Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Ann Clin Transl Neurol ; 7(1): 121-125, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31846234

RESUMO

The combination of congenital bilateral perisylvian syndrome (CBPS) with lower motor neuron dysfunction remains unusual and suggests a potential common genetic insult affecting basic neurodevelopmental processes. Here we identify a putatively pathogenic missense mutation in the MCF2 gene in a boy with CBPS. Using in utero electroporation to genetically manipulate cortical neurons during corticogenesis, we demonstrate that the mouse Mcf2 gene controls the embryonic migration of cortical projection neurons. Strikingly, we find that the CBPS-associated MCF2 mutation impairs cortical laminar positioning, supporting the hypothesis that alterations in the process of embryonic neuronal migration can lead to rare cases of CBPS.


Assuntos
Anormalidades Múltiplas/genética , Córtex Cerebral/embriologia , Fatores de Troca do Nucleotídeo Guanina/genética , Deficiência Intelectual/genética , Malformações do Desenvolvimento Cortical/genética , Doença dos Neurônios Motores/genética , Proteínas Proto-Oncogênicas/genética , Adulto , Animais , Movimento Celular , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto , Adulto Jovem
2.
Am J Hum Genet ; 103(4): 568-578, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30290152

RESUMO

Infantile and childhood-onset cataracts form a heterogeneous group of disorders; among the many genetic causes, numerous pathogenic variants in additional genes associated with autosomal-recessive infantile cataracts remain to be discovered. We identified three consanguineous families affected by bilateral infantile cataracts. Using exome sequencing, we found homozygous loss-of-function variants in DNMBP: nonsense variant c.811C>T (p.Arg271∗) in large family F385 (nine affected individuals; LOD score = 5.18 at θ = 0), frameshift deletion c.2947_2948del (p.Asp983∗) in family F372 (two affected individuals), and frameshift variant c.2852_2855del (p.Thr951Metfs∗41) in family F3 (one affected individual). The phenotypes of all affected individuals include infantile-onset cataracts. RNAi-mediated knockdown of the Drosophila ortholog still life (sif), enriched in lens-secreting cells, affects the development of these cells as well as the localization of E-cadherin, alters the distribution of septate junctions in adjacent cone cells, and leads to a ∼50% reduction in electroretinography amplitudes in young flies. DNMBP regulates the shape of tight junctions, which correspond to the septate junctions in invertebrates, as well as the assembly pattern of E-cadherin in human epithelial cells. E-cadherin has an important role in lens vesicle separation and lens epithelial cell survival in humans. We therefore conclude that DNMBP loss-of-function variants cause infantile-onset cataracts in humans.


Assuntos
Catarata/genética , Proteínas do Citoesqueleto/genética , Predisposição Genética para Doença/genética , Variação Genética/genética , Perda de Heterozigosidade/genética , Adulto , Alelos , Animais , Caderinas/genética , Criança , Drosophila/genética , Células Epiteliais/patologia , Exoma/genética , Feminino , Homozigoto , Humanos , Escore Lod , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Junções Íntimas/patologia
3.
Eur J Hum Genet ; 26(3): 330-339, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29343805

RESUMO

Kinesin proteins are critical for various cellular functions such as intracellular transport and cell division, and many members of the family have been linked to monogenic disorders and cancer. We report eight individuals with intellectual disability and microcephaly from four unrelated families with parental consanguinity. In the affected individuals of each family, homozygosity for likely pathogenic variants in KIF14 were detected; two loss-of-function (p.Asn83Ilefs*3 and p.Ser1478fs), and two missense substitutions (p.Ser841Phe and p.Gly459Arg). KIF14 is a mitotic motor protein that is required for spindle localization of the mitotic citron rho-interacting kinase, CIT, also mutated in microcephaly. Our results demonstrate the involvement of KIF14 in development and reveal a wide phenotypic variability ranging from fetal lethality to moderate developmental delay and microcephaly.


Assuntos
Deficiência Intelectual/genética , Cinesinas/genética , Microcefalia/genética , Proteínas Oncogênicas/genética , Criança , Pré-Escolar , Feminino , Humanos , Deficiência Intelectual/patologia , Cinesinas/química , Cinesinas/metabolismo , Mutação com Perda de Função , Microcefalia/patologia , Mutação de Sentido Incorreto , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Linhagem , Fenótipo , Domínios Proteicos , Síndrome
4.
Hum Genomics ; 10(1): 26, 2016 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-27421267

RESUMO

BACKGROUND: The recent availability of whole-exome sequencing has opened new possibilities for the evaluation of individuals with genetically undiagnosed intellectual disability. RESULTS: We report two affected siblings, offspring of first-cousin parents, with intellectual disability, hypotonia, short stature, growth hormone deficiency, and delayed bone age. All members of the nuclear family were genotyped, and exome sequencing was performed in one of the affected individuals. We used an in-house algorithm (CATCH v1.1) that combines homozygosity mapping with exome sequencing results and provides a list of candidate variants. One identified novel homozygous missense variant in KALRN (NM_003947.4:c.3644C>A: p.(Thr1215Lys)) was predicted to be pathogenic by all pathogenicity prediction software used (SIFT, PolyPhen, Mutation Taster). KALRN encodes the protein kalirin, which is a GTP-exchange factor protein with a reported role in cytoskeletal remodeling and dendritic spine formation in neurons. It is known that mice with ablation of Kalrn exhibit age-dependent functional deficits and behavioral phenotypes. CONCLUSION: Exome sequencing provided initial evidence linking KALRN to monogenic intellectual disability in man, and we propose that KALRN is the causative gene for the autosomal recessive phenotype in this family.


Assuntos
Deficiências do Desenvolvimento/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Deficiência Intelectual/genética , Proteínas Serina-Treonina Quinases/genética , Adolescente , Criança , Consanguinidade , Análise Mutacional de DNA , Exoma/genética , Feminino , Estudos de Associação Genética , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto
5.
Am J Hum Genet ; 98(4): 615-26, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26996948

RESUMO

Glycosylphosphatidylinositol (GPI) is a glycolipid that anchors >150 various proteins to the cell surface. At least 27 genes are involved in biosynthesis and transport of GPI-anchored proteins (GPI-APs). To date, mutations in 13 of these genes are known to cause inherited GPI deficiencies (IGDs), and all are inherited as recessive traits. IGDs mainly manifest as intellectual disability, epilepsy, coarse facial features, and multiple organ anomalies. These symptoms are caused by the decreased surface expression of GPI-APs or by structural abnormalities of GPI. Here, we present five affected individuals (from two consanguineous families from Egypt and Pakistan and one non-consanguineous family from Japan) who show intellectual disability, hypotonia, and early-onset seizures. We identified pathogenic variants in PIGG, a gene in the GPI pathway. In the consanguineous families, homozygous variants c.928C>T (p.Gln310(∗)) and c.2261+1G>C were found, whereas the Japanese individual was compound heterozygous for c.2005C>T (p.Arg669Cys) and a 2.4 Mb deletion involving PIGG. PIGG is the enzyme that modifies the second mannose with ethanolamine phosphate, which is removed soon after GPI is attached to the protein. Physiological significance of this transient modification has been unclear. Using B lymphoblasts from affected individuals of the Egyptian and Japanese families, we revealed that PIGG activity was almost completely abolished; however, the GPI-APs had normal surface levels and normal structure, indicating that the pathogenesis of PIGG deficiency is not yet fully understood. The discovery of pathogenic variants in PIGG expands the spectrum of IGDs and further enhances our understanding of this etiopathogenic class of intellectual disability.


Assuntos
Variação Genética , Glicosilfosfatidilinositóis/genética , Deficiência Intelectual/genética , Manosiltransferases/genética , Hipotonia Muscular/genética , Convulsões/genética , Anormalidades Múltiplas/genética , Adolescente , Linhagem Celular Tumoral , Criança , Consanguinidade , Egito , Técnicas de Genotipagem , Glicosilfosfatidilinositóis/metabolismo , Células HEK293 , Heterozigoto , Homozigoto , Humanos , Lactente , Japão , Mutação , Paquistão , Linhagem , Adulto Jovem
6.
Nat Genet ; 48(4): 398-406, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26950094

RESUMO

Basal cell carcinoma (BCC) of the skin is the most common malignant neoplasm in humans. BCC is primarily driven by the Sonic Hedgehog (Hh) pathway. However, its phenotypic variation remains unexplained. Our genetic profiling of 293 BCCs found the highest mutation rate in cancer (65 mutations/Mb). Eighty-five percent of the BCCs harbored mutations in Hh pathway genes (PTCH1, 73% or SMO, 20% (P = 6.6 × 10(-8)) and SUFU, 8%) and in TP53 (61%). However, 85% of the BCCs also harbored additional driver mutations in other cancer-related genes. We observed recurrent mutations in MYCN (30%), PPP6C (15%), STK19 (10%), LATS1 (8%), ERBB2 (4%), PIK3CA (2%), and NRAS, KRAS or HRAS (2%), and loss-of-function and deleterious missense mutations were present in PTPN14 (23%), RB1 (8%) and FBXW7 (5%). Consistent with the mutational profiles, N-Myc and Hippo-YAP pathway target genes were upregulated. Functional analysis of the mutations in MYCN, PTPN14 and LATS1 suggested their potential relevance in BCC tumorigenesis.


Assuntos
Carcinoma Basocelular/genética , Transdução de Sinais/efeitos da radiação , Neoplasias Cutâneas/genética , Anilidas/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinogênese/genética , Carcinoma Basocelular/tratamento farmacológico , Carcinoma Basocelular/patologia , Análise Mutacional de DNA , Progressão da Doença , Exoma , Estudos de Associação Genética , Predisposição Genética para Doença , Células HEK293 , Humanos , Mutação , Piridinas/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Transcriptoma
7.
Nat Commun ; 5: 5690, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25471132

RESUMO

Alteration of the number of copies of double minutes (DMs) with oncogenic EGFR mutations in response to tyrosine kinase inhibitors is a novel adaptive mechanism of glioblastoma. Here we provide evidence that such mutations in DMs, called here amplification-linked extrachromosomal mutations (ALEMs), originate extrachromosomally and could therefore be completely eliminated from the cancer cells. By exome sequencing of seven glioblastoma patients we reveal ALEMs in EGFR, PDGFRA and other genes. These mutations together with DMs are lost by cancer cells in culture. We confirm the extrachromosomal origin of such mutations by showing that wild-type and mutated DMs may coexist in the same tumour. Analysis of 4,198 tumours suggests the presence of ALEMs across different tumour types with the highest prevalence in glioblastomas and low-grade gliomas. The extrachromosomal nature of ALEMs explains the observed drastic changes in the amounts of mutated oncogenes (like EGFR or PDGFRA) in glioblastoma in response to environmental changes.


Assuntos
Neoplasias Encefálicas/genética , DNA Circular/genética , Glioblastoma/genética , Neoplasias Encefálicas/patologia , Amplificação de Genes , Instabilidade Genômica , Glioma/genética , Glioma/patologia , Humanos , Mutação , Gradação de Tumores
8.
Bone ; 68: 142-5, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25180662

RESUMO

INTRODUCTION: Hypophosphatemic rickets (HR) comprises a rare group of inherited diseases. Very recently, mutations in the dentin matrix protein 1 (DMP1) gene were identified in patients with an extremely rare autosomal recessive form of HR (ARHR). To date, very few cases of these mutations were reported. MATERIALS AND METHODS: A Lebanese consanguineous family with 2 affected sisters was studied. Patients aged 45 and 47years old presented with short stature, severe genu varum, cranial hyperostosis and a very high bone density that led to a diagnosis of a familial sclerosing bone dysplasia. Molecular analysis of known genes involved in osteopetrosis showed normal results. A combination of genotyping and exome sequencing was performed in order to elucidate the genetic basis of this pathology. RESULTS: Biochemical analysis was consistent with normal serum calcium and 1-25(OH)2D levels, low to normal serum phosphorus and elevated PTH values. Serum c-terminal FGF-23 was elevated in one of the two patients. A homozygous mutation disrupting the initiation codon of the DMP1 gene (OMIM 600980), NM_001079911.2: c.1A>G, p.Met1Val, was identified by exome sequencing and confirmed by Sanger sequencing. CONCLUSION: We report here a family of ARHR secondary to a DMP1 mutation located in the first coding exon of the gene. Our cases show that some ARHR cases may develop with age an unaccountable increase in bone density and bone overgrowth.


Assuntos
Exoma/genética , Proteínas da Matriz Extracelular/genética , Osteocondrodisplasias/genética , Fosfoproteínas/genética , Adulto , Sequência de Bases , Análise Mutacional de DNA , Família , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação/genética , Osteocondrodisplasias/sangue , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/urina , Radiografia
9.
Nat Commun ; 5: 4654, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25105841

RESUMO

Children with Down syndrome (DS) and acute lymphoblastic leukaemia (ALL) have poorer survival and more relapses than non-DS children with ALL, highlighting an urgent need for deeper mechanistic understanding of DS-ALL. Here, using full-exome or cancer genes-targeted sequencing of 42 ALL samples from 39 DS patients, we uncover driver mutations in RAS, (KRAS and NRAS) recurring to a similar extent (15/42) as JAK2 (12/42) mutations or P2RY8-CRLF2 fusions (14/42). RAS mutations are almost completely mutually exclusive with JAK2 mutations (P=0.016), driving a combined total of two-thirds of analysed cases. Clonal architecture analysis reveals that both RAS and JAK2 drove sub-clonal expansions primarily initiated by CRLF2 rearrangements, and/or mutations in chromatin remodellers and lymphocyte differentiation factors. Remarkably, in 2/3 relapsed cases, there is a switch from a primary JAK2- or PTPN11-mutated sub-clone to a RAS-mutated sub-clone in relapse. These results provide important new insights informing the patient stratification strategies for targeted therapeutic approaches for DS-ALL.


Assuntos
Síndrome de Down/genética , Janus Quinase 2/genética , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/metabolismo , Criança , Cromatina/química , Análise Mutacional de DNA , Síndrome de Down/metabolismo , Feminino , Deleção de Genes , Regulação Leucêmica da Expressão Gênica , Humanos , Linfócitos/citologia , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Citocinas/genética
10.
Genome Res ; 24(2): 349-55, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24389049

RESUMO

There is increasing interest in clinical genetics pertaining to the utilization of high-throughput sequencing data for accurate diagnoses of monogenic diseases. Moreover, massive whole-exome sequencing of tumors has provided significant advances in the understanding of cancer development through the recognition of somatic driver variants. To improve the identification of the variants from HTS, we developed VariantMaster, an original program that accurately and efficiently extracts causative variants in familial and sporadic genetic diseases. The algorithm takes into account predicted variants (SNPs and indels) in affected individuals or tumor samples and utilizes the row (BAM) data to robustly estimate the conditional probability of segregation in a family, as well as the probability of it being de novo or somatic. In familial cases, various modes of inheritance are considered: X-linked, autosomal dominant, and recessive (homozygosity or compound heterozygosity). Moreover, VariantMaster integrates phenotypes and genotypes, and employs Annovar to produce additional information such as allelic frequencies in the general population and damaging scores to further reduce the number of putative variants. As a proof of concept, we successfully applied VariantMaster to identify (1) de novo mutations in a previously described data set, (2) causative variants in a rare Mendelian genetic disease, and (3) known and new "driver" mutations in previously reported cancer data sets. Our results demonstrate that VariantMaster is considerably more accurate in terms of precision and sensitivity compared with previously published algorithms.


Assuntos
Predisposição Genética para Doença , Mutação INDEL/genética , Polimorfismo de Nucleotídeo Único/genética , Software , Algoritmos , Biologia Computacional/métodos , Bases de Dados Genéticas , Frequência do Gene , Genótipo , Humanos , Fenótipo , Análise de Sequência de DNA
11.
EMBO Mol Med ; 6(2): 259-77, 2014 02.
Artigo em Inglês | MEDLINE | ID: mdl-24375627

RESUMO

Down syndrome (trisomy 21) is the most common viable chromosomal disorder with intellectual impairment and several other developmental abnormalities. Here, we report the generation and characterization of induced pluripotent stem cells (iPSCs) derived from monozygotic twins discordant for trisomy 21 in order to eliminate the effects of the variability of genomic background. The alterations observed by genetic analysis at the iPSC level and at first approximation in early development illustrate the developmental disease transcriptional signature of Down syndrome. Moreover, we observed an abnormal neural differentiation of Down syndrome iPSCs in vivo when formed teratoma in NOD-SCID mice, and in vitro when differentiated into neuroprogenitors and neurons. These defects were associated with changes in the architecture and density of neurons, astroglial and oligodendroglial cells together with misexpression of genes involved in neurogenesis, lineage specification and differentiation. Furthermore, we provide novel evidence that dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A) on chromosome 21 likely contributes to these defects. Importantly, we found that targeting DYRK1A pharmacologically or by shRNA results in a considerable correction of these defects.


Assuntos
Síndrome de Down/patologia , Síndrome de Down/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Modelos Biológicos , Gêmeos Monozigóticos/genética , Animais , Apoptose/genética , Diferenciação Celular/genética , Proliferação de Células , Síndrome de Down/genética , Ontologia Genética , Genoma Humano/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/patologia , Neurogênese/genética , Neurônios/metabolismo , Neurônios/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Transcriptoma/genética , Quinases Dyrk
12.
Am J Hum Genet ; 93(2): 346-56, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23891471

RESUMO

Primary ciliary dyskinesia (PCD) is a ciliopathy characterized by airway disease, infertility, and laterality defects, often caused by dual loss of the inner dynein arms (IDAs) and outer dynein arms (ODAs), which power cilia and flagella beating. Using whole-exome and candidate-gene Sanger resequencing in PCD-affected families afflicted with combined IDA and ODA defects, we found that 6/38 (16%) carried biallelic mutations in the conserved zinc-finger gene BLU (ZMYND10). ZMYND10 mutations conferred dynein-arm loss seen at the ultrastructural and immunofluorescence level and complete cilia immotility, except in hypomorphic p.Val16Gly (c.47T>G) homozygote individuals, whose cilia retained a stiff and slowed beat. In mice, Zmynd10 mRNA is restricted to regions containing motile cilia. In a Drosophila model of PCD, Zmynd10 is exclusively expressed in cells with motile cilia: chordotonal sensory neurons and sperm. In these cells, P-element-mediated gene silencing caused IDA and ODA defects, proprioception deficits, and sterility due to immotile sperm. Drosophila Zmynd10 with an equivalent c.47T>G (p.Val16Gly) missense change rescued mutant male sterility less than the wild-type did. Tagged Drosophila ZMYND10 is localized primarily to the cytoplasm, and human ZMYND10 interacts with LRRC6, another cytoplasmically localized protein altered in PCD. Using a fly model of PCD, we conclude that ZMYND10 is a cytoplasmic protein required for IDA and ODA assembly and that its variants cause ciliary dysmotility and PCD with laterality defects.


Assuntos
Cílios/genética , Dineínas/genética , Infertilidade Masculina/genética , Síndrome de Kartagener/genética , Proteínas/genética , Sistema Respiratório/metabolismo , Proteínas Supressoras de Tumor/genética , Animais , Axonema/genética , Axonema/metabolismo , Axonema/patologia , Cílios/metabolismo , Cílios/patologia , Proteínas do Citoesqueleto , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Dineínas/metabolismo , Exoma , Feminino , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patologia , Masculino , Camundongos , Mutação , Linhagem , Estrutura Terciária de Proteína , Proteínas/metabolismo , Sistema Respiratório/patologia , Proteínas Supressoras de Tumor/metabolismo
13.
Cancer Res ; 72(23): 6279-89, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23204322

RESUMO

Oncogene-induced DNA replication stress is thought to drive genomic instability in cancer. In particular, replication stress can explain the high prevalence of focal genomic deletions mapping within very large genes in human tumors. However, the origin of single-nucleotide substitutions (SNS) in nonfamilial cancers is strongly debated. Some argue that cancers have a mutator phenotype, whereas others argue that the normal DNA replication error rates are sufficient to explain the number of observed SNSs. Here, we sequenced the exomes of 24, mostly precancerous, colon polyps. Analysis of the sequences revealed mutations in the APC, CTNNB1, and BRAF genes as the presumptive cancer-initiating events and many passenger SNSs. We used the number of SNSs in the various lesions to calculate mutation rates for normal colon and adenomas and found that colon adenomas exhibit a mutator phenotype. Interestingly, the SNSs in the adenomas mapped more often than expected within very large genes, where focal deletions in response to DNA replication stress also map. We propose that single-stranded DNA generated in response to oncogene-induced replication stress compromises the repair of deaminated cytosines and other damaged bases, leading to the observed SNS mutator phenotype.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Exoma , Adenoma/patologia , Neoplasias Colorretais/patologia , Reparo do DNA/genética , Genoma Humano , Instabilidade Genômica , Humanos , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único
14.
Nat Genet ; 44(2): 133-9, 2011 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-22197931

RESUMO

We performed exome sequencing to detect somatic mutations in protein-coding regions in seven melanoma cell lines and donor-matched germline cells. All melanoma samples had high numbers of somatic mutations, which showed the hallmark of UV-induced DNA repair. Such a hallmark was absent in tumor sample-specific mutations in two metastases derived from the same individual. Two melanomas with non-canonical BRAF mutations harbored gain-of-function MAP2K1 and MAP2K2 (MEK1 and MEK2, respectively) mutations, resulting in constitutive ERK phosphorylation and higher resistance to MEK inhibitors. Screening a larger cohort of individuals with melanoma revealed the presence of recurring somatic MAP2K1 and MAP2K2 mutations, which occurred at an overall frequency of 8%. Furthermore, missense and nonsense somatic mutations were frequently found in three candidate melanoma genes, FAT4, LRP1B and DSC1.


Assuntos
Exoma/genética , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 2/genética , Melanoma/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Mutação , Neoplasias Cutâneas/genética , Sequência de Bases , Caderinas/genética , Linhagem Celular Tumoral , Estudos de Coortes , Reparo do DNA/genética , Desmocolinas , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas B-raf/genética , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética , Raios Ultravioleta/efeitos adversos
15.
Hum Mutat ; 29(1): 130-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17918732

RESUMO

Building on our discovery that mutations in the transmembrane serine protease, TMPRSS3, cause nonsyndromic deafness, we have investigated the contribution of other TMPRSS family members to the auditory function. To identify which of the 16 known TMPRSS genes had a strong likelihood of involvement in hearing function, three types of biological evidence were examined: 1) expression in inner ear tissues; 2) location in a genomic interval that contains a yet unidentified gene for deafness; and 3) evaluation of hearing status of any available Tmprss knockout mouse strains. This analysis demonstrated that, besides TMPRSS3, another TMPRSS gene was essential for hearing and, indeed, mice deficient for Hepsin (Hpn) also known as Tmprss1 exhibited profound hearing loss. In addition, TMPRSS2, TMPRSS5, and CORIN, also named TMPRSS10, showed strong likelihood of involvement based on their inner ear expression and mapping position within deafness loci PKSR7, DFNB24, and DFNB25, respectively. These four TMPRSS genes were then screened for mutations in affected members of the DFNB24 and DFNB25 deafness families, and in a cohort of 362 sporadic deaf cases. This large mutation screen revealed numerous novel sequence variations including three potential pathogenic mutations in the TMPRSS5 gene. The mutant forms of TMPRSS5 showed reduced or absent proteolytic activity. Subsequently, TMPRSS genes with evidence of involvement in deafness were further characterized, and their sites of expression were determined. Tmprss1, 3, and 5 proteins were detected in spiral ganglion neurons. Tmprss3 was also present in the organ of Corti. TMPRSS1 and 3 proteins appeared stably anchored to the endoplasmic reticulum membranes, whereas TMPRSS5 was also detected at the plasma membrane. Collectively, these results provide evidence that TMPRSS1 and TMPRSS3 play and TMPRSS5 may play important and specific roles in hearing.


Assuntos
Perda Auditiva/genética , Proteínas de Membrana/genética , Serina Endopeptidases/genética , Animais , Orelha Interna/metabolismo , Perda Auditiva/enzimologia , Humanos , Imuno-Histoquímica , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/análise , Serina Endopeptidases/metabolismo
16.
Front Biosci ; 13: 1557-67, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981648

RESUMO

Recently, we and others have shown that mutations in TMPRSS3 were responsible for autosomal recessive non-syndromic hearing loss. TMPRSS3 is a member of the Type II Transmembrane Serine Protease (TTSP) family and encodes for a protease that also contains LDLRA (low-density lipoprotein receptor class A) and SRCR (scavenger receptor cysteine rich) domains. Fourteen pathogenic mutations, which occur not only in the catalytic domain but also in the LDLRA and SRCR domains, have been identified to date that cause the DFNB8/10 forms of deafness. In vitro experiments demonstrated that TMPRSS3 mutants were proteolytically inactive indicating that TMPRSS3 protease activity is critical for normal auditory function. However, how missense mutations in the LDLRA and SRCR domains affect the proteolytic activity of TMPRSS3 remains to be elucidated. Although the role of TMPRSS3 in the auditory system is currently not completely understood, it has been shown to regulate the activity of the ENaC sodium channel in vitro and could therefore participate in the regulation of sodium concentration in the cochlea. TMPRSS3 mutations are not a common cause of hereditary deafness, the elucidation of its function is nevertheless important for better understanding of hearing, and provide biological targets for therapeutic interventions.


Assuntos
Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Serina Endopeptidases/metabolismo , Adolescente , Criança , Pré-Escolar , Mapeamento Cromossômico , Cromossomos Humanos Par 21/genética , Análise Mutacional de DNA , Saúde da Família , Genes Recessivos , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/fisiologia
17.
Pathology ; 38(4): 336-42, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16916724

RESUMO

AIMS: The molecular pathogenesis of essential thrombocythaemia (ET) is heterogeneous. We aimed to determine the relative sensitivity of four separate molecular assays used to detect the presence of the JAK2 V617F mutation in peripheral blood from patients with essential thrombocythaemia and related myeloproliferative disorders. METHODS: Purified granulocytes from 60 patients were analysed for the presence of the JAK2 V617F mutation by direct sequencing, denaturing high-performance liquid chromatography (DHPLC), allele-specific polymerase chain reaction (PCR) and allele-specific enrichment. Clinical data were collected for all patients and correlated with assay results. RESULTS: Direct sequencing and DHPLC were relatively insensitive assays for mutation detection, together identifying only 53% of the JAK2 V617F positive cases of ET. Allele-specific PCR and allele-specific enrichment were significantly more sensitive assays and were able to identify additional ET patients that were positive for the JAK2 V617F mutation in only a minority of circulating granulocytes. Enrichment for the mutation was demonstrated in blood platelets from two of these patients. CONCLUSIONS: The observed biological difference in circulating granulocyte involvement by the JAK2 V617F clone necessitates a sensitive molecular assay for the diagnostic investigation of thrombocytosis.


Assuntos
Análise Mutacional de DNA/métodos , Heterogeneidade Genética , Granulócitos/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Trombocitemia Essencial/sangue , Trombocitemia Essencial/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Cromatografia Líquida de Alta Pressão , DNA/análise , DNA/genética , Feminino , Granulócitos/patologia , Humanos , Janus Quinase 2 , Masculino , Pessoa de Meia-Idade , Mutação , Reação em Cadeia da Polimerase , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sensibilidade e Especificidade
18.
Hum Mol Genet ; 11(23): 2829-36, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393794

RESUMO

TMPRSS3 encodes a transmembrane serine protease that contains both LDLRA and SRCR domains and is mutated in non-syndromic autosomal recessive deafness (DFNB8/10). To study its function, we cloned the mouse ortholog which maps to Mmu17, which is structurally similar to the human gene and encodes a polypeptide with 88% identity to the human protein. RT-PCR and RNA in situ hybridization on rat and mouse cochlea revealed that Tmprss3 is expressed in the spiral ganglion, the cells supporting the organ of Corti and the stria vascularis. RT-PCR on mouse tissues showed expression in the thymus, stomach, testis and E19 embryos. Transient expression of wild-type or tagged TMPRSS3 protein showed a primary localization in the endoplasmic reticulum. The epithelial amiloride-sensitive sodium channel (ENaC), which is expressed in many sodium-reabsorbing tissues including the inner ear and is regulated by membrane-bound channel activating serine proteases (CAPs), is a potential substrate of TMPRSS3. In the Xenopus oocyte expression system, proteolytic processing of TMPRSS3 was associated with increased ENaC mediated currents. In contrast, 6 TMPRSS3 mutants (D103G, R109W, C194F, W251C, P404L, C407R) causing deafness and a mutant in the catalytic triad of TMPRSS3 (S401A), failed to undergo proteolytic cleavage and activate ENaC. These data indicate that important signaling pathways in the inner ear are controlled by proteolytic cleavage and suggest: (i) the existence of an auto-catalytic processing by which TMPRSS3 would become active, and (ii) that ENaC could be a substrate of TMPRSS3 in the inner ear.


Assuntos
Surdez/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação de Sentido Incorreto/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Canais de Sódio/metabolismo , Animais , Sítios de Ligação , Western Blotting , Análise Mutacional de DNA , Primers do DNA/química , Surdez/metabolismo , Retículo Endoplasmático/metabolismo , Canais Epiteliais de Sódio , Feminino , Genes Recessivos/genética , Genótipo , Humanos , Hibridização In Situ , Técnicas In Vitro , Masculino , Camundongos , Oócitos/metabolismo , Órgão Espiral/metabolismo , Transporte Proteico , RNA Mensageiro/metabolismo , Coelhos , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gânglio Espiral da Cóclea/metabolismo , Estria Vascular/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA