Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 28(2): 290-306, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30304514

RESUMO

LonP1 is crucial for maintaining mitochondrial proteostasis and mitigating cell stress. We identified a novel homozygous missense LONP1 variant, c.2282 C > T, (p.Pro761Leu), by whole-exome and Sanger sequencing in two siblings born to healthy consanguineous parents. Both siblings presented with stepwise regression during infancy, profound hypotonia and muscle weakness, severe intellectual disability and progressive cerebellar atrophy on brain imaging. Muscle biopsy revealed the absence of ragged-red fibers, however, scattered cytochrome c oxidase-negative staining and electron dense mitochondrial inclusions were observed. Primary cultured fibroblasts from the siblings showed normal levels of mtDNA and mitochondrial transcripts, and normal activities of oxidative phosphorylation complexes I through V. Interestingly, fibroblasts of both siblings showed glucose-repressed oxygen consumption compared to their mother, whereas galactose and palmitic acid utilization were similar. Notably, the siblings' fibroblasts had reduced pyruvate dehydrogenase (PDH) activity and elevated intracellular lactate:pyruvate ratios, whereas plasma ratios were normal. We demonstrated that in the siblings' fibroblasts, PDH dysfunction was caused by increased levels of the phosphorylated E1α subunit of PDH, which inhibits enzyme activity. Blocking E1α phosphorylation activated PDH and reduced intracellular lactate concentrations. In addition, overexpressing wild-type LonP1 in the siblings' fibroblasts down-regulated phosphoE1α. Furthermore, in vitro studies demonstrated that purified LonP1-P761L failed to degrade phosphorylated E1α, in contrast to wild-type LonP1. We propose a novel mechanism whereby homozygous expression of the LonP1-P761L variant leads to PDH deficiency and energy metabolism dysfunction, which promotes severe neurologic impairment and neurodegeneration.


Assuntos
Proteases Dependentes de ATP/genética , Doenças Cerebelares/genética , Proteínas Mitocondriais/genética , Mutação , Doenças Neurodegenerativas/genética , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Alelos , Doenças Cerebelares/enzimologia , DNA Mitocondrial/metabolismo , Homozigoto , Humanos , Recém-Nascido , Lactatos/metabolismo , Masculino , Doenças Neurodegenerativas/enzimologia , Linhagem , Fosforilação , Subunidades Proteicas/metabolismo , Proteólise , Doença da Deficiência do Complexo de Piruvato Desidrogenase/patologia
2.
Genet Med ; 20(4): 435-443, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28771251

RESUMO

PurposeGenetic testing is an integral diagnostic component of pediatric medicine. Standard of care is often a time-consuming stepwise approach involving chromosomal microarray analysis and targeted gene sequencing panels, which can be costly and inconclusive. Whole-genome sequencing (WGS) provides a comprehensive testing platform that has the potential to streamline genetic assessments, but there are limited comparative data to guide its clinical use.MethodsWe prospectively recruited 103 patients from pediatric non-genetic subspecialty clinics, each with a clinical phenotype suggestive of an underlying genetic disorder, and compared the diagnostic yield and coverage of WGS with those of conventional genetic testing.ResultsWGS identified diagnostic variants in 41% of individuals, representing a significant increase over conventional testing results (24%; P = 0.01). Genes clinically sequenced in the cohort (n = 1,226) were well covered by WGS, with a median exonic coverage of 40 × ±8 × (mean ±SD). All the molecular diagnoses made by conventional methods were captured by WGS. The 18 new diagnoses made with WGS included structural and non-exonic sequence variants not detectable with whole-exome sequencing, and confirmed recent disease associations with the genes PIGG, RNU4ATAC, TRIO, and UNC13A.ConclusionWGS as a primary clinical test provided a higher diagnostic yield than conventional genetic testing in a clinically heterogeneous cohort.


Assuntos
Estudos de Associação Genética , Doenças Genéticas Inatas/diagnóstico , Doenças Genéticas Inatas/genética , Predisposição Genética para Doença , Testes Genéticos , Análise de Sequência de DNA , Sequenciamento Completo do Genoma , Biologia Computacional/métodos , Variações do Número de Cópias de DNA , Exoma , Feminino , Estudos de Associação Genética/métodos , Estudos de Associação Genética/normas , Testes Genéticos/métodos , Testes Genéticos/normas , Variação Genética , Humanos , Masculino , Anotação de Sequência Molecular , Fenótipo , Análise de Sequência de DNA/métodos , Análise de Sequência de DNA/normas , Sequenciamento do Exoma/métodos , Sequenciamento do Exoma/normas , Sequenciamento Completo do Genoma/métodos , Sequenciamento Completo do Genoma/normas
3.
Hum Mutat ; 37(7): 703-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27071356

RESUMO

Biallelic loss-of-function mutations in SPG11 cause a wide spectrum of recessively inherited, neurodegenerative disorders including hereditary spastic paraplegia (HSP), amyotrophic lateral sclerosis, and Charcot-Marie-Tooth disease. By comprehensive screening of three large cohorts of HSP index patients, we identified 83 alleles with "small" mutations and 13 alleles that carry large genomic rearrangements. Including relevant data from previous studies, we estimate that copy number variants (CNVs) account for ∼19% of pathogenic SPG11 alleles. The breakpoints for all novel and some previously reported CNVs were determined by long-range PCR and sequencing. This revealed several Alu-associated recombination hotspots. We also found evidence for additional mutational mechanisms, including for a two-step event in which an Alu retrotransposition preceded the actual rearrangement. Apparently independent samples with identical breakpoints were analyzed by microsatellite PCRs. The resulting haplotypes suggested the existence of two rearrangement founder alleles. Our findings widen the spectra of mutations and mutational mechanisms in SPG11, underscore the pivotal role played by Alus, and are of high diagnostic relevance for a wide spectrum of clinical phenotypes including the most frequent form of recessive HSP.


Assuntos
Variações do Número de Cópias de DNA , Proteínas/genética , Paraplegia Espástica Hereditária/genética , Alelos , Elementos Alu , Pontos de Quebra do Cromossomo , Cromossomos Humanos/genética , Efeito Fundador , Humanos , Mutação , Análise de Sequência de DNA
4.
BMC Genomics ; 16 Suppl 1: S12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25923536

RESUMO

BACKGROUND: We report a consanguineous couple that has experienced three consecutive pregnancy losses following the foetal ultrasound finding of short limbs. Post-termination examination revealed no skeletal dysplasia, but some subtle proximal limb shortening in two foetuses, and a spectrum of mildly dysmorphic features. Karyotype was normal in all three foetuses (46, XX) and comparative genomic hybridization microarray analysis detected no pathogenic copy number variants. RESULTS: Whole-exome sequencing and genome-wide homozygosity mapping revealed a previously reported frameshift mutation in the OBSL1 gene (c.1273insA p.T425nfsX40), consistent with a diagnosis of 3-M Syndrome 2 (OMIM #612921), which had not been anticipated from the clinical findings. CONCLUSIONS: Our study provides novel insight into the early clinical manifestations of this form of 3-M syndrome, and demonstrates the utility of whole exome sequencing as a tool for prenatal diagnosis in particular when there is a family history suggestive of a recurrent set of clinical symptoms.


Assuntos
Autopsia , Proteínas do Citoesqueleto/genética , Nanismo/diagnóstico , Nanismo/genética , Feto/metabolismo , Mutação da Fase de Leitura/genética , Hipotonia Muscular/diagnóstico , Hipotonia Muscular/genética , Coluna Vertebral/anormalidades , Análise Mutacional de DNA , Exoma , Feminino , Humanos , Masculino , Linhagem , Fenótipo , Gravidez
5.
Am J Med Genet A ; 167A(11): 2544-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26097063

RESUMO

Craniosynostosis is a clinically and genetically heterogeneous condition. Knowledge of the specific genetic diagnosis in patients presenting with this condition is important for surgical and medical management. The most common single gene causes of syndromic craniosynostosis are mutations in FGFR1, FGFR2, FGFR3, TWIST1, and EFNB1. Recently, a new single gene cause of craniosynostosis was published, together with phenotype data that highlight the clinical importance of making this specific molecular diagnosis. Phenotypic features of "ERF-related craniosynostosis" include sagittal or multiple-suture synostosis, Chiari malformation, and language delay. In order to determine the contribution of ERF mutations to genetically undiagnosed patients with craniosynostosis, we sequenced the coding regions of ERF in 40 patients with multi-suture or sagittal suture synostosis. We identified heterozygous ERF mutations in two individuals (5%). One mutation positive individual had pansynostosis, while the second had bilateral coronal and metopic synostosis. Both presented in infancy or childhood (age 3 months, and 6 years 9 months, respectively). One had CNS abnormalities including Chiari I malformation. Dysmorphic features included hypertelorism, proptosis, depressed nasal bridge, and retrognathia, in keeping with previously reported cases. The individuals did not require repeated cranial surgeries. ERF-related craniosynostosis should be suspected in patients presenting with multiple suture or sagittal synostosis.


Assuntos
Suturas Cranianas/patologia , Craniossinostoses/genética , Mutação/genética , Proteínas Repressoras/genética , Sequência de Bases , Criança , Pré-Escolar , Estudos de Coortes , Suturas Cranianas/diagnóstico por imagem , Craniossinostoses/diagnóstico por imagem , Heterozigoto , Humanos , Dados de Sequência Molecular , Fenótipo , Síndrome , Tomografia Computadorizada por Raios X
6.
Hum Mutat ; 35(5): 513-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24599881

RESUMO

Our increasing knowledge of how genomic variants affect human health and the falling costs of whole-genome sequencing are driving the development of individualized genetic medicine. This new clinical paradigm uses knowledge of an individual's genomic variants to guide health care decisions throughout life, to anticipate, diagnose, and manage disease. While individualized genetic medicine offers the promise of transformative change in health care, it forces us to reconsider existing ethical, scientific, and clinical paradigms. The potential benefits of presymptomatic identification of at risk individuals, improved diagnostics, individualized therapy, accurate prognosis, and avoidance of adverse drug reactions coexist with the potential risks of uninterpretable results, psychological harm, outmoded counseling models, and increased health care costs. Here, we review the challenges of integrating genomic analysis into clinical practice and describe a prototype for implementing genetic medicine. Our multidisciplinary team of bioinformaticians, health economists, ethicists, geneticists, genetic counselors, and clinicians has designed a "Genome Clinic" research project that addresses multiple challenges in genomic medicine-ranging from the development of bioinformatics tools for the clinical assessment of genomic variants and the discovery of disease genes to health policy inquiries, assessment of clinical care models, patient preference, and the ethics of consent.


Assuntos
Variação Genética , Genômica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Medicina de Precisão , Pesquisa Biomédica/economia , Biologia Computacional , Testes Genéticos , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala/economia , Humanos
7.
Hum Genet ; 133(3): 321-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24154661

RESUMO

Beckwith-Wiedemann syndrome (BWS), an overgrowth and tumor predisposition syndrome is clinically heterogeneous. Its variable presentation makes molecular diagnosis particularly important for appropriate counseling of patients with respect to embyronal tumor risk and recurrence risk. BWS is characterized by macrosomia, omphalocele, and macroglossia. Additional clinical features can include hemihyperplasia, embryonal tumors, umbilical hernia, and ear anomalies. BWS is etiologically heterogeneous arising from dysregulation of one or both of the chromosome 11p15.5 imprinting centers (IC) and/or imprinted growth regulatory genes on chromosome 11p15.5. Most BWS cases are sporadic and result from loss of maternal methylation at imprinting center 2 (IC2), gain of maternal methylation at imprinting center 1 (IC1) or paternal uniparental disomy (UPD). Heritable forms of BWS (15 %) have been attributed mainly to mutations in the growth suppressor gene CDKN1C, but have also infrequently been identified in patients with copy number variations (CNVs) in the chromosome 11p15.5 region. Four hundred and thirty-four unrelated BWS patients referred to the molecular diagnostic laboratory were tested by methylation-specific multiplex ligation-dependent probe amplification. Molecular alterations were detected in 167 patients, where 103 (62 %) showed loss of methylation at IC2, 23 (14 %) had gain of methylation at IC1, and 41 (25 %) showed changes at both ICs usually associated with paternal UPD. In each of the three groups, we identified patients in whom the abnormalities in the chromosome 11p15.5 region were due to CNVs. Surprisingly, 14 patients (9 %) demonstrated either deletions or duplications of the BWS critical region that were confirmed using comparative genomic hybridization array analysis. The majority of these CNVs were associated with a methylation change at IC1. Our results suggest that CNVs in the 11p15.5 region contribute significantly to the etiology of BWS. We highlight the importance of performing deletion/duplication testing in addition to methylation analysis in the molecular investigation of BWS to improve our understanding of the molecular basis of this disorder, and to provide accurate genetic counseling.


Assuntos
Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos Par 11/genética , Variações do Número de Cópias de DNA/genética , Deleção Cromossômica , Cromossomos Humanos Par 4/genética , Hibridização Genômica Comparativa , Inibidor de Quinase Dependente de Ciclina p57/genética , Metilação de DNA , Feminino , Rearranjo Gênico , Impressão Genômica , Genótipo , Humanos , Masculino , Linhagem , Fenótipo
8.
Thorax ; 69(3): 254-60, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24149827

RESUMO

BACKGROUND: The phenotypic spectrum of cystic fibrosis (CF) has expanded to include patients affected by single-organ diseases. Extensive genotyping and nasal potential difference (NPD) testing have been proposed to assist in the diagnosis of CF when sweat testing is inconclusive. However, the diagnostic yield of extensive genotyping and NPD and the concordance between NPD and the sweat test have not been carefully evaluated. METHODS: We evaluated the diagnostic outcomes of genotyping (with 122 mutations included as disease causing), sweat testing and NPD in a prospectively ascertained cohort of undiagnosed patients who presented with chronic sino-pulmonary disease (RESP), chronic/recurrent pancreatitis (PANC) or obstructive azoospermia (AZOOSP). RESULTS: 202 patients (68 RESP, 42 PANC and 92 AZOOSP) were evaluated; 17.3%, 22.8% and 59.9% had abnormal, borderline and normal sweat chloride results, respectively. Only 17 (8.4%) patients were diagnosable as having CF by genotyping. Compared to sweat testing, NPD identified more patients as having CF (33.2%) with fewer borderline results (18.8%). The level of agreement according to kappa statistics (and the observed percentage of agreement) between sweat chloride and NPD in RESP, PANC and AZOOSP subjects was 'moderate' (65% observed agreement), 'poor' (33% observed agreement) and 'fair' (28% observed agreement), respectively. The degree of agreement only improved marginally when subjects with borderline sweat chloride results were excluded from the analysis. CONCLUSIONS: The diagnosis of CF or its exclusion is not always straightforward and may remain elusive even with comprehensive evaluation, particularly among individuals who present at an older age with single-organ manifestations suggestive of CF.


Assuntos
Fibrose Cística/diagnóstico , Fibrose Cística/metabolismo , Mucosa Nasal/metabolismo , Cloreto de Sódio/metabolismo , Adulto , Alelos , Biomarcadores/metabolismo , Estudos de Coortes , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo , Valor Preditivo dos Testes , Estudos Prospectivos , Fatores de Risco , Sensibilidade e Especificidade , Suor/metabolismo
9.
Bioinformatics ; 29(15): 1843-50, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23736532

RESUMO

MOTIVATION: The prioritization and identification of disease-causing mutations is one of the most significant challenges in medical genomics. Currently available methods address this problem for non-synonymous single nucleotide variants (SNVs) and variation in promoters/enhancers; however, recent research has implicated synonymous (silent) exonic mutations in a number of disorders. RESULTS: We have curated 33 such variants from literature and developed the Silent Variant Analyzer (SilVA), a machine-learning approach to separate these from among a large set of rare polymorphisms. We evaluate SilVA's performance on in silico 'infection' experiments, in which we implant known disease-causing mutations into a human genome, and show that for 15 of 33 disorders, we rank the implanted mutation among the top five most deleterious ones. Furthermore, we apply the SilVA method to two additional datasets: synonymous variants associated with Meckel syndrome, and a collection of silent variants clinically observed and stratified by a molecular diagnostics laboratory, and show that SilVA is able to accurately predict the harmfulness of silent variants in these datasets. AVAILABILITY: SilVA is open source and is freely available from the project website: http://compbio.cs.toronto.edu/silva CONTACT: silva-snv@cs.toronto.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Inteligência Artificial , Doença/genética , Genoma Humano , Mutação , Transtornos da Motilidade Ciliar/genética , Simulação por Computador , Encefalocele/genética , Éxons , Genômica/métodos , Humanos , Doenças Renais Policísticas/genética , Polimorfismo Genético , Retinose Pigmentar
10.
Genet Med ; 16(8): 625-32, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24556927

RESUMO

PURPOSE: The purpose of this study was to determine the molecular consequences of the variant c.3700 A>G in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, a variant that has been predicted to cause a missense mutation in the CFTR protein (p.Ile1234Val). METHODS: Clinical assays of CFTR function were performed, and genomic DNA from patients homozygous for c.3700 A>G and their family members was sequenced. Total RNA was extracted from epithelial cells of the patients, transcribed into complementary DNA, and sequenced. CFTR complementary DNA clones containing the missense mutation p.Ile1234Val or a truncated exon 19 (p.Ile1234_Arg1239del) were constructed and heterologously expressed to test CFTR protein synthesis and processing. RESULTS: In vivo functional measurements revealed that the individuals homozygous for the variant c.3700 A>G exhibited defective CFTR function. We show that this mutation in exon 19 activates a cryptic donor splice site 18 bp upstream of the original donor splice site, resulting in deletion of six amino acids (r.3700_3717del; p.Ile1234_Arg1239del). This deletion, similar to p.Phe508del, causes a primary defect in folding and processing. Importantly, Lumacaftor (VX-809), currently in clinical trial for cystic fibrosis patients with the major cystic fibrosis-causing mutation, p.Phe508del, partially ameliorated the processing defect caused by p.Ile1234_Arg1239del. CONCLUSION: These studies highlight the need to verify molecular and clinical consequences of CFTR variants to define possible therapeutic strategies.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/genética , Isoleucina/metabolismo , Valina/metabolismo , Adolescente , Adulto , Aminopiridinas/farmacologia , Animais , Benzodioxóis/farmacologia , Linhagem Celular , Cricetinae , Fibrose Cística/tratamento farmacológico , Éxons , Células HEK293 , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto , Catar , Splicing de RNA
11.
Hum Mutat ; 34(8): 1057-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23636887

RESUMO

We have developed PhenoTips: open source software for collecting and analyzing phenotypic information for patients with genetic disorders. Our software combines an easy-to-use interface, compatible with any device that runs a Web browser, with a standardized database back end. The PhenoTips' user interface closely mirrors clinician workflows so as to facilitate the recording of observations made during the patient encounter. Collected data include demographics, medical history, family history, physical and laboratory measurements, physical findings, and additional notes. Phenotypic information is represented using the Human Phenotype Ontology; however, the complexity of the ontology is hidden behind a user interface, which combines simple selection of common phenotypes with error-tolerant, predictive search of the entire ontology. PhenoTips supports accurate diagnosis by analyzing the entered data, then suggesting additional clinical investigations and providing Online Mendelian Inheritance in Man (OMIM) links to likely disorders. By collecting, classifying, and analyzing phenotypic information during the patient encounter, PhenoTips allows for streamlining of clinic workflow, efficient data entry, improved diagnosis, standardization of collected patient phenotypes, and sharing of anonymized patient phenotype data for the study of rare disorders. Our source code and a demo version of PhenoTips are available at http://phenotips.org.


Assuntos
Bases de Dados Genéticas , Doenças Genéticas Inatas/diagnóstico , Doenças Genéticas Inatas/genética , Pesquisa em Genética , Fenótipo , Software , Interface Usuário-Computador , Algoritmos , Criança , Sistemas de Gerenciamento de Base de Dados , Bases de Dados Factuais , Ontologia Genética , Humanos , Armazenamento e Recuperação da Informação
12.
Hum Genet ; 132(11): 1245-52, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23812740

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a myocardial disease characterized by fibro-fatty replacement of right ventricular free wall myocardium and life-threatening ventricular arrhythmias. A missense mutation, c.1073C>T (p.S358L) in the transmembrane protein 43 (TMEM43) gene, has been genetically identified to cause ARVC type 5 in a founder population from Newfoundland. It is unclear whether this mutation occurs in other populations outside of this founder population or if other variants of TMEM43 are associated with ARVC disease. We sought to identify non-Newfoundland individuals with TMEM43 variants among patient samples sent for genetic assessment for possible ARVC. Of 195 unrelated individuals with suspected ARVC, mutation of desmosomal proteins was seen in 28 and the p.S358L TMEM43 mutation in six. We identified a de novo p.S358L mutation in a non-Newfoundland patient and five separate rare TMEM43 (four novel) sequence variants in non-Newfoundland patients, each occurring in an evolutionarily conserved amino acid. TMEM43 mutations occur outside of the founder population of the island of Newfoundland where it was originally described. TMEM43 sequencing should be incorporated into clinical genetic testing for ARVC patients.


Assuntos
Displasia Arritmogênica Ventricular Direita/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Arritmias Cardíacas/genética , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Desmossomos/genética , Desmossomos/metabolismo , Efeito Fundador , Predisposição Genética para Doença , Ventrículos do Coração/fisiopatologia , Heterozigoto , Humanos , Proteínas de Membrana/metabolismo , Terra Nova e Labrador , Linhagem , Análise de Sequência de DNA
13.
Am J Hum Genet ; 87(5): 631-42, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-21056402

RESUMO

DNA copy-number variations (CNVs) underlie many neuropsychiatric conditions, but they have been less studied in cancer. We report the association of a 17p13.1 CNV, childhood-onset developmental delay (DD), and cancer. Through a screen of over 4000 patients with diverse diagnoses, we identified eight probands harboring microdeletions at TP53 (17p13.1). We used a purpose-built high-resolution array with 93.75% breakpoint accuracy to fine map these microdeletions. Four patients were found to have a common phenotype including DD, hypotonia, and hand and foot abnormalities, constituting a unique syndrome. Notably, these patients were not affected with cancer. Moreover, none of the TP53-deletion patients affected with cancer (n = 4) had neurocognitive impairments. DD patients have larger deletions, which encompass but do not disrupt TP53, whereas cancer-affected patients harbor CNVs with at least one breakpoint within TP53. Most 17p13.1 deletions arise by Alu-mediated nonallelic homologous recombination. Furthermore, we identify a critical genomic region associated with DD and containing six underexpressed genes. We conclude that, although they overlap, 17p13.1 CNVs are associated with distinct phenotypes depending on the position of the breakpoint with respect to TP53. Further, detailed characterization of breakpoints revealed a common formation signature. Future studies should consider whether other loci in the genome also give rise to phenotypically distinct disorders by means of a common mechanism, resulting in a similar formation signature.


Assuntos
Neoplasias/genética , Adulto , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 17 , Variações do Número de Cópias de DNA , Deficiências do Desenvolvimento/genética , Genes p53 , Humanos , Fenótipo , Síndrome
14.
Am J Med Genet A ; 161A(1): 13-20, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23239666

RESUMO

Mosaicism for genome-wide paternal uniparental disomy (UPD) has been reported in only seven live born individuals to date. Clinical presentation includes manifestations of multiple paternal UPD syndromes with high variability, likely due to the variable levels of mosaicism in different somatic tissues. We report an eighth case in a female patient with mosaicism for genome-wide paternal UPD which highlights the complex clinical presentation. Our patient had features of Beckwith-Wiedemann syndrome (BWS), Angelman syndrome, and congenital hyperinsulinism. The clinical findings included prematurity, organomegaly, hemihyperplasia, developmental delay, benign tumors, and cystic lesions. The diagnosis in our patient was established utilizing microarray-based genome-wide DNA methylation analysis performed on leukocyte DNA. Targeted multiplex ligation-dependent probe amplification (MLPA) analysis of chromosome regions 11p15 and 15q13 confirmed mosaicism for paternal UPD at these genomic regions. This case represents the first report of microarray-based genome-wide DNA methylation analysis in the diagnosis of genome-wide paternal UPD. The application of microarray-based genome-wide DNA methylation analysis on selected individuals with complex clinical presentations could be a valuable diagnostic tool to improve the detection rate of mosaic genome-wide paternal UPD. This approach, which screens many loci simultaneously, is more cost-effective and less labor-intensive than performing multiple targeted DNA methylation-based assays. Identification of individuals with mosaicism for genome-wide paternal UPD is an important goal as it confers a low recurrence risk for the family and identifies individuals who require surveillance due to increased tumor risk.


Assuntos
Estudo de Associação Genômica Ampla/métodos , Impressão Genômica , Mosaicismo , Dissomia Uniparental/diagnóstico , Dissomia Uniparental/genética , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Cromossomos Humanos/genética , Hiperinsulinismo Congênito/diagnóstico , Hiperinsulinismo Congênito/genética , DNA/isolamento & purificação , Metilação de DNA , Feminino , Seguimentos , Perfilação da Expressão Gênica , Genômica , Genótipo , Humanos , Reação em Cadeia da Polimerase Multiplex , Análise de Sequência de DNA , População Branca
16.
Am J Med Genet A ; 158A(6): 1388-94, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22585446

RESUMO

Beckwith-Wiedemann syndrome (BWS) is an overgrowth disorder with variability in clinical manifestations and molecular causes. In most cases, patients with BWS have normal development. Cases with developmental delay are usually attributed to neonatal hypoglycemia or chromosome abnormalities involving copy number variation for genes beyond the critical BWS region at 11p15.5. Brain abnormalities have not previously been recognized within the BWS phenotypic spectrum. We report on seven cases of BWS associated with posterior fossa abnormalities. Of these, two cases presented with Blake's pouch cyst, two with Dandy-Walker variant (DWV; hypoplasia of the inferior part of the vermis), one with Dandy-Walker malformation (DWM) and one with a complex of DWM, dysgenesis of the corpus callosum and brain stem abnormality. In all these cases, molecular findings involved the centromeric imprinted domain on chromosome locus 11p15.5, which includes imprinting center 2 (IC2) and the imprinted growth suppressor gene, CDKN1C. Three cases had loss of methylation at IC2, two had CDKN1C mutations, and one had loss of methylation at IC2 and a microdeletion. In one case no mutation/methylation abnormality was detected. These findings together with previously reported correlations suggest that genes in imprinted domain 2 at 11p15.5 are involved in normal midline development of several organs including the brain. Our data suggest that brain malformations may present as a finding within the BWS phenotype when the molecular etiology involves imprinted domain 2. Brain imaging may be useful in identifying such malformations in individuals with BWS and neurodevelopmental issues.


Assuntos
Síndrome de Beckwith-Wiedemann/diagnóstico , Encéfalo/anormalidades , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/genética , Encéfalo/patologia , Pré-Escolar , Cromossomos Humanos Par 11 , Inibidor de Quinase Dependente de Ciclina p57/genética , Metilação de DNA , Evolução Fatal , Feminino , Deleção de Genes , Impressão Genômica , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Mutação
17.
Muscle Nerve ; 45(5): 752-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22499106

RESUMO

INTRODUCTION: Congenital muscular dystrophies (CMD) with hypoglycosylated α-dystroglycan due to POMT1 mutations are associated with clinical phenotypes that vary in severity. METHODS: We describe a patient with congenital hypotonia, generalized weakness, elevated creatine kinase (CK), and normal brain imaging. RESULTS: Histochemical analysis of the index case's muscle showed deficiency of glycosylated α-dystroglycan and secondary merosin deficiency. Genetic testing revealed a novel mutation in exon 20 of the POMT1 gene. CONCLUSIONS: Our patient's milder form of CMD adds to the emerging evidence of an expanding phenotype caused by POMT1 mutations. The histopathological findings of the muscle biopsy in this case support the need for careful clinical, genetic, and histochemical diagnostic interpretation.


Assuntos
Manosiltransferases/genética , Distrofias Musculares/genética , Mutação , Encéfalo/patologia , Creatina Quinase/sangue , Distroglicanas/metabolismo , Eletromiografia , Testes Genéticos , Glicosilação , Humanos , Lactente , Laminina/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofias Musculares/patologia , Distrofias Musculares/fisiopatologia , Fenótipo
19.
Can J Neurol Sci ; 38(3): 465-74, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21515508

RESUMO

INTRODUCTION: We carried out a population-based study of dystrophin mutations in patients followed by members of the Canadian Paediatric Neuromuscular Group (CPNG) over a ten-year period. OBJECTIVES: We aimed to describe the changes in diagnostic testing for dystrophinopathy and to determine the frequency of dystrophin mutations from 2000 to 2009. METHODS: De-identified data containing the clinical phenotypes, diagnostic methods, and mutational reports from dystrophinopathy patients followed by CPNG centres from January 2000 to December 2009 were analyzed using descriptive statistics. RESULTS: 773 patients had a confirmed diagnosis of dystrophinopathy based on genetic testing (97%), muscle biopsy (2%), or family history (1%). 573 (74%) had complete deletion/duplication analysis of all 79 exons or whole gene sequencing, resulting in 366 (64%) deletions, 64 (11%) duplications, and 143 (25%) point mutations. The percentage of patients who were diagnosed using currently accepted genetic testing methods varied across Canada, with a mean of 63% (SD 23). 246 (43%) mutations involved exons 45 to 53. The top ten deletions (n=147, 26%) were exons 45-47, 45-48, 45, 45-50, 45-55, 51, 45-49, 45-52, 49-50, and 46-47. 169 (29%) mutations involved exons 2 to 20. The most common duplications (n=29, 5.1%) were exons 2, 2-7, 2-17, 3-7, 8-11, 10, 10-11, and 12. CONCLUSION: This is the most comprehensive report of dystrophin mutations in Canada. Consensus guidelines regarding the diagnostic approach to dystrophinopathy will hopefully reduce the geographical variation in mutation detection rates in the coming decade.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/epidemiologia , Distrofia Muscular de Duchenne/genética , Mutação/genética , Canadá , Planejamento em Saúde Comunitária , Éxons/genética , Feminino , Testes Genéticos/métodos , Humanos , Estudos Longitudinais , Masculino , Distrofia Muscular de Duchenne/classificação , Distrofia Muscular de Duchenne/diagnóstico , Fenótipo , Prevalência , Estudos Retrospectivos , Estatística como Assunto , Fatores de Tempo
20.
Am J Med Genet A ; 152A(7): 1808-11, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20583174

RESUMO

Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency is an autosomal recessive disorder affecting mitochondrial fatty acid oxidation due to mutations in the HADHA gene. We report on a 22-month-old child who was identified on expanded newborn screening with an abnormal acylcarnitine pattern and increased C14OH. Molecular analysis showed that the child was homozygous for the common mutation, c.1526G > C (p.Glu510Gln) in the HADHA gene. Carrier testing on the parental samples revealed that the father was heterozygous for the mutation whereas the mother did not carry the mutation. Short tandem repeat testing with markers covering both short and long arms of chromosome 2 showed that the child has paternal uniparental isodisomy. We highlight the importance of parental testing in cases of homozygosity in autosomal recessive disorders and its impact on genetic counseling of the family.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/deficiência , Cromossomos Humanos Par 2/genética , Pai , Dissomia Uniparental/genética , 3-Hidroxiacil-CoA Desidrogenases/genética , Sequência de Bases , Segregação de Cromossomos/genética , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , 3-Hidroxiacil-CoA Desidrogenase de Cadeia Longa , Masculino , Dados de Sequência Molecular , Linhagem , Gravidez
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa