Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
J Clin Immunol ; 44(4): 98, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38598033

RESUMO

Biallelic null or hypomorphic variants in JAK3 cause SCID and less frequently Omenn syndrome. We investigated homozygous hypomorphic JAK3 mutations in two patients, and expression and function of a novel JAK3R431P variant in Omenn syndrome. Immunophenotyping of PBMC from the patient with the novel JAK3R431P variant was undertaken, by flow cytometry and Phosflow after stimulation with IL-2, IL-7, and IL-15. JAK3 expression was investigated by Western blotting. We report two patients with homozygous hypomorphic JAK3 variants and clinical features of Omenn syndrome. One patient had a previously described JAK3R775H variant, and the second had a novel JAK3R431P variant. One patient with a novel JAK3R431P variant had normal expression of JAK3 in immortalised EBV-LCL cells but reduced phosphorylation of STAT5 after stimulation with IL-2, IL-7, and IL-15 consistent with impaired kinase activity. These results suggest the JAK3R431P variant to be hypomorphic. Both patients are alive and well after allogeneic haematopoietic stem cell transplantation. They have full donor chimerism, restitution of thymopoiesis and development of appropriate antibody responses following vaccination. We expand the phenotype of hypomorphic JAK3 deficiency and demonstrate the importance of functional testing of novel variants in disease-causing genes.


Assuntos
Janus Quinase 3 , Imunodeficiência Combinada Severa , Humanos , Lactente , Interleucina-15 , Interleucina-2 , Interleucina-7 , Janus Quinase 3/genética , Leucócitos Mononucleares , Imunodeficiência Combinada Severa/diagnóstico , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia
3.
medRxiv ; 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38196618

RESUMO

To discover rare disease-gene associations, we developed a gene burden analytical framework and applied it to rare, protein-coding variants from whole genome sequencing of 35,008 cases with rare diseases and their family members recruited to the 100,000 Genomes Project (100KGP). Following in silico triaging of the results, 88 novel associations were identified including 38 with existing experimental evidence. We have published the confirmation of one of these associations, hereditary ataxia with UCHL1 , and independent confirmatory evidence has recently been published for four more. We highlight a further seven compelling associations: hypertrophic cardiomyopathy with DYSF and SLC4A3 where both genes show high/specific heart expression and existing associations to skeletal dystrophies or short QT syndrome respectively; monogenic diabetes with UNC13A with a known role in the regulation of ß cells and a mouse model with impaired glucose tolerance; epilepsy with KCNQ1 where a mouse model shows seizures and the existing long QT syndrome association may be linked; early onset Parkinson's disease with RYR1 with existing links to tremor pathophysiology and a mouse model with neurological phenotypes; anterior segment ocular abnormalities associated with POMK showing expression in corneal cells and with a zebrafish model with developmental ocular abnormalities; and cystic kidney disease with COL4A3 showing high renal expression and prior evidence for a digenic or modifying role in renal disease. Confirmation of all 88 associations would lead to potential diagnoses in 456 molecularly undiagnosed cases within the 100KGP, as well as other rare disease patients worldwide, highlighting the clinical impact of a large-scale statistical approach to rare disease gene discovery.

4.
Eur Respir J ; 60(5)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35728977

RESUMO

BACKGROUND: Bronchiectasis can result from infectious, genetic, immunological and allergic causes. 60-80% of cases are idiopathic, but a well-recognised genetic cause is the motile ciliopathy, primary ciliary dyskinesia (PCD). Diagnosis of PCD has management implications including addressing comorbidities, implementing genetic and fertility counselling and future access to PCD-specific treatments. Diagnostic testing can be complex; however, PCD genetic testing is moving rapidly from research into clinical diagnostics and would confirm the cause of bronchiectasis. METHODS: This observational study used genetic data from severe bronchiectasis patients recruited to the UK 100,000 Genomes Project and patients referred for gene panel testing within a tertiary respiratory hospital. Patients referred for genetic testing due to clinical suspicion of PCD were excluded from both analyses. Data were accessed from the British Thoracic Society audit, to investigate whether motile ciliopathies are underdiagnosed in people with bronchiectasis in the UK. RESULTS: Pathogenic or likely pathogenic variants were identified in motile ciliopathy genes in 17 (12%) out of 142 individuals by whole-genome sequencing. Similarly, in a single centre with access to pathological diagnostic facilities, 5-10% of patients received a PCD diagnosis by gene panel, often linked to normal/inconclusive nasal nitric oxide and cilia functional test results. In 4898 audited patients with bronchiectasis, <2% were tested for PCD and <1% received genetic testing. CONCLUSIONS: PCD is underdiagnosed as a cause of bronchiectasis. Increased uptake of genetic testing may help to identify bronchiectasis due to motile ciliopathies and ensure appropriate management.


Assuntos
Bronquiectasia , Transtornos da Motilidade Ciliar , Ciliopatias , Síndrome de Kartagener , Humanos , Mutação , Bronquiectasia/diagnóstico , Bronquiectasia/genética , Cílios , Transtornos da Motilidade Ciliar/diagnóstico , Transtornos da Motilidade Ciliar/genética , Ciliopatias/complicações , Síndrome de Kartagener/diagnóstico , Síndrome de Kartagener/genética
5.
Nat Genet ; 53(4): 500-510, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33782605

RESUMO

Spleen tyrosine kinase (SYK) is a critical immune signaling molecule and therapeutic target. We identified damaging monoallelic SYK variants in six patients with immune deficiency, multi-organ inflammatory disease such as colitis, arthritis and dermatitis, and diffuse large B cell lymphomas. The SYK variants increased phosphorylation and enhanced downstream signaling, indicating gain of function. A knock-in (SYK-Ser544Tyr) mouse model of a patient variant (p.Ser550Tyr) recapitulated aspects of the human disease that could be partially treated with a SYK inhibitor or transplantation of bone marrow from wild-type mice. Our studies demonstrate that SYK gain-of-function variants result in a potentially treatable form of inflammatory disease.


Assuntos
Artrite/genética , Colite/genética , Dermatite/genética , Linfoma Difuso de Grandes Células B/genética , Quinase Syk/genética , Adulto , Animais , Artrite/imunologia , Artrite/patologia , Artrite/terapia , Sequência de Bases , Transplante de Medula Óssea , Colite/imunologia , Colite/patologia , Colite/terapia , Dermatite/imunologia , Dermatite/patologia , Dermatite/terapia , Família , Feminino , Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Lactente , Linfoma Difuso de Grandes Células B/imunologia , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/terapia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Linhagem , Inibidores de Proteínas Quinases/farmacologia , Quinase Syk/antagonistas & inibidores , Quinase Syk/deficiência
6.
Life Sci Alliance ; 3(8)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32527837

RESUMO

The RNA exosome is a ubiquitously expressed complex of nine core proteins (EXOSC1-9) and associated nucleases responsible for RNA processing and degradation. Mutations in EXOSC3, EXOSC8, EXOSC9, and the exosome cofactor RBM7 cause pontocerebellar hypoplasia and motor neuronopathy. We investigated the consequences of exosome mutations on RNA metabolism and cellular survival in zebrafish and human cell models. We observed that levels of mRNAs encoding p53 and ribosome biogenesis factors are increased in zebrafish lines with homozygous mutations of exosc8 or exosc9, respectively. Consistent with higher p53 levels, mutant zebrafish have a reduced head size, smaller brain, and cerebellum caused by an increased number of apoptotic cells during development. Down-regulation of EXOSC8 and EXOSC9 in human cells leads to p53 protein stabilisation and G2/M cell cycle arrest. Increased p53 transcript levels were also observed in muscle samples from patients with EXOSC9 mutations. Our work provides explanation for the pathogenesis of exosome-related disorders and highlights the link between exosome function, ribosome biogenesis, and p53-dependent signalling. We suggest that exosome-related disorders could be classified as ribosomopathies.


Assuntos
Doenças Cerebelares/genética , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Ribossomos/metabolismo , Adulto , Animais , Linhagem Celular Tumoral , Doenças Cerebelares/fisiopatologia , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Exossomos/genética , Feminino , Homozigoto , Humanos , Masculino , Mutação , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribossomos/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
Blood ; 136(9): 1055-1066, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32518946

RESUMO

Molecular dissection of inborn errors of immunity can help to elucidate the nonredundant functions of individual genes. We studied 3 children with an immune dysregulation syndrome of susceptibility to infection, lymphadenopathy, hepatosplenomegaly, developmental delay, autoimmunity, and lymphoma of B-cell (n = 2) or T-cell (n = 1) origin. All 3 showed early autologous T-cell reconstitution following allogeneic hematopoietic stem cell transplantation. By whole-exome sequencing, we identified rare homozygous germline missense or nonsense variants in a known epigenetic regulator of gene expression: ten-eleven translocation methylcytosine dioxygenase 2 (TET2). Mutated TET2 protein was absent or enzymatically defective for 5-hydroxymethylating activity, resulting in whole-blood DNA hypermethylation. Circulating T cells showed an abnormal immunophenotype including expanded double-negative, but depleted follicular helper, T-cell compartments and impaired Fas-dependent apoptosis in 2 of 3 patients. Moreover, TET2-deficient B cells showed defective class-switch recombination. The hematopoietic potential of patient-derived induced pluripotent stem cells was skewed toward the myeloid lineage. These are the first reported cases of autosomal-recessive germline TET2 deficiency in humans, causing clinically significant immunodeficiency and an autoimmune lymphoproliferative syndrome with marked predisposition to lymphoma. This disease phenotype demonstrates the broad role of TET2 within the human immune system.


Assuntos
Proteínas de Ligação a DNA/deficiência , Mutação em Linhagem Germinativa , Mutação com Perda de Função , Transtornos Linfoproliferativos/genética , Proteínas Proto-Oncogênicas/deficiência , Imunodeficiência Combinada Severa/genética , Aloenxertos , Apoptose , Subpopulações de Linfócitos B/patologia , Técnicas de Reprogramação Celular , Códon sem Sentido , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Dioxigenases , Evolução Fatal , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Recém-Nascido , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/patologia , Masculino , Mutação de Sentido Incorreto , Neoplasias Primárias Múltiplas/genética , Linhagem , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Imunodeficiência Combinada Severa/patologia , Subpopulações de Linfócitos T/patologia , Sequenciamento do Exoma
8.
Neurology ; 90(21): e1842-e1848, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29720545

RESUMO

OBJECTIVE: To describe a patient with a multifocal demyelinating motor neuropathy with onset in childhood and a mutation in phosphatase and tensin homolog (PTEN), a tumor suppressor gene associated with inherited tumor susceptibility conditions, macrocephaly, autism, ataxia, tremor, and epilepsy. Functional implications of this protein have been investigated in Parkinson and Alzheimer diseases. METHODS: We performed whole-exome sequencing in the patient's genomic DNA validated by Sanger sequencing. Immunoblotting, in vitro enzymatic assay, and label-free shotgun proteomic profiling were performed in the patient's fibroblasts. RESULTS: The predominant clinical presentation of the patient was a childhood onset, asymmetric progressive multifocal motor neuropathy. In addition, he presented with macrocephaly, autism spectrum disorder, and skin hamartomas, considered as clinical criteria for PTEN-related hamartoma tumor syndrome. Extensive tumor screening did not detect any malignancies. We detected a novel de novo heterozygous c.269T>C, p.(Phe90Ser) PTEN variant, which was absent in both parents. The pathogenicity of the variant is supported by altered expression of several PTEN-associated proteins involved in tumorigenesis. Moreover, fibroblasts showed a defect in catalytic activity of PTEN against the secondary substrate, phosphatidylinositol 3,4-trisphosphate. In support of our findings, focal hypermyelination leading to peripheral neuropathy has been reported in PTEN-deficient mice. CONCLUSION: We describe a novel phenotype, PTEN-associated multifocal demyelinating motor neuropathy with a skin hamartoma syndrome. A similar mechanism may potentially underlie other forms of Charcot-Marie-Tooth disease with involvement of the phosphatidylinositol pathway.


Assuntos
Hamartoma/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Neuropatia Hereditária Motora e Sensorial/genética , PTEN Fosfo-Hidrolase/genética , Adulto , Predisposição Genética para Doença , Hamartoma/complicações , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/complicações , Neuropatia Hereditária Motora e Sensorial/complicações , Humanos , Masculino , Mutação , Sequenciamento do Exoma
9.
Hum Mol Genet ; 27(7): 1186-1195, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29361167

RESUMO

Mitochondrial dynamics play an important role in cellular homeostasis and a variety of human diseases are linked to its dysregulated function. Here, we describe a 15-year-old boy with a novel disease caused by altered mitochondrial dynamics. The patient was the second child of consanguineous Jewish parents. He developed progressive muscle weakness and exercise intolerance at 6 years of age. His muscle biopsy revealed mitochondrial myopathy with numerous ragged red and cytochrome c oxidase (COX) negative fibers and combined respiratory chain complex I and IV deficiency. MtDNA copy number was elevated and no deletions of the mtDNA were detected in muscle DNA. Whole exome sequencing identified a homozygous nonsense mutation (p.Q92*) in the MIEF2 gene encoding the mitochondrial dynamics protein of 49 kDa (MID49). Immunoblotting revealed increased levels of proteins promoting mitochondrial fusion (MFN2, OPA1) and decreased levels of the fission protein DRP1. Fibroblasts of the patient showed elongated mitochondria, and significantly higher frequency of fusion events, mtDNA abundance and aberrant mitochondrial cristae ultrastructure, compared with controls. Thus, our data suggest that mutations in MIEF2 result in imbalanced mitochondrial dynamics and a combined respiratory chain enzyme defect in skeletal muscle, leading to mitochondrial myopathy.


Assuntos
Fibroblastos/metabolismo , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais , Doenças Musculares , Mutação de Sentido Incorreto , Fatores de Alongamento de Peptídeos , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Fibroblastos/patologia , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Humanos , Masculino , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Doenças Musculares/genética , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Fatores de Alongamento de Peptídeos/genética , Fatores de Alongamento de Peptídeos/metabolismo , Cultura Primária de Células
10.
Neurol Genet ; 2(2): e59, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27123478

RESUMO

OBJECTIVE: To define the mechanism responsible for fatigue, lethargy, and weakness in 2 cousins who had a normal muscle biopsy. METHODS: Exome sequencing, long-range PCR, and Sanger sequencing to identify the pathogenic mutation. Functional analysis in the patient fibroblasts included oxygen consumption measurements, extracellular acidification studies, Western blotting, and calcium imaging, followed by overexpression of the wild-type protein. RESULTS: Analysis of the exome sequencing depth revealed a homozygous deletion of exon 1 of MICU1 within a 2,755-base pair deletion. No MICU1 protein was detected in patient fibroblasts, which had impaired mitochondrial calcium uptake that was rescued through the overexpression of the wild-type allele. CONCLUSIONS: MICU1 mutations cause fatigue and lethargy in patients with normal mitochondrial enzyme activities in muscle. The fluctuating clinical course is likely mediated through the mitochondrial calcium uniporter, which is regulated by MICU1.

11.
Neuromuscul Disord ; 25(6): 516-21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25900305

RESUMO

Mutations in the transient receptor potential vanilloid 4 (TRPV4) gene have been associated with autosomal dominant skeletal dysplasias and peripheral nervous system syndromes (PNSS). PNSS include Charcot-Marie-Tooth disease (CMT) type 2C, congenital spinal muscular atrophy and arthrogryposis and scapuloperoneal spinal muscular atrophy. We report the clinical, electrophysiological and muscle biopsy findings in two unrelated patients with two novel heterozygous missense mutations in the TRPV4 gene. Whole exome sequencing was carried out on genomic DNA using Illumina Truseq(TM) 62Mb exome capture. Patient 1 harbours a de novo c.805C > T (p.Arg269Cys) mutation. Clinically, this patient shows signs of both scapuloperoneal spinal muscular atrophy and skeletal dysplasia. Patient 2 harbours a novel c.184G > A (p.Asp62Asn) mutation. While the clinical phenotype is compatible with CMT type 2C with the patient's muscle harbours basophilic inclusions. Mutations in the TRPV4 gene have a broad phenotypic variability and disease severity and may share a similar pathogenic mechanism with Heat Shock Protein related neuropathies.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Nanismo/genética , Atrofia Muscular Espinal/genética , Osteocondrodisplasias/genética , Canais de Cátion TRPV/genética , Doença de Charcot-Marie-Tooth/patologia , Criança , Nanismo/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Atrofia Muscular Espinal/patologia , Mutação de Sentido Incorreto , Osteocondrodisplasias/patologia , Fenótipo
13.
JAMA Neurol ; 72(1): 106-11, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25420100

RESUMO

IMPORTANCE: Progressive external ophthalmoplegia (PEO) is a common feature in adults with mitochondrial (mt) DNA maintenance disorders associated with somatic mtDNA deletions in muscle, yet the causal genetic defect in many patients remains undetermined. OBSERVATIONS: Whole-exome sequencing identified a novel, heterozygous p.(Gly671Trp) mutation in the AFG3L2 gene encoding an mt protease--previously associated with dominant spinocerebellar ataxia type 28 disease--in a patient with indolent ataxia and PEO. Targeted analysis of a larger, genetically undetermined cohort of patients with PEO with suspected mtDNA maintenance abnormalities identified a second unrelated patient with a similar phenotype and a novel, heterozygous p.(Tyr689His) AFG3L2 mutation. Analysis of patient fibroblasts revealed mt fragmentation and decreased AFG3L2 transcript expression. Western blotting of patient fibroblast and muscle showed decreased AFG3L2 protein levels. CONCLUSIONS AND RELEVANCE: Our observations suggest that AFG3L2 mutations are another important cause, albeit rare, of a late-onset ataxic PEO phenotype due to a disturbance of mtDNA maintenance.


Assuntos
Proteases Dependentes de ATP/genética , DNA Mitocondrial/genética , Doenças Mitocondriais/genética , Degenerações Espinocerebelares/genética , ATPases Associadas a Diversas Atividades Celulares , Idoso , Animais , Estudos de Casos e Controles , Evolução Molecular , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Estudo de Associação Genômica Ampla , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação , Oftalmoplegia Externa Progressiva Crônica/genética , Ataxias Espinocerebelares/congênito , Degenerações Espinocerebelares/patologia
14.
Am J Hum Genet ; 95(3): 332-9, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25192047

RESUMO

Synaptotagmin 2 is a synaptic vesicle protein that functions as a calcium sensor for neurotransmission but has not been previously associated with human disease. Via whole-exome sequencing, we identified heterozygous missense mutations in the C2B calcium-binding domain of the gene encoding Synaptotagmin 2 in two multigenerational families presenting with peripheral motor neuron syndromes. An essential calcium-binding aspartate residue, Asp307Ala, was disrupted by a c.920A>C change in one family that presented with an autosomal-dominant presynaptic neuromuscular junction disorder resembling Lambert-Eaton myasthenic syndrome. A c.923C>T variant affecting an adjacent residue (p.Pro308Leu) produced a presynaptic neuromuscular junction defect and a dominant hereditary motor neuropathy in a second family. Characterization of the mutation homologous to the human c.920A>C variant in Drosophila Synaptotagmin revealed a dominant disruption of synaptic vesicle exocytosis using this transgenic model. These findings indicate that Synaptotagmin 2 regulates neurotransmitter release at human peripheral motor nerve terminals. In addition, mutations in the Synaptotagmin 2 C2B domain represent an important cause of presynaptic congenital myasthenic syndromes and link them with hereditary motor axonopathies.


Assuntos
Genes Dominantes/genética , Síndrome Miastênica de Lambert-Eaton/genética , Doença dos Neurônios Motores/genética , Mutação/genética , Doenças do Sistema Nervoso Periférico/genética , Sinaptotagmina II/genética , Adolescente , Adulto , Idoso , Animais , Criança , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Eletrofisiologia , Exocitose/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Transmissão Sináptica , Adulto Jovem
15.
PLoS Genet ; 10(6): e1004424, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24901367

RESUMO

Identifying the genetic basis for mitochondrial diseases is technically challenging given the size of the mitochondrial proteome and the heterogeneity of disease presentations. Using next-generation exome sequencing, we identified in a patient with severe combined mitochondrial respiratory chain defects and corresponding perturbation in mitochondrial protein synthesis, a homozygous p.Arg323Gln mutation in TRIT1. This gene encodes human tRNA isopentenyltransferase, which is responsible for i6A37 modification of the anticodon loops of a small subset of cytosolic and mitochondrial tRNAs. Deficiency of i6A37 was previously shown in yeast to decrease translational efficiency and fidelity in a codon-specific manner. Modelling of the p.Arg323Gln mutation on the co-crystal structure of the homologous yeast isopentenyltransferase bound to a substrate tRNA, indicates that it is one of a series of adjacent basic side chains that interact with the tRNA backbone of the anticodon stem, somewhat removed from the catalytic center. We show that patient cells bearing the p.Arg323Gln TRIT1 mutation are severely deficient in i6A37 in both cytosolic and mitochondrial tRNAs. Complete complementation of the i6A37 deficiency of both cytosolic and mitochondrial tRNAs was achieved by transduction of patient fibroblasts with wild-type TRIT1. Moreover, we show that a previously-reported pathogenic m.7480A>G mt-tRNASer(UCN) mutation in the anticodon loop sequence A36A37A38 recognised by TRIT1 causes a loss of i6A37 modification. These data demonstrate that deficiencies of i6A37 tRNA modification should be considered a potential mechanism of human disease caused by both nuclear gene and mitochondrial DNA mutations while providing insight into the structure and function of TRIT1 in the modification of cytosolic and mitochondrial tRNAs.


Assuntos
Alquil e Aril Transferases/genética , Doenças Mitocondriais/genética , Sulfurtransferases/genética , Células Cultivadas , Deficiência de Citocromo-c Oxidase/genética , Citosol , DNA Mitocondrial/genética , Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Feminino , Humanos , Masculino , Mitocôndrias/genética , Biossíntese de Proteínas/genética , RNA/genética , RNA Mitocondrial , RNA de Transferência/genética , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Schizosaccharomyces/enzimologia , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética
16.
Mov Disord ; 27(6): 789-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22508347

RESUMO

BACKGROUND: Neurodegeneration with brain iron accumulation is clinically and genetically heterogeneous because of mutations in at least 7 nuclear genes. METHODS: We performed homozygosity mapping and whole-exome sequencing in 2 brothers with brain iron accumulation from a consanguineous family. RESULTS: We identified a homozygous missense mutation in both brothers in the very recently identified chromosome 19 open-reading frame 12 gene. The disease presented before age 10 with slowly progressive tremor, dystonia, and spasticity. Additional features were optic atrophy, peripheral neuropathy, and learning difficulties. A raised serum creatine kinase indicated neuromuscular involvement, and compensatory mitochondrial proliferation implicated mitochondrial dysfunction as a pathological mechanism. CONCLUSIONS: Further studies are needed to explore the function of the chromosome 19 open-reading frame 12 gene, and extended genetic analysis on larger patient cohorts will provide more information about the presentation and frequency of this disease.


Assuntos
Encéfalo/metabolismo , Distúrbios Distônicos/genética , Ferro/metabolismo , Degeneração Neural/genética , Atrofia Óptica/genética , Doenças do Sistema Nervoso Periférico/genética , Adolescente , Encéfalo/patologia , Criança , Consanguinidade , Distúrbios Distônicos/metabolismo , Distúrbios Distônicos/patologia , Humanos , Masculino , Mutação de Sentido Incorreto , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Atrofia Óptica/metabolismo , Atrofia Óptica/patologia , Linhagem , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Síndrome
17.
Blood ; 118(10): 2656-8, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21765025

RESUMO

The human syndrome of dendritic cell, monocyte, B and natural killer lymphoid deficiency presents as a sporadic or autosomal dominant trait causing susceptibility to mycobacterial and other infections, predisposition to myelodysplasia and leukemia, and, in some cases, pulmonary alveolar proteinosis. Seeking a genetic cause, we sequenced the exomes of 4 unrelated persons, 3 with sporadic disease, looking for novel, heterozygous, and probably deleterious variants. A number of genes harbored novel variants in person, but only one gene, GATA2, was mutated in all 4 persons. Each person harbored a different mutation, but all were predicted to be highly deleterious and to cause loss or mutation of the C-terminal zinc finger domain. Because GATA2 is the only common mutated gene in 4 unrelated persons, it is highly probable to be the cause of dendritic cell, monocyte, B, and natural killer lymphoid deficiency. This disorder therefore constitutes a new genetic form of heritable immunodeficiency and leukemic transformation.


Assuntos
Linfócitos B/patologia , Células Dendríticas/patologia , Suscetibilidade a Doenças/etiologia , Éxons/genética , Fator de Transcrição GATA2/genética , Células Matadoras Naturais/patologia , Tecido Linfoide/patologia , Monócitos/patologia , Mutação/genética , Fator de Transcrição GATA2/química , Humanos , Conformação Proteica
18.
PLoS One ; 4(3): e4978, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19308252

RESUMO

Several previous studies have investigated the role of common promoter variants in the vascular endothelial growth factor (VEGF) gene in causing congenital cardiovascular malformation (CVM). However, results have been discrepant between studies and no study to date has comprehensively characterised variation throughout the gene. We genotyped 771 CVM cases, of whom 595 had the outflow tract malformation Tetralogy of Fallot (TOF), and carried out TDT and case-control analyses using haplotype-tagging SNPs in VEGF. We carried out a meta-analysis of previous case-control or family-based studies that had typed VEGF promoter SNPs, which included an additional 570 CVM cases. To identify rare variants potentially causative of CVM, we carried out mutation screening in all VEGF exons and splice sites in 93 TOF cases. There was no significant effect of any VEGF haplotype-tagging SNP on the risk of CVM in our analyses of 771 probands. When the results of this and all previous studies were combined, there was no significant effect of the VEGF promoter SNPs rs699947 (OR 1.05 [95% CI 0.95-1.17]); rs1570360 (OR 1.17 [95% CI 0.99-1.26]); and rs2010963 (OR 1.04 [95% CI 0.93-1.16]) on the risk of CVM in 1341 cases. Mutation screening of 93 TOF cases revealed no VEGF coding sequence variants and no changes at splice consensus sequences. Genetic variation in VEGF appears to play a small role, if any, in outflow tract CVM susceptibility.


Assuntos
Anormalidades Cardiovasculares/genética , Variação Genética , Fator A de Crescimento do Endotélio Vascular/genética , Anormalidades Cardiovasculares/epidemiologia , Anormalidades Cardiovasculares/etiologia , Estudos de Casos e Controles , Genótipo , Humanos , Mutação , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Risco , Tetralogia de Fallot/genética
19.
J Heart Valve Dis ; 11(2): 242-7; discussion 248, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12000167

RESUMO

BACKGROUND AND AIMS OF THE STUDY: The reduced availability of homografts and investigations into the ideal valved conduit have prompted a search for alternative options. This prospective study evaluated the Contegra, a new biological valved conduit consisting of a glutaraldehyde-preserved heterologous bovine jugular vein having a trileaflet venous valve. METHODS: Between April 1999 and May 2001, 26 patients (mean age 13.5+/-12.5 years; range: 4 months to 43 years), of mean body weight 33.9+/-26.7 kg (range: 4.0-91.0 kg), underwent implantation of a Contegra valved conduit for the following diagnoses: pulmonary valve replacement during Ross operation (n = 11), pulmonary atresia with ventricular septal defect (n = 6), tetralogy of Fallot (n = 3), truncus arteriosus (n = 3), Taussig-Bing syndrome (n = 2), and pulmonary valve regurgitation (n = 1). Conduit size was 14 mm in two patients, 16 mm in four, 18 mm in three, 20 mm in three, and 22 mm in 14. Mean cardiopulmonary bypass time was 156+/-33 min (range: 105-233 min), and mean aortic cross-clamp time 79+/-29 min (range: 0-128 min). RESULTS: There were no hospital deaths. During a mean follow up of 14.7+/-6.9 months (range: 1-26 months) there was one late death (4% mortality) that was unrelated to the valved conduit. Three children underwent reoperation; in all cases this was nonconduit-related. All 25 late survivors are currently free from reoperation and complications due to presence of the conduit. Echocardiographically monitored valve regurgitation was absent in 12 cases, trivial in nine and mild in four. The discharge situation was maintained during systematic follow up. No difference was found between the pressure gradient at discharge and at the last follow up. CONCLUSION: The new conduit provided consistently good results during its first two years of use. The main advantages were: (i) excellent off-the-shelf availability; (ii) large variety of available sizes (12 to 22 mm internal diameter); (iii) easy tailoring and suturing; (iv) adequate hemodynamics; (v) no need for proximal or distal extension; and (vi) reduced cost (half that of a conventional homograft).


Assuntos
Ventrículos do Coração/cirurgia , Veias Jugulares/cirurgia , Artéria Pulmonar/patologia , Artéria Pulmonar/cirurgia , Valva Pulmonar/patologia , Valva Pulmonar/cirurgia , Adolescente , Adulto , Criança , Proteção da Criança , Pré-Escolar , Ecocardiografia , Seguimentos , Humanos , Lactente , Bem-Estar do Lactente , Alta do Paciente , Estudos Prospectivos , Reoperação , Análise de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA