Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Genet Med ; 25(11): 100950, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37551667

RESUMO

PURPOSE: Coffin-Siris and Nicolaides-Baraitser syndromes are recognizable neurodevelopmental disorders caused by germline variants in BAF complex subunits. The SMARCC2 BAFopathy was recently reported. Herein, we present clinical and molecular data on a large cohort. METHODS: Clinical symptoms for 41 novel and 24 previously published affected individuals were analyzed using the Human Phenotype Ontology. For genotype-phenotype correlations, molecular data were standardized and grouped into non-truncating and likely gene-disrupting (LGD) variants. Missense variant protein expression and BAF-subunit interactions were examined using 3D protein modeling, co-immunoprecipitation, and proximity-ligation assays. RESULTS: Neurodevelopmental delay with intellectual disability, muscular hypotonia, and behavioral disorders were the major manifestations. Clinical hallmarks of BAFopathies were rare. Clinical presentation differed significantly, with LGD variants being predominantly inherited and associated with mildly reduced or normal cognitive development, whereas non-truncating variants were mostly de novo and presented with severe developmental delay. These distinct manifestations and non-truncating variant clustering in functional domains suggest different pathomechanisms. In vitro testing showed decreased protein expression for N-terminal missense variants similar to LGD. CONCLUSION: This study improved SMARCC2 variant classification and identified discernible SMARCC2-associated phenotypes for LGD and non-truncating variants, which were distinct from other BAFopathies. The pathomechanism of most non-truncating variants has yet to be investigated.


Assuntos
Anormalidades Múltiplas , Deficiência Intelectual , Micrognatismo , Transtornos do Neurodesenvolvimento , Humanos , Anormalidades Múltiplas/genética , Face , Micrognatismo/genética , Deficiência Intelectual/genética , Deficiência Intelectual/complicações , Fácies , Fenótipo , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
2.
J Clin Immunol ; 42(8): 1766-1777, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35976468

RESUMO

Haploinsufficiency of suppressor of cytokine signaling 1 (SOCS1) is a recently discovered autoinflammatory disorder with significant rheumatologic, immunologic, and hematologic manifestations. Here we report a case of SOCS1 haploinsufficiency in a 5-year-old child with profound arthralgias and immune-mediated thrombocytopenia unmasked by SARS-CoV-2 infection. Her clinical manifestations were accompanied by excessive B cell activity, eosinophilia, and elevated IgE levels. Uniquely, this is the first report of SOCS1 haploinsufficiency in the setting of a chromosomal deletion resulting in complete loss of a single SOCS1 gene with additional clinical findings of bone marrow hypocellularity and radiologic evidence of severe enthesitis. Immunologic profiling showed a prominent interferon signature in the patient's peripheral blood mononuclear cells, which were also hypersensitive to stimulation by type I and type II interferons. The patient showed excellent clinical and functional laboratory response to tofacitinib, a Janus kinase inhibitor that disrupts interferon signaling. Our case highlights the need to utilize a multidisciplinary diagnostic approach and consider a comprehensive genetic evaluation for inborn errors of immunity in patients with an atypical immune-mediated thrombocytopenia phenotype.


Assuntos
COVID-19 , Síndromes Mielodisplásicas , Trombocitopenia , Feminino , Humanos , Proteína 1 Supressora da Sinalização de Citocina/genética , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Haploinsuficiência , Leucócitos Mononucleares/metabolismo , Medula Óssea , SARS-CoV-2 , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Interferons/metabolismo
3.
Am J Med Genet A ; 188(7): 2209-2216, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35365979

RESUMO

Multilocus imprinting disturbances (MLID) have been associated with up to 12% of patients with Beckwith-Wiedemann syndrome, Silver-Russell syndrome, and pseudohypoparathyroidism type 1B (PHP1B). Single-gene defects affecting components of the subcortical maternal complex (SCMC) have been reported in cases with multilocus hypomethylation defects. We present a patient with speech and language impairment with mild Angelman syndrome (AS) features who demonstrates maternal hypomethylation at 15q11.2 (SNRPN) as well as 11p15.5 (KCNQ1OT1) imprinted loci, but normal methylation at 6q24.2 (PLAGL1), 7p12.1 (GRB10), 7q32.2 (MEST), 11p15.5 (H19), 14q32.2 (MEG3), 19q13.43 (PEG3), and 20q13.32 (GNAS and GNAS-AS1). The proband also has no copy number nor sequence variants within the AS imprinting center or in UBE3A. Maternal targeted next generation sequencing did not identify any pathogenic variants in ZPF57, NLRP2, NLRP5, NLRP7, KHDC3L, PADI6, TLE6, OOEP, UHRF1 or ZAR1. The presence of very delayed, yet functional speech, behavioral difficulties, EEG abnormalities but without clinical seizures, and normocephaly are consistent with the 15q11.2 hypomethylation defect observed in this patient. To our knowledge, this is the first report of MLID in a patient with mild, likely mosaic, Angelman syndrome.


Assuntos
Síndrome de Angelman , Síndrome de Beckwith-Wiedemann , Deficiência Intelectual , Transtornos do Desenvolvimento da Linguagem , Proteínas Adaptadoras de Transdução de Sinal/genética , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Proteínas Estimuladoras de Ligação a CCAAT , Criança , Metilação de DNA , Impressão Genômica , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Transtornos do Desenvolvimento da Linguagem/genética , Ubiquitina-Proteína Ligases
4.
J Hum Genet ; 66(4): 439-443, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33067531

RESUMO

Kagami-Ogata syndrome (KOS14) is a rare imprinting disorder characterized by a unique constellation of phenotypes including bell-shaped small thorax with coat-hanger appearance of the ribs. We encountered an African American female infant with KOS14 phenotype and 46,XX,t(2;14)(q11.2;q32.2)mat. After excluding upd(14)pat and an epimutation (hypermethylation) and a deletion affecting the maternally derived 14q32.2 imprinted region, we performed whole-genome sequencing, revealing that the translocation was generated between noncoding region at 2q11.2 and intron 6 of MEG3 at 14q32.2. Subsequent Sanger sequencing for the fusion points showed that the chromosomal fusion on the der(2) chromosome occurred between Chr2:102,193,994 (bp) and Chr14:101,314,628 (bp) in association with an insertion of 5-bp segment of unknown origin and that on the der(14) chromosome took place between Chr14:101,314,627 (bp) and Chr2:102,193,995 (bp) in association with an insertion of 1-bp segment of unknown origin (according to GRCh37/hg19). The results, together with the previous data in patients with KOS14, imply that the MEG3 disruption by 46,XX,t(2;14)(q11.2;q32.2)mat caused silencing of all MEGs including RTL1as and resultant excessive RTL1 expression, leading to the development of KOS14. To our knowledge, while Robertsonian translocations involving chromosome 14 have been reported in KOS14, this is the first case of KOS14 caused by a chromosomal translocation involving the 14q32.2 imprinted region.


Assuntos
Transtornos Cromossômicos/patologia , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 2/genética , RNA Longo não Codificante/genética , Translocação Genética , Dissomia Uniparental/patologia , Adolescente , Transtornos Cromossômicos/genética , Feminino , Humanos , Recém-Nascido , Síndrome , Dissomia Uniparental/genética
5.
J Pediatr Hematol Oncol ; 43(3): e336-e340, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33122585

RESUMO

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure syndrome usually caused by heterozygous variants in ribosomal proteins (RP) and which leads to severe anemia. Genetic studies in DBA rely primarily on multigene panels that often result in variants of unknown significance. Our objective was to optimize polysome profiling to functionally validate new large subunit RP variants. We determined the optimal experimental conditions for B-cell polysome profiles then performed this analysis on 2 children with DBA and novel missense RPL5 (uL18) and RPL26 (uL24) variants of unknown significance. Both patients had reduced 60S and 80S fractions when compared with an unaffected parent consistent with a large ribosomal subunit defect. Polysome profiling using primary B-cells is an adjunctive tool that can assist in validation of large subunit RP variants of uncertain significance. Further studies are necessary to validate this method in patients with known DBA mutations, small RP subunit variants, and silent carriers.


Assuntos
Anemia de Diamond-Blackfan/genética , Polirribossomos/genética , Proteínas Ribossômicas/genética , Linfócitos B/metabolismo , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto
6.
Am J Hum Genet ; 100(4): 650-658, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28343630

RESUMO

Intellectual disability (ID) is a highly heterogeneous disorder involving at least 600 genes, yet a genetic diagnosis remains elusive in ∼35%-40% of individuals with moderate to severe ID. Recent meta-analyses statistically analyzing de novo mutations in >7,000 individuals with neurodevelopmental disorders highlighted mutations in PPM1D as a possible cause of ID. PPM1D is a type 2C phosphatase that functions as a negative regulator of cellular stress-response pathways by mediating a feedback loop of p38-p53 signaling, thereby contributing to growth inhibition and suppression of stress-induced apoptosis. We identified 14 individuals with mild to severe ID and/or developmental delay and de novo truncating PPM1D mutations. Additionally, deep phenotyping revealed overlapping behavioral problems (ASD, ADHD, and anxiety disorders), hypotonia, broad-based gait, facial dysmorphisms, and periods of fever and vomiting. PPM1D is expressed during fetal brain development and in the adult brain. All mutations were located in the last or penultimate exon, suggesting escape from nonsense-mediated mRNA decay. Both PPM1D expression analysis and cDNA sequencing in EBV LCLs of individuals support the presence of a stable truncated transcript, consistent with this hypothesis. Exposure of cells derived from individuals with PPM1D truncating mutations to ionizing radiation resulted in normal p53 activation, suggesting that p53 signaling is unaffected. However, a cell-growth disadvantage was observed, suggesting a possible effect on the stress-response pathway. Thus, we show that de novo truncating PPM1D mutations in the last and penultimate exons cause syndromic ID, which provides additional insight into the role of cell-cycle checkpoint genes in neurodevelopmental disorders.


Assuntos
Éxons , Deficiência Intelectual/genética , Mutação , Proteína Fosfatase 2C/genética , Adolescente , Ciclo Celular , Criança , Pré-Escolar , Humanos , Deficiência Intelectual/patologia , Adulto Jovem
7.
J Clin Immunol ; 40(2): 267-276, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31853824

RESUMO

We report three new cases of a germline heterozygous gain-of-function missense (p.(Met1141Lys)) mutation in the C2 domain of phospholipase C gamma 2 (PLCG2) associated with symptoms consistent with previously described auto-inflammation and phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) syndrome and pediatric common variable immunodeficiency (CVID). Functional evaluation showed platelet hyper-reactivity, increased B cell receptor-triggered calcium influx and ERK phosphorylation. Expression of the altered p.(Met1141Lys) variant in a PLCγ2-knockout DT40 cell line showed clearly enhanced BCR-triggered influx of external calcium when compared to control-transfected cells. Our results further expand the molecular basis of pediatric CVID and phenotypic spectrum of PLCγ2-related defects.


Assuntos
Linfócitos B/imunologia , Imunodeficiência de Variável Comum/diagnóstico , Mutação em Linhagem Germinativa/genética , Síndromes de Imunodeficiência/diagnóstico , Mutação de Sentido Incorreto/genética , Fosfolipase C gama/genética , Autoimunidade/genética , Sinalização do Cálcio , Linhagem Celular , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Fenótipo , Domínios Proteicos/genética
8.
Ann Neurol ; 86(6): 899-912, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31600826

RESUMO

OBJECTIVE: Pathogenic variants in KCNB1, encoding the voltage-gated potassium channel KV 2.1, are associated with developmental and epileptic encephalopathy (DEE). Previous functional studies on a limited number of KCNB1 variants indicated a range of molecular mechanisms by which variants affect channel function, including loss of voltage sensitivity, loss of ion selectivity, and reduced cell-surface expression. METHODS: We evaluated a series of 17 KCNB1 variants associated with DEE or other neurodevelopmental disorders (NDDs) to rapidly ascertain channel dysfunction using high-throughput functional assays. Specifically, we investigated the biophysical properties and cell-surface expression of variant KV 2.1 channels expressed in heterologous cells using high-throughput automated electrophysiology and immunocytochemistry-flow cytometry. RESULTS: Pathogenic variants exhibited diverse functional defects, including altered current density and shifts in the voltage dependence of activation and/or inactivation, as homotetramers or when coexpressed with wild-type KV 2.1. Quantification of protein expression also identified variants with reduced total KV 2.1 expression or deficient cell-surface expression. INTERPRETATION: Our study establishes a platform for rapid screening of KV 2.1 functional defects caused by KCNB1 variants associated with DEE and other NDDs. This will aid in establishing KCNB1 variant pathogenicity and the mechanism of dysfunction, which will enable targeted strategies for therapeutic intervention based on molecular phenotype. ANN NEUROL 2019;86:899-912.


Assuntos
Variação Genética/genética , Ensaios de Triagem em Larga Escala/métodos , Transtornos do Neurodesenvolvimento/genética , Canais de Potássio Shab/genética , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Transtornos do Neurodesenvolvimento/diagnóstico , Estrutura Secundária de Proteína , Canais de Potássio Shab/química
9.
Pediatr Res ; 87(4): 735-739, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31618753

RESUMO

BACKGROUND: As clinical exome sequencing (CES) becomes more common, understanding which patients are most likely to benefit and in what manner is critical for the general pediatrics community to appreciate. METHODS: Five hundred and twenty-three patients referred to the Pediatric Genetics clinic at Michigan Medicine were systematically phenotyped by the presence or absence of abnormalities for 13 body/organ systems by a Clinical Genetics team. All patients then underwent CES. RESULTS: Overall, 30% of patients who underwent CES had an identified pathogenic mutation. The most common phenotypes were developmental delay (83%), neuromuscular system abnormalities (81%), and multiple congenital anomalies (42%). In all, 67% of patients had a variant of uncertain significance (VUS) or gene of uncertain significance (GUS); 23% had no variants reported. There was a significant difference in the average number of body systems affected among these groups (pathogenic 5.89, VUS 6.0, GUS 6.12, and no variant 4.6; P < 0.00001). Representative cases highlight four ways in which CES is changing clinical pediatric practice. CONCLUSIONS: Patients with identified variants are enriched for multiple organ system involvement. Furthermore, our phenotyping provides broad insights into which patients are most likely to benefit from genetics referral and CES and how those results can help guide clinical practice more generally.


Assuntos
Anormalidades Congênitas/genética , Análise Mutacional de DNA , Sequenciamento do Exoma , Testes Genéticos , Mutação , Anormalidades Congênitas/diagnóstico , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Fenótipo , Valor Preditivo dos Testes , Estudos Retrospectivos
10.
Genet Med ; 21(9): 2007-2014, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30760892

RESUMO

PURPOSE: EPHB4 variants were recently reported to cause capillary malformation-arteriovenous malformation 2 (CM-AVM2). CM-AVM2 mimics RASA1-related CM-AVM1 and hereditary hemorrhagic telangiectasia (HHT), as clinical features include capillary malformations (CMs), telangiectasia, and arteriovenous malformations (AVMs). Epistaxis, another clinical feature that overlaps with HHT, was reported in several cases. Based on the clinical overlap of CM-AVM2 and HHT, we hypothesized that patients considered clinically suspicious for HHT with no variant detected in an HHT gene (ENG, ACVRL1, or SMAD4) may have an EPHB4 variant. METHODS: Exome sequencing or a next-generation sequencing panel including EPHB4 was performed on individuals with previously negative molecular genetic testing for the HHT genes and/or RASA1. RESULTS: An EPHB4 variant was identified in ten unrelated cases. Seven cases had a pathogenic EPHB4 variant, including one with mosaicism. Three cases had an EPHB4 variant of uncertain significance. The majority had epistaxis (6/10 cases) and telangiectasia (8/10 cases), as well as CMs. Two of ten cases had a central nervous system AVM. CONCLUSIONS: Our results emphasize the importance of considering CM-AVM2 as part of the clinical differential for HHT and other vascular malformation syndromes. Yet, these cases highlight significant differences in the cutaneous presentations of CM-AVM2 versus HHT.


Assuntos
Capilares/anormalidades , Testes Genéticos , Receptor EphB4/genética , Telangiectasia Hemorrágica Hereditária/genética , Malformações Vasculares/genética , Receptores de Activinas Tipo II/genética , Adolescente , Capilares/patologia , Criança , Endoglina/genética , Feminino , Humanos , Masculino , Mutação , Proteína Smad4/genética , Telangiectasia Hemorrágica Hereditária/diagnóstico , Telangiectasia Hemorrágica Hereditária/patologia , Malformações Vasculares/patologia , Sequenciamento do Exoma
12.
Hum Mutat ; 39(1): 103-113, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29024177

RESUMO

Loss-of-function mutations in the X-linked gene FLNA can lead to abnormal neuronal migration, vascular and cardiac defects, and congenital intestinal pseudo-obstruction (CIPO), the latter characterized by anomalous intestinal smooth muscle layering. Survival in male hemizygotes for such mutations is dependent on retention of residual FLNA function but it is unclear why a subgroup of males with mutations in the 5' end of the gene can present with CIPO alone. Here, we demonstrate evidence for the presence of two FLNA isoforms differing by 28 residues at the N-terminus initiated at ATG+1 and ATG+82 . A male with CIPO (c.18_19del) exclusively expressed FLNA ATG+82 , implicating the longer protein isoform (ATG+1 ) in smooth muscle development. In contrast, mutations leading to reduction of both isoforms are associated with compound phenotypes affecting the brain, heart, and intestine. RNA-seq data revealed three distinct transcription start sites, two of which produce a protein isoform utilizing ATG+1 while the third utilizes ATG+82 . Transcripts sponsoring translational initiation at ATG+1 predominate in intestinal smooth muscle, and are more abundant compared with the level measured in fibroblasts. Together these observations describe a new mechanism of tissue-specific regulation of FLNA that could reflect the differing mechanical requirements of these cell types during development.


Assuntos
Filaminas/genética , Estudos de Associação Genética , Heterogeneidade Genética , Mutação com Perda de Função , Fenótipo , Transcrição Gênica , Adolescente , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Criança , Sequência Conservada , Análise Mutacional de DNA , Feminino , Filaminas/química , Filaminas/metabolismo , Trato Gastrointestinal/metabolismo , Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Masculino , Músculo Liso/metabolismo , Isoformas de Proteínas , Adulto Jovem
13.
Hum Mol Genet ; 25(3): 597-608, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26647312

RESUMO

De novo truncating mutations in Additional sex combs-like 3 (ASXL3) have been identified in individuals with Bainbridge-Ropers syndrome (BRS), characterized by failure to thrive, global developmental delay, feeding problems, hypotonia, dysmorphic features, profound speech delays and intellectual disability. We identified three novel de novo heterozygous truncating variants distributed across ASXL3, outside the original cluster of ASXL3 mutations previously described for BRS. Primary skin fibroblasts established from a BRS patient were used to investigate the functional impact of pathogenic variants. ASXL3 mRNA transcripts from the mutated allele are prone to nonsense-mediated decay, and expression of ASXL3 is reduced. We found that ASXL3 interacts with BAP1, a hydrolase that removes mono-ubiquitin from histone H2A lysine 119 (H2AK119Ub1) as a component of the Polycomb repressive deubiquitination (PR-DUB) complex. A significant increase in H2AK119Ub1 was observed in ASXL3 patient fibroblasts, highlighting an important functional role for ASXL3 in PR-DUB mediated deubiquitination. Transcriptomes of ASXL3 patient and control fibroblasts were compared to investigate the impact of chromatin changes on transcriptional regulation. Out of 564 significantly differentially expressed genes (DEGs) in ASXL3 patient fibroblasts, 52% were upregulated and 48% downregulated. DEGs were enriched in molecular processes impacting transcriptional regulation, development and proliferation, consistent with the features of BRS. This is the first single gene disorder linked to defects in deubiquitination of H2AK119Ub1 and suggests an important role for dynamic regulation of H2A mono-ubiquitination in transcriptional regulation and the pathophysiology of BRS.


Assuntos
Deficiências do Desenvolvimento/genética , Insuficiência de Crescimento/genética , Histonas/metabolismo , Deficiência Intelectual/genética , Transtornos do Desenvolvimento da Linguagem/genética , Mutação , Fatores de Transcrição/metabolismo , Pré-Escolar , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Insuficiência de Crescimento/metabolismo , Insuficiência de Crescimento/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Genes Dominantes , Heterozigoto , Histonas/genética , Humanos , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Transtornos do Desenvolvimento da Linguagem/metabolismo , Transtornos do Desenvolvimento da Linguagem/patologia , Masculino , Cultura Primária de Células , Ligação Proteica , Síndrome , Fatores de Transcrição/genética , Transcriptoma , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação
14.
Neurogenetics ; 17(3): 173-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27094857

RESUMO

Exome sequencing is an effective way to identify genetic causes of etiologically heterogeneous conditions such as developmental delay and intellectual disabilities. Using exome sequencing, we have identified four patients with similar phenotypes of developmental delay, intellectual disability, failure to thrive, hypotonia, ataxia, and tooth enamel defects who all have the same de novo R331W missense variant in C-terminal binding protein 1 (CTBP1). CTBP1 is a transcriptional regulator critical for development by coordinating different regulatory pathways. The R331W variant found in these patients is within the C-terminal portion of the PLDLS (Pro-Leu-Asp-Leu-Ser) binding cleft, which is the domain through which CTBP1, interacts with chromatin-modifying enzymes and mediates chromatin-dependent gene repression pathways. This is the first report of mutations within CTBP1 in association with any human disease.


Assuntos
Oxirredutases do Álcool/genética , Ataxia/genética , Proteínas de Ligação a DNA/genética , Esmalte Dentário/patologia , Deficiências do Desenvolvimento/genética , Hipotonia Muscular/genética , Mutação de Sentido Incorreto , Adulto , Ataxia/complicações , Criança , Deficiências do Desenvolvimento/complicações , Feminino , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Masculino , Hipotonia Muscular/complicações , Sequenciamento do Exoma , Adulto Jovem
15.
Hum Mol Genet ; 22(1): 1-17, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22949511

RESUMO

Although biallelic mutations in non-collagen genes account for <10% of individuals with osteogenesis imperfecta, the characterization of these genes has identified new pathways and potential interventions that could benefit even those with mutations in type I collagen genes. We identified mutations in FKBP10, which encodes the 65 kDa prolyl cis-trans isomerase, FKBP65, in 38 members of 21 families with OI. These include 10 families from the Samoan Islands who share a founder mutation. Of the mutations, three are missense; the remainder either introduce premature termination codons or create frameshifts both of which result in mRNA instability. In four families missense mutations result in loss of most of the protein. The clinical effects of these mutations are short stature, a high incidence of joint contractures at birth and progressive scoliosis and fractures, but there is remarkable variability in phenotype even within families. The loss of the activity of FKBP65 has several effects: type I procollagen secretion is slightly delayed, the stabilization of the intact trimer is incomplete and there is diminished hydroxylation of the telopeptide lysyl residues involved in intermolecular cross-link formation in bone. The phenotype overlaps with that seen with mutations in PLOD2 (Bruck syndrome II), which encodes LH2, the enzyme that hydroxylates the telopeptide lysyl residues. These findings define a set of genes, FKBP10, PLOD2 and SERPINH1, that act during procollagen maturation to contribute to molecular stability and post-translational modification of type I procollagen, without which bone mass and quality are abnormal and fractures and contractures result.


Assuntos
Artrogripose/genética , Colágeno Tipo I/metabolismo , Genes Recessivos , Lisina/metabolismo , Mutação , Osteogênese Imperfeita/genética , Proteínas de Ligação a Tacrolimo/genética , Feminino , Humanos , Hidroxilação , Masculino , Processamento de Proteína Pós-Traducional
16.
Am J Med Genet A ; 167A(12): 3038-45, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26420380

RESUMO

The ability to identify the clinical nature of the recurrent duplication of chromosome 17q12 has been limited by its rarity and the diverse range of phenotypes associated with this genomic change. In order to further define the clinical features of affected patients, detailed clinical information was collected in the largest series to date (30 patients and 2 of their siblings) through a multi-institutional collaborative effort. The majority of patients presented with developmental delays varying from mild to severe. Though dysmorphic features were commonly reported, patients do not have consistent and recognizable features. Cardiac, ophthalmologic, growth, behavioral, and other abnormalities were each present in a subset of patients. The newly associated features potentially resulting from 17q12 duplication include height and weight above the 95th percentile, cataracts, microphthalmia, coloboma, astigmatism, tracheomalacia, cutaneous mosaicism, pectus excavatum, scoliosis, hypermobility, hypospadias, diverticulum of Kommerell, pyloric stenosis, and pseudohypoparathryoidism. The majority of duplications were inherited with some carrier parents reporting learning disabilities or microcephaly. We identified additional, potentially contributory copy number changes in a subset of patients, including one patient each with 16p11.2 deletion and 15q13.3 deletion. Our data further define and expand the clinical spectrum associated with duplications of 17q12 and provide support for the role of genomic modifiers contributing to phenotypic variability.


Assuntos
Anormalidades Múltiplas/genética , Duplicação Cromossômica , Adolescente , Criança , Pré-Escolar , Variações do Número de Cópias de DNA , Deficiências do Desenvolvimento/genética , Face/anormalidades , Feminino , Humanos , Lactente , Masculino , Microcefalia/genética , Fenótipo , Adulto Jovem
17.
Nat Genet ; 37(10): 1044-6, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186812

RESUMO

Hereditary neuralgic amyotrophy (HNA) is an autosomal dominant recurrent neuropathy affecting the brachial plexus. HNA is triggered by environmental factors such as infection or parturition. We report three mutations in the gene septin 9 (SEPT9) in six families with HNA linked to chromosome 17q25. HNA is the first monogenetic disease caused by mutations in a gene of the septin family. Septins are implicated in formation of the cytoskeleton, cell division and tumorigenesis.


Assuntos
Neurite do Plexo Braquial/genética , Cromossomos Humanos Par 17/genética , GTP Fosfo-Hidrolases/genética , Mutação , Sequência de Aminoácidos , Animais , Sequência de Bases , Cães , Humanos , Camundongos , Dados de Sequência Molecular , Ratos , Septinas
18.
Hum Mutat ; 34(10): 1415-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23878096

RESUMO

We describe the molecular and clinical characterization of nine individuals with recurrent, 3.4-Mb, de novo deletions of 3q13.2-q13.31 detected by chromosomal microarray analysis. All individuals have hypotonia and language and motor delays; they variably express mild to moderate cognitive delays (8/9), abnormal behavior (7/9), and autism spectrum disorders (3/9). Common facial features include downslanting palpebral fissures with epicanthal folds, a slightly bulbous nose, and relative macrocephaly. Twenty-eight genes map to the deleted region, including four strong candidate genes, DRD3, ZBTB20, GAP43, and BOC, with important roles in neural and/or muscular development. Analysis of the breakpoint regions based on array data revealed directly oriented human endogenous retrovirus (HERV-H) elements of ~5 kb in size and of >95% DNA sequence identity flanking the deletion. Subsequent DNA sequencing revealed different deletion breakpoints and suggested nonallelic homologous recombination (NAHR) between HERV-H elements as a mechanism of deletion formation, analogous to HERV-I-flanked and NAHR-mediated AZFa deletions. We propose that similar HERV elements may also mediate other recurrent deletion and duplication events on a genome-wide scale. Observation of rare recurrent chromosomal events such as these deletions helps to further the understanding of mechanisms behind naturally occurring variation in the human genome and its contribution to genetic disease.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 3/genética , Transtornos Cognitivos/genética , Deficiências do Desenvolvimento/genética , Retrovirus Endógenos/genética , Hipotonia Muscular/genética , Adolescente , Adulto , Sequência de Bases , Criança , Pré-Escolar , Pontos de Quebra do Cromossomo , Transtornos Cognitivos/diagnóstico , Hibridização Genômica Comparativa , Deficiências do Desenvolvimento/diagnóstico , Fácies , Feminino , Ordem dos Genes , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Hipotonia Muscular/diagnóstico , Fenótipo , Alinhamento de Sequência , Síndrome , Adulto Jovem
20.
Am J Med Genet A ; 161A(4): 717-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23495017

RESUMO

Deletions at 2p16.3 involving exons of NRXN1 are associated with susceptibility for autism and schizophrenia, and similar deletions have been identified in individuals with developmental delay and dysmorphic features. We have identified 34 probands with exonic NRXN1 deletions following referral for clinical microarray-based comparative genomic hybridization. To more firmly establish the full phenotypic spectrum associated with exonic NRXN1 deletions, we report the clinical features of 27 individuals with NRXN1 deletions, who represent 23 of these 34 families. The frequency of exonic NRXN1 deletions among our postnatally diagnosed patients (0.11%) is significantly higher than the frequency among reported controls (0.02%; P = 6.08 × 10(-7) ), supporting a role for these deletions in the development of abnormal phenotypes. Generally, most individuals with NRXN1 exonic deletions have developmental delay (particularly speech), abnormal behaviors, and mild dysmorphic features. In our cohort, autism spectrum disorders were diagnosed in 43% (10/23), and 16% (4/25) had epilepsy. The presence of NRXN1 deletions in normal parents and siblings suggests reduced penetrance and/or variable expressivity, which may be influenced by genetic, environmental, and/or stochastic factors. The pathogenicity of these deletions may also be affected by the location of the deletion within the gene. Counseling should appropriately represent this spectrum of possibilities when discussing recurrence risks or expectations for a child found to have a deletion in NRXN1.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Deleção de Genes , Proteínas do Tecido Nervoso/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Adolescente , Adulto , Transtorno Autístico/genética , Proteínas de Ligação ao Cálcio , Criança , Pré-Escolar , Hibridização Genômica Comparativa , Deficiências do Desenvolvimento/genética , Éxons , Fácies , Feminino , Interação Gene-Ambiente , Estudo de Associação Genômica Ampla , Humanos , Lactente , Deficiência Intelectual/genética , Masculino , Pessoa de Meia-Idade , Moléculas de Adesão de Célula Nervosa , Penetrância , Fenótipo , Esquizofrenia/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA