement protein A2 (HTRA2) encoding a mitochondria-localized serine protease. All individuals presented a recognizable phenotype with neonatal- or infantile-onset neurodegeneration and death within the first month of life. Hallmark features were central hypopnea/apnea leading to respiratory insufficiency, seizures, neutropenia, 3-MGA-uria, tonus dysregulation, and dysphagia. Tremor, jitteriness, dystonia, and/or clonus were also common. HTRA2 defect should be grouped under the IEM with 3-MGA-uria as discriminating feature. Clinical characteristics overlap with other disorders of this group suggesting a common underlying pathomechanism. Urinary organic acid analysis is a noninvasive and inexpensive test that can guide further genetic testing in children with suggestive clinical findings.


Assuntos
Deficiências do Desenvolvimento , Epilepsia , Serina Peptidase 2 de Requerimento de Alta Temperatura A/deficiência , Erros Inatos do Metabolismo , Doenças Mitocondriais , Transtornos dos Movimentos , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/genética , Distonia/diagnóstico , Distonia/etiologia , Distonia/genética , Epilepsia/diagnóstico , Epilepsia/etiologia , Epilepsia/genética , Evolução Fatal , Feminino , Humanos , Lactente , Recém-Nascido , Doenças do Recém-Nascido , Masculino , Erros Inatos do Metabolismo/complicações , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/genética , Doenças Mitocondriais/complicações , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Transtornos dos Movimentos/diagnóstico , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/genética , Insuficiência Respiratória/diagnóstico , Insuficiência Respiratória/etiologia , Insuficiência Respiratória/genética , Tremor/diagnóstico , Tremor/etiologia , Tremor/genética
7.
J Hum Genet ; 62(7): 723-727, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28356563

RESUMO

The iron-sulfur (Fe-S) cluster (ISC) biogenesis pathway is indispensable for many fundamental biological processes and pathogenic variations in genes encoding several components of the Fe-S biogenesis machinery, such as NFU1, BOLA3, IBA57 and ISCA2 are already implicated in causing four types of multiple mitochondrial dysfunctions syndromes (MMDS). We report on two unrelated families, with two affected children each with early onset neurological deterioration, seizures, extensive white matter abnormalities, cortical migrational abnormalities, lactic acidosis and early demise. Exome sequencing of two affected individuals, one from each family, revealed a homozygous c.259G>A [p.(Glu87Lys)] variant in ISCA1 and Mendelian segregation was confirmed in both families. The ISCA1 variant lies in the only shared region of homozygosity between the two families suggesting the possibility of a founder effect. In silico functional analyses and structural modeling of the protein predict the identified ISCA1 variant to be detrimental to protein stability and function. Notably the phenotype observed in all affected subjects with the ISCA1 pathogenic variant is similar to that previously described in all four types of MMDS. Our findings suggest association of a pathogenic variant in ISCA1 with another MMDS.


Assuntos
Predisposição Genética para Doença , Proteínas Ferro-Enxofre/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Mutação/genética , Sequência de Aminoácidos , Sequência de Bases , Pré-Escolar , Evolução Fatal , Feminino , Homozigoto , Humanos , Lactente , Proteínas Ferro-Enxofre/química , Masculino , Proteínas Mitocondriais/química , Linhagem , Síndrome
8.
Mol Cell Probes ; 30(1): 18-21, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26631803

RESUMO

The SOX18 gene encodes a transcription factor that plays a notable role in certain developmental contexts such as lymphangiogenesis, hair follicle development and vasculogenesis. SOX18 mutations are linked to recessive and dominant hypotrichosis-lymphedema-telangiectasia syndrome (HLTS). In this study we report on a novel heterozygous mutation in SOX18 in a Jordanian patient suffering from HLTS that was revealed by Whole Exome Sequencing. In this case, a frameshift caused by 14-nucleotide duplication in SOX18 appeared de novo resulting in a premature translational stop at the N-terminal region of the central trans-activation domain. Here we present the clinical manifestations of the above mentioned molecular lesion in the light of what is known from published SOX18 mutations.


Assuntos
Análise Mutacional de DNA/métodos , Exoma/genética , Hipotricose , Linfedema , Mutação , Fatores de Transcrição SOXF/genética , Telangiectasia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Sequência de Bases , Humanos , Recém-Nascido , Jordânia , Masculino , Dados de Sequência Molecular , Síndrome
9.
J Exp Med ; 203(7): 1671-7, 2006 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-16818677

RESUMO

Endothelial cell-selective adhesion molecule (ESAM) is specifically expressed at endothelial tight junctions and on platelets. To test whether ESAM is involved in leukocyte extravasation, we have generated mice carrying a disrupted ESAM gene and analyzed them in three different inflammation models. We found that recruitment of lymphocytes into inflamed skin was unaffected by the gene disruption. However, the migration of neutrophils into chemically inflamed peritoneum was inhibited by 70% at 2 h after stimulation, recovering at later time points. Analyzing neutrophil extravasation directly by intravital microscopy in the cremaster muscle revealed that leukocyte extravasation was reduced (50%) in ESAM(-/-) mice without affecting leukocyte rolling and adhesion. Depletion of >98% of circulating platelets did not abolish the ESAM deficiency-related inhibitory effect on neutrophil extravasation, indicating that it is only ESAM at endothelial tight junctions that is relevant for the extravasation process. Knocking down ESAM expression in endothelial cells resulted in reduced levels of activated Rho, a GTPase implicated in the destabilization of tight junctions. Indeed, vascular permeability stimulated by vascular endothelial growth factor was reduced in ESAM(-/-) mice. Collectively, ESAM at endothelial tight junctions participates in the migration of neutrophils through the vessel wall, possibly by influencing endothelial cell contacts.


Assuntos
Permeabilidade Capilar/imunologia , Moléculas de Adesão Celular/fisiologia , Movimento Celular/imunologia , Neutrófilos/patologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Permeabilidade Capilar/genética , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Comunicação Celular/genética , Comunicação Celular/imunologia , Movimento Celular/genética , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Neutrófilos/imunologia
10.
J Dtsch Dermatol Ges ; 14(6): 627-629, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27240080
11.
J Dtsch Dermatol Ges ; 14(6): 627-629, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27240081
12.
Mol Cell Biol ; 27(3): 983-92, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17130243

RESUMO

Dendritic cells (DCs) have long been recognized as key regulators of immune responses. However, the process of their recruitment to peripheral tissues and turnover during homeostasis remains largely unknown. The chemokine CXCL14 (BRAK) is constitutively expressed in skin and other epithelial tissues. Recently, the human chemokine was proposed to play a role in the homeostatic recruitment of macrophage and/or DC precursors toward the periphery, such as skin. Although so far no physiological function could be demonstrated for the murine CXCL14, it shows a remarkable homology to the human chemokine. In order to elucidate the in vivo role of CXCL14, we generated a mouse defective for this chemokine. We studied various components of the immune system with emphasis on monocytes/macrophages and DC/Langerhans cell (LC) populations in different tissues during steady state but did not find a significant difference between knockout (CXCL14(-)(/)(-)) and control mice. Functionally, LCs were able to become activated, to migrate out of skin, and to elicit a delayed type of hypersensitivity reaction. Overall, our data indicate that murine CXCL14 is dispensable for the homeostatic recruitment of antigen-presenting cells toward the periphery and for LC functionality.


Assuntos
Movimento Celular , Quimiocinas CXC/metabolismo , Células Dendríticas/citologia , Tecido Linfoide/citologia , Pele/citologia , Animais , Cruzamento , Contagem de Células , Quimiocinas CXC/deficiência , Células Dendríticas/imunologia , Epitélio/imunologia , Feminino , Marcação de Genes , Inflamação , Células de Langerhans/citologia , Células de Langerhans/imunologia , Tecido Linfoide/imunologia , Macrófagos Peritoneais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pele/imunologia , Pele/patologia , Cicatrização
13.
Clin Case Rep ; 7(8): 1522-1525, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31428380

RESUMO

Conradi-Hünermann-Happle syndrome is a rare X-linked dominant syndrome affecting the skin, skeletal system, and eyes. Here, we report on a female patient with a de novo heterozygous missense mutation c.301C>T (p.Trp101Arg) of the EMP (emopamil binding protein) gene.

14.
J Appl Genet ; 60(2): 151-162, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30706430

RESUMO

Autosomal recessive primary microcephaly (MCPH) is a group of rare neurodevelopmental diseases with severe microcephaly at birth. One type of the disorder, MCPH2, is caused by biallelic mutations in the WDR62 gene, which encodes the WD repeat-containing protein 62. Patients with WDR62 mutation may have a wide range of malformations of cortical development in addition to congenital microcephaly. We describe two patients, a boy and a girl, with severe congenital microcephaly, global developmental delay, epilepsy, and failure to thrive. MRI showed hemispherical asymmetry, diffuse pachygyria, thick gray matter, indistinct gray-white matter junction, and corpus callosum and white matter hypoplasia. Whole exome sequencing revealed the same novel homozygous missense mutation, c.668T>C, p.Phe223Ser in exon 6 of the WDR62 gene. The healthy parents were heterozygous for this mutation. The mutation affects a highly conserved region in one of the WD repeats of the WDR62 protein. Haplotype analysis showed genetic relatedness between the families of the patients. Our findings expand the spectrum of mutations randomly distributed in the WDR62 gene. A review is also provided of the brain malformations described in WDR62 mutations in association with congenital microcephaly.


Assuntos
Deficiências do Desenvolvimento/genética , Microcefalia/genética , Proteínas do Tecido Nervoso/genética , Proteínas de Ciclo Celular , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/patologia , Haplótipos , Homozigoto , Humanos , Masculino , Microcefalia/diagnóstico por imagem , Microcefalia/fisiopatologia , Mutação de Sentido Incorreto/genética , Linhagem , Substância Branca/diagnóstico por imagem , Substância Branca/patologia
15.
Mol Cell Biol ; 25(2): 699-705, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15632070

RESUMO

Tenomodulin (Tnmd) is a member of a new family of type II transmembrane glycoproteins. It is predominantly expressed in tendons, ligaments, and eyes, whereas the only other family member, chondromodulin I (ChM-I), is highly expressed in cartilage and at lower levels in the eye and thymus. The C-terminal extracellular domains of both proteins were shown to modulate endothelial-cell proliferation and tube formation in vitro and in vivo. We analyzed Tnmd function in vivo and provide evidence that Tnmd is processed in vivo and that the proteolytically cleaved C-terminal domain can be found in tendon extracts. Loss of Tnmd expression in gene targeted mice abated tenocyte proliferation and led to a reduced tenocyte density. The deposited amounts of extracellular matrix proteins, including collagen types I, II, III, and VI and decorin, lumican, aggrecan, and matrilin-2, were not affected, but the calibers of collagen fibrils varied significantly and exhibited increased maximal diameters. Tnmd-deficient mice did not have changes in tendon vessel density, and mice lacking both Tnmd and ChM-I had normal retinal vascularization and neovascularization after oxygen-induced retinopathy. These results suggest that Tnmd is a regulator of tenocyte proliferation and is involved in collagen fibril maturation but do not confirm an in vivo involvement of Tnmd in angiogenesis.


Assuntos
Proteínas de Membrana/metabolismo , Tendões/citologia , Tendões/fisiologia , Animais , Animais Recém-Nascidos , Colágeno/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Feminino , Marcação de Genes , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mucinas/metabolismo , Retina/citologia , Retina/metabolismo , Neovascularização Retiniana , Sialomucinas
16.
Ophthalmic Genet ; 39(6): 678-683, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30289319

RESUMO

PURPOSE: To report five novel genetic variants in seven unrelated consanguineous families with achromatopsia (ACHM). METHODS: Patients were examined with multimodal retinal imaging and full-field electroretinography (ffERG). Genetic testing was conducted using next-generation sequencing (NGS). RESULTS: Three novel homozygous variants were detected in CNGA3: a missense c.967G > C (p.Ala323Pro) variant was detected in exon 8 (one patient), a splice site variant c.101 + 1G > A in intron 2 (three patients), and a splice site variant c.395 + 1G > T in intron 4(one patient). Another two novel variants were found in PDE6C: a homozygous missense variant c.1899C > A (p.His633Gln) in exon 15 (one patient) and a homozygous splice site variant c.1072-1G > C in intron 7 (one patient). Mutation segregation assessment was possible in 3 of the 7 families. All patients had nonrecordable ffERG 30-Hz flicker responses, reduced single-flash cone responses but preserved rod responses. Patients presented with variable degrees of foveal outer retinal layer loss and variable patterns of foveal hyperautofluorescence. CONCLUSIONS: These novel variants expand the genotypes associated with ACHM and may help in future therapy development for ACHM.


Assuntos
Defeitos da Visão Cromática/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Proteínas do Olho/genética , Variação Genética , Adulto , Criança , Pré-Escolar , Defeitos da Visão Cromática/diagnóstico por imagem , Defeitos da Visão Cromática/fisiopatologia , Consanguinidade , Análise Mutacional de DNA , Eletrorretinografia , Éxons/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Íntrons/genética , Masculino , Imagem Multimodal , Retina/fisiopatologia , Estudos Retrospectivos , Tomografia de Coerência Óptica
17.
Eur J Hum Genet ; 26(4): 592-598, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29449720

RESUMO

Congenital neurological disorders are genetically highly heterogeneous. Rare forms of hereditary neurological disorders are still difficult to be adequately diagnosed. Pertinent studies, especially when reporting only single families, need independent confirmation. We present three unrelated families in which whole-exome sequencing identified the homozygous non-sense variants c.430[C>T];[C>T] p.(Arg144*), c.1219[C>T];[C>T] p.(Gln407*) and c.1408[C>T];[C>T] p.(Arg470*) in GTPBP2. Their clinical presentations include early onset and apparently non-progressive motor and cognitive impairment, and thereby overlap with findings in a recently described family harbouring a homozygous GTPBP2 splice site variant. Notable differences include structural brain abnormalities (e.g., agenesis of the corpus callosum, exclusive to our patients), and evidence for brain iron accumulation (exclusive to the previously described family). This report confirms pathogenicity of biallelic GTPBP2 inactivation and broadens the phenotypic spectrum. It also underlines that a potential involvement of brain iron accumulation needs clarification. Further patients will have to be identified and characterised in order to fully define the core features of GTPBP2-associated neurological disorder, but future approaches to molecular diagnosis of neurodevelopmental disorders should implement GTPBP2.


Assuntos
Agenesia do Corpo Caloso/genética , Deficiência Intelectual/genética , Sobrecarga de Ferro/genética , Mutação com Perda de Função , Proteínas Monoméricas de Ligação ao GTP/genética , Agenesia do Corpo Caloso/patologia , Alelos , Criança , Feminino , Proteínas de Ligação ao GTP , Humanos , Deficiência Intelectual/patologia , Sobrecarga de Ferro/patologia , Masculino , Fenótipo , Síndrome
18.
Mol Cell Biol ; 22(18): 6627-35, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12192060

RESUMO

Chondromodulin I (chm-I), a type II transmembrane protein, is highly expressed in the avascular zones of cartilage but is downregulated in the hypertrophic region, which is invaded by blood vessels during enchondral ossification. In vitro and in vivo assays with the purified protein have shown chondrocyte-modulating and angiogenesis-inhibiting functions. To investigate chm-I function in vivo, we generated transgenic mice lacking chm-I mRNA and protein. Null mice are viable and fertile and show no morphological changes. No abnormalities in vascular invasion and cartilage development were detectable. No evidence was found for a compensating function of tendin, a recently published homologue highly expressed in tendons and also, at low levels, in cartilage. Furthermore, no differences in the expression of other angiogenic or antiangiogenic factors such as transforming growth factor beta1 (TGF-beta1), TGF-beta2, TGF-beta3, fibroblast growth factor 2, and vascular endothelial growth factor were found. The surprising lack of phenotype in the chm-I-deficient mice suggests either a different function for chm-I in vivo than has been proposed or compensatory changes in uninvestigated angiogenic or angiogenesis-inhibiting factors. Further analysis using double-knockout technology will be necessary to analyze the function of chm-I in the complex process of enchondral ossification.


Assuntos
Cartilagem/embriologia , Cartilagem/crescimento & desenvolvimento , Olho/embriologia , Olho/crescimento & desenvolvimento , Substâncias de Crescimento/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana , Osteogênese , Alelos , Animais , Southern Blotting , Cartilagem/irrigação sanguínea , Colágeno/biossíntese , Regulação para Baixo , Fatores de Crescimento Endotelial/metabolismo , Olho/irrigação sanguínea , Fator 2 de Crescimento de Fibroblastos/metabolismo , Substâncias de Crescimento/genética , Imuno-Histoquímica , Linfocinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Genéticos , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timo/embriologia , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta2 , Fator de Crescimento Transformador beta3 , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Sci Rep ; 7(1): 6149, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28733637

RESUMO

Farber disease (FD) is a rare autosomal recessive disease caused by mutations in the acid ceramidase gene (ASAH1). Low ceramidase activity results in the accumulation of fatty substances, mainly ceramides. Hallmark symptoms at clinical level are periarticular nodules, lipogranulomas, swollen and painful joints and a hoarse voice. FD phenotypes are heterogeneous varying from mild to very severe cases, with the patients not surviving past their first year of life. The diagnostic aspects of FD are poorly developed due to the rarity of the disease. In the present study, the screening for ceramides and related molecules was performed in Farber affected patients (n = 10), carriers (n = 11) and control individuals (n = 192). This study has the highest number of enrolled Farber patients and carriers reported to present. Liquid chromatography multiple reaction mass spectrometry (LC/MRM-MS) studies revealed that the ceramide C26:0 and especially its isoform 1 is a highly sensitive and specific biomarker for FD (p < 0.0001). The new biomarker can be determined directly in the dried blood spot extracts with low sample consumption. This allows for easy sample preparation, high reproducibility and use in high throughput screenings.


Assuntos
Biomarcadores/análise , Portador Sadio/diagnóstico , Ceramidas/análise , Lipogranulomatose de Farber/diagnóstico , Ceramidase Ácida/genética , Adulto , Pré-Escolar , Cromatografia Líquida , Teste em Amostras de Sangue Seco , Lipogranulomatose de Farber/genética , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Mutação , Adulto Jovem
20.
Eur J Hum Genet ; 25(2): 176-182, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27848944

RESUMO

We report our results of 1000 diagnostic WES cases based on 2819 sequenced samples from 54 countries with a wide phenotypic spectrum. Clinical information given by the requesting physicians was translated to HPO terms. WES processes were performed according to standardized settings. We identified the underlying pathogenic or likely pathogenic variants in 307 families (30.7%). In further 253 families (25.3%) a variant of unknown significance, possibly explaining the clinical symptoms of the index patient was identified. WES enabled timely diagnosing of genetic diseases, validation of causality of specific genetic disorders of PTPN23, KCTD3, SCN3A, PPOX, FRMPD4, and SCN1B, and setting dual diagnoses by detecting two causative variants in distinct genes in the same patient. We observed a better diagnostic yield in consanguineous families, in severe and in syndromic phenotypes. Our results suggest that WES has a better yield in patients that present with several symptoms, rather than an isolated abnormality. We also validate the clinical benefit of WES as an effective diagnostic tool, particularly in nonspecific or heterogeneous phenotypes. We recommend WES as a first-line diagnostic in all cases without a clear differential diagnosis, to facilitate personal medical care.


Assuntos
Exoma , Testes Genéticos/métodos , Técnicas de Genotipagem/métodos , Análise de Sequência de DNA/métodos , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Flavoproteínas/genética , Testes Genéticos/normas , Técnicas de Genotipagem/normas , Humanos , Lactente , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais/genética , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Núcleo Familiar , Fenótipo , Canais de Potássio/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Protoporfirinogênio Oxidase/genética , Análise de Sequência de DNA/normas , Canais de Sódio/genética , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa