Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-35483879

RESUMO

Tuberous sclerosis complex (TSC) is an inheritable disorder characterized by the formation of benign yet disorganized tumors in multiple organ systems. Germline mutations in the TSC1 (hamartin) or more frequently TSC2 (tuberin) genes are causative for TSC. The malignant manifestations of TSC, pulmonary lymphangioleiomyomatosis (LAM) and renal angiomyolipoma (AML), may also occur as independent sporadic perivascular epithelial cell tumor (PEComa) characterized by somatic TSC2 mutations. Thus, discerning TSC from the copresentation of sporadic LAM and sporadic AML may be obscured in TSC patients lacking additional features. In this report, we present a case study on a single patient initially reported to have sporadic LAM and a mucinous duodenal adenocarcinoma deficient in DNA mismatch repair proteins. Moreover, the patient had a history of Wilms' tumor, which was reclassified as AML following the LAM diagnosis. Therefore, we investigated the origins and relatedness of these tumors. Using germline whole-genome sequencing, we identified a premature truncation in one of the patient's TSC2 alleles. Using immunohistochemistry, loss of tuberin expression was observed in AML and LAM tissue. However, no evidence of a somatic loss of heterozygosity or DNA methylation epimutations was observed at the TSC2 locus, suggesting alternate mechanisms may contribute to loss of the tumor suppressor protein. In the mucinous duodenal adenocarcinoma, no causative mutations were found in the DNA mismatch repair genes MLH1, MSH2, MSH6, or PMS2 Rather, clonal deconvolution analyses were used to identify mutations contributing to pathogenesis. This report highlights both the utility of using multiple sequencing techniques and the complexity of interpreting the data in a clinical context.


Assuntos
Adenocarcinoma , Angiomiolipoma , Neoplasias Renais , Leucemia Mieloide Aguda , Esclerose Tuberosa , Angiomiolipoma/genética , Angiomiolipoma/patologia , Feminino , Humanos , Masculino , Esclerose Tuberosa/diagnóstico , Esclerose Tuberosa/genética , Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/genética
2.
J Hum Genet ; 66(2): 161-169, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32778763

RESUMO

Gout is a complex inflammatory arthritis affecting ~20% of people with an elevated serum urate level (hyperuricemia). Gout and hyperuricemia are essentially specific to humans and other higher primates, with varied prevalence across ancestral groups. SLC2A9 and ABCG2 are major loci associated with both urate and gout in multiple ancestral groups. However, fine mapping has been challenging due to extensive linkage disequilibrium underlying the associated regions. We used trans-ancestral fine mapping integrated with primate-specific genomic information to address this challenge. Trans-ancestral meta-analyses of GWAS cohorts of either European (EUR) or East Asian (EAS) ancestry resulted in single-variant resolution mappings for SLC2A9 (rs3775948 for urate and rs4697701 for gout) and ABCG2 (rs2622621 for gout). Tests of colocalization of variants in both urate and gout suggested existence of a shared candidate causal variant for SLC2A9 only in EUR and for ABCG2 only in EAS. The fine-mapped gout variant rs4697701 was within an ancient enhancer, whereas rs2622621 was within a primate-specific transposable element, both supported by functional evidence from the Roadmap Epigenomics project in human primary tissues relevant to urate and gout. Additional primate-specific elements were found near both loci and those adjacent to SLC2A9 overlapped with known statistical epistatic interactions associated with urate as well as multiple super-enhancers identified in urate-relevant tissues. We conclude that by leveraging ancestral differences trans-ancestral fine mapping has identified ancestral and functional variants for SLC2A9 or ABCG2 with primate-specific regulatory effects on urate and gout.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Proteínas Facilitadoras de Transporte de Glucose/genética , Gota/genética , Hiperuricemia/genética , Proteínas de Neoplasias/genética , Locos de Características Quantitativas , Característica Quantitativa Herdável , Sequências Reguladoras de Ácido Nucleico , Animais , Evolução Molecular , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Gota/patologia , Humanos , Hiperuricemia/patologia , Masculino , Polimorfismo de Nucleotídeo Único , Primatas , Especificidade da Espécie , Ácido Úrico/sangue
3.
Eur J Hum Genet ; 29(3): 396-401, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32879452

RESUMO

Germline pathogenic variants in AMER1 cause osteopathia striata with cranial sclerosis (OSCS: OMIM 300373), an X-linked sclerosing bone disorder. Female heterozygotes exhibit metaphyseal striations in long bones, macrocephaly, cleft palate, and, occasionally, learning disability. Male hemizygotes typically manifest the condition as fetal or neonatal death. Somatically acquired variants in AMER1 are found in neoplastic tissue in 15-30% of patients with Wilms tumor; however, to date, only one individual with OSCS has been reported with a Wilms tumor. Here we present four cases of Wilms tumor in unrelated individuals with OSCS, including the single previously published case. We also report the first case of bilateral Wilms tumor in a patient with OSCS. Tumor tissue analysis showed no clear pattern of histological subtypes. In Beckwith-Wiedemann syndrome, which has a known predisposition to Wilms tumor development, clinical protocols have been developed for tumor surveillance. In the absence of further evidence, we propose a similar protocol for patients with OSCS to be instituted as an initial precautionary approach to tumor surveillance. Further evidence is needed to refine this protocol and to evaluate the possibility of development of other neoplasms later in life, in patients with OSCS.


Assuntos
Osteosclerose/genética , Fenótipo , Tumor de Wilms/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Pré-Escolar , Feminino , Mutação em Linhagem Germinativa , Humanos , Lactente , Osteosclerose/complicações , Osteosclerose/patologia , Proteínas Supressoras de Tumor/genética , Tumor de Wilms/etiologia , Tumor de Wilms/patologia , Adulto Jovem
4.
Genes (Basel) ; 11(12)2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33265914

RESUMO

Osteopathia striata with cranial sclerosis (OSCS) is an X-linked dominant condition characterised by metaphyseal striations, macrocephaly, cleft palate, and developmental delay in affected females. Males have a more severe phenotype with multi-organ malformations, and rarely survive. To date, only frameshift and nonsense variants in exon 2, the single coding exon of AMER1, or whole gene deletions have been reported to cause OSCS. In this study, we describe two families with phenotypic features typical of OSCS. Exome sequencing and multiplex ligation-dependent probe amplification (MLPA) did not identify pathogenic variants in AMER1. Therefore, genome sequencing was employed which identified two deletions containing the non-coding exon 1 of AMER1 in the families. These families highlight the importance of considering variants or deletions of upstream non-coding exons in conditions such as OSCS, noting that often such exons are not captured on probe or enrichment-based platforms because of their high G/C content.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Éxons/genética , Osteosclerose/genética , Proteínas Supressoras de Tumor/genética , Criança , Feminino , Deleção de Genes , Humanos , Masculino
5.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31970420

RESUMO

CONTEXT: The WNT/ß-catenin pathway is central to the pathogenesis of various human diseases including those affecting bone development and tumor progression. OBJECTIVE: To evaluate the role of a gain-of-function variant in CTNNB1 in a child with a sclerosing bone dysplasia and an adrenocortical adenoma. DESIGN: Whole exome sequencing with corroborative biochemical analyses. PATIENTS: We recruited a child with a sclerosing bone dysplasia and an adrenocortical adenoma together with her unaffected parents. INTERVENTION: Whole exome sequencing and performance of immunoblotting and luciferase-based assays to assess the cellular consequences of a de novo variant in CTNNB1. MAIN OUTCOME MEASURE(S)/RESULT: A de novo variant in CTNNB1 (c.131C>T; p.[Pro44Leu]) was identified in a patient with a sclerosing bone dysplasia and an adrenocortical adenoma. A luciferase-based transcriptional assay of WNT signaling activity verified that the activity of ß-catenin was increased in the cells transfected with a CTNNB1p.Pro44Leu construct (P = 4.00 × 10-5). The ß-catenin p.Pro44Leu variant was also associated with a decrease in phosphorylation at Ser45 and Ser33/Ser37/Thr41 in comparison to a wild-type (WT) CTNNB1 construct (P = 2.16 × 10-3, P = 9.34 × 10-8 respectively). CONCLUSION: Increased ß-catenin activity associated with a de novo gain-of-function CTNNB1 variant is associated with osteosclerotic phenotype and adrenocortical neoplasia.


Assuntos
Neoplasias do Córtex Suprarrenal/patologia , Carcinoma Adrenocortical/patologia , Doenças do Desenvolvimento Ósseo/patologia , Mutação , beta Catenina/genética , Neoplasias do Córtex Suprarrenal/genética , Carcinoma Adrenocortical/genética , Doenças do Desenvolvimento Ósseo/genética , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Fenótipo , Prognóstico , Sequenciamento do Exoma
6.
Eur J Hum Genet ; 28(4): 445-452, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31792352

RESUMO

Connective tissue disorders are a spectrum of diseases that affect the integrity of tissues including skin, vasculature, and joints. They are often caused by variants that disrupt genes encoding components of extracellular matrix (ECM). The fibulin glycoproteins are ECM proteins important for integrity of tissues including dermis, retina, fascia, and vasculature. The fibulin family consists of seven members (fibulins-1 to -7) and is defined by a fibulin-type domain at the C-terminus. The family is associated with human diseases, for instance a variant in FBLN1, encoding fibulin-1, is associated with synpolydactyly, while one in EFEMP1, encoding fibulin-3, causes Doyne honeycomb degeneration of the retina. Loss-of-function of fibulins-4 and -5 causes cutis laxa, while variants in fibulins-5 and -6 are associated with age-related macular degeneration. Of note, EFEMP1 is not currently associated with any connective tissue disorder. Here we show biallelic loss-of-function variants in EFEMP1 in an individual with multiple and recurrent abdominal and thoracic herniae, myopia, hypermobile joints, scoliosis, and thin translucent skin. Fibroblasts from this individual express significantly lower EFEMP1 transcript than age-matched control cells. A skin biopsy, visualised using light microscopy, showed normal structure and abundance of elastic fibres. The phenotype of this individual is remarkably similar to the Efemp1 knockout mouse model that displays multiple herniae with premature aging and scoliosis. We conclude that loss of EFEMP1 function in this individual is the cause of a connective tissue disorder with a novel combination of phenotypic features, and can perhaps explain similar, previously reported cases in the literature.


Assuntos
Doenças do Tecido Conjuntivo/genética , Proteínas da Matriz Extracelular/genética , Mutação com Perda de Função , Fenótipo , Adulto , Alelos , Células Cultivadas , Doenças do Tecido Conjuntivo/patologia , Proteínas da Matriz Extracelular/metabolismo , Humanos , Masculino
7.
PLoS Genet ; 14(6): e1007399, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29912901

RESUMO

Wilms tumour is a childhood tumour that arises as a consequence of somatic and rare germline mutations, the characterisation of which has refined our understanding of nephrogenesis and carcinogenesis. Here we report that germline loss of function mutations in TRIM28 predispose children to Wilms tumour. Loss of function of this transcriptional co-repressor, which has a role in nephrogenesis, has not previously been associated with cancer. Inactivation of TRIM28, either germline or somatic, occurred through inactivating mutations, loss of heterozygosity or epigenetic silencing. TRIM28-mutated tumours had a monomorphic epithelial histology that is uncommon for Wilms tumour. Critically, these tumours were negative for TRIM28 immunohistochemical staining whereas the epithelial component in normal tissue and other Wilms tumours stained positively. These data, together with a characteristic gene expression profile, suggest that inactivation of TRIM28 provides the molecular basis for defining a previously described subtype of Wilms tumour, that has early age of onset and excellent prognosis.


Assuntos
Mutação em Linhagem Germinativa , Neoplasias Renais/genética , Mutação com Perda de Função , Recidiva Local de Neoplasia/genética , Proteína 28 com Motivo Tripartido/genética , Tumor de Wilms/genética , Adulto , Biomarcadores Tumorais/genética , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Humanos , Rim/patologia , Neoplasias Renais/epidemiologia , Neoplasias Renais/patologia , Masculino , Recidiva Local de Neoplasia/epidemiologia , Recidiva Local de Neoplasia/patologia , Prognóstico , Urotélio/patologia , Sequenciamento do Exoma , Tumor de Wilms/epidemiologia , Tumor de Wilms/patologia , Adulto Jovem
8.
Am J Hum Genet ; 102(6): 1115-1125, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29805041

RESUMO

Spondylocarpotarsal synostosis syndrome (SCTS) is characterized by intervertebral fusions and fusion of the carpal and tarsal bones. Biallelic mutations in FLNB cause this condition in some families, whereas monoallelic variants in MYH3, encoding embryonic heavy chain myosin 3, have been implicated in dominantly inherited forms of the disorder. Here, five individuals without FLNB mutations from three families were hypothesized to be affected by recessive SCTS on account of sibling recurrence of the phenotype. Initial whole-exome sequencing (WES) showed that all five were heterozygous for one of two independent splice-site variants in MYH3. Despite evidence indicating that three of the five individuals shared two allelic haplotypes encompassing MYH3, no second variant could be located in the WES datasets. Subsequent genome sequencing of these three individuals demonstrated a variant altering a 5' UTR splice donor site (rs557849165 in MYH3) not represented by exome-capture platforms. When the cohort was expanded to 16 SCTS-affected individuals without FLNB mutations, nine had truncating mutations transmitted by unaffected parents, and six inherited the rs557849165 variant in trans, an observation at odds with the population allele frequency for this variant. The rs557849165 variant disrupts splicing in the 5' UTR but is still permissive of MYH3 translational initiation, albeit with reduced efficiency. Although some MYH3 variants cause dominant SCTS, these data indicate that others (notably truncating variants) do not, except in the context of compound heterozygosity for a second hypomorphic allele. These observations make genetic diagnosis challenging in the context of simplex presentations of the disorder.


Assuntos
Anormalidades Múltiplas/genética , Genes Recessivos , Vértebras Lombares/anormalidades , Doenças Musculoesqueléticas/genética , Mutação/genética , Cadeias Pesadas de Miosina/genética , Escoliose/congênito , Sinostose/genética , Vértebras Torácicas/anormalidades , Alelos , Mapeamento Cromossômico , Feminino , Filaminas/genética , Haplótipos/genética , Heterozigoto , Humanos , Masculino , Linhagem , Fenótipo , Splicing de RNA/genética , Escoliose/genética , Síndrome , Sequenciamento do Exoma
9.
J Clin Invest ; 127(11): 3923-3936, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28945198

RESUMO

Netrin-1 is a secreted protein that was first identified 20 years ago as an axon guidance molecule that regulates midline crossing in the CNS. It plays critical roles in various tissues throughout development and is implicated in tumorigenesis and inflammation in adulthood. Despite extensive studies, no inherited human disease has been directly associated with mutations in NTN1, the gene coding for netrin-1. Here, we have identified 3 mutations in exon 7 of NTN1 in 2 unrelated families and 1 sporadic case with isolated congenital mirror movements (CMM), a disorder characterized by involuntary movements of one hand that mirror intentional movements of the opposite hand. Given the diverse roles of netrin-1, the absence of manifestations other than CMM in NTN1 mutation carriers was unexpected. Using multimodal approaches, we discovered that the anatomy of the corticospinal tract (CST) is abnormal in patients with NTN1-mutant CMM. When expressed in HEK293 or stable HeLa cells, the 3 mutated netrin-1 proteins were almost exclusively detected in the intracellular compartment, contrary to WT netrin-1, which is detected in both intracellular and extracellular compartments. Since netrin-1 is a diffusible extracellular cue, the pathophysiology likely involves its loss of function and subsequent disruption of axon guidance, resulting in abnormal decussation of the CST.


Assuntos
Transtornos dos Movimentos/genética , Netrina-1/genética , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Sequência Conservada , Feminino , Frequência do Gene , Estudos de Associação Genética , Células HEK293 , Células HeLa , Heterozigoto , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto , Linhagem , Deleção de Sequência
10.
Nat Commun ; 8: 16077, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28681861

RESUMO

Multinucleate cellular syncytial formation is a hallmark of skeletal muscle differentiation. Myomaker, encoded by Mymk (Tmem8c), is a well-conserved plasma membrane protein required for myoblast fusion to form multinucleated myotubes in mouse, chick, and zebrafish. Here, we report that autosomal recessive mutations in MYMK (OMIM 615345) cause Carey-Fineman-Ziter syndrome in humans (CFZS; OMIM 254940) by reducing but not eliminating MYMK function. We characterize MYMK-CFZS as a congenital myopathy with marked facial weakness and additional clinical and pathologic features that distinguish it from other congenital neuromuscular syndromes. We show that a heterologous cell fusion assay in vitro and allelic complementation experiments in mymk knockdown and mymkinsT/insT zebrafish in vivo can differentiate between MYMK wild type, hypomorphic and null alleles. Collectively, these data establish that MYMK activity is necessary for normal muscle development and maintenance in humans, and expand the spectrum of congenital myopathies to include cell-cell fusion deficits.


Assuntos
Proteínas de Membrana/genética , Síndrome de Möbius/genética , Morfogênese/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Mutação , Mioblastos/metabolismo , Síndrome de Pierre Robin/genética , Proteínas de Peixe-Zebra/genética , Adulto , Sequência de Aminoácidos , Animais , Fusão Celular , Criança , Modelos Animais de Doenças , Embrião não Mamífero , Feminino , Expressão Gênica , Genes Recessivos , Teste de Complementação Genética , Humanos , Lactente , Masculino , Proteínas de Membrana/deficiência , Síndrome de Möbius/metabolismo , Síndrome de Möbius/patologia , Proteínas Musculares/deficiência , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Mioblastos/patologia , Linhagem , Síndrome de Pierre Robin/metabolismo , Síndrome de Pierre Robin/patologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência
11.
Am J Med Genet A ; 173(7): 1739-1746, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28498505

RESUMO

Frontometaphyseal dysplasia (FMD) is caused by gain-of-function mutations in the X-linked gene FLNA in approximately 50% of patients. Recently we characterized an autosomal dominant form of FMD (AD-FMD) caused by mutations in MAP3K7, which accounts for the condition in the majority of patients who lack a FLNA mutation. We previously also described a patient with a de novo variant in TAB2, which we hypothesized was causative of another form of AD-FMD. In this study, a cohort of 20 individuals with AD-FMD is clinically evaluated. This cohort consists of 15 individuals with the recently described, recurrent mutation (c.1454C>T) in MAP3K7, as well as three individuals with missense mutations that result in substitutions in the N-terminal kinase domain of TGFß-activated kinase 1 (TAK1), encoded by MAP3K7. Additionally, two individuals have missense variants in the gene TAB2, which encodes a protein with a close functional relationship to TAK1, TAK1-associated binding protein 2 (TAB2). Although the X-linked and autosomal dominant forms of FMD are very similar, there are distinctions to be made between the two conditions. Individuals with AD-FMD have characteristic facial features, and are more likely to be deaf, have scoliosis and cervical fusions, and have a cleft palate. Furthermore, there are features only found in AD-FMD in our review of the literature including valgus deformity of the feet and predisposition to keloid scarring. Finally, intellectual disability is present in a small number of subjects with AD-FMD but has not been described in association with X-linked FMD.

12.
Am J Hum Genet ; 99(2): 392-406, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27426733

RESUMO

Frontometaphyseal dysplasia (FMD) is a progressive sclerosing skeletal dysplasia affecting the long bones and skull. The cause of FMD in some individuals is gain-of-function mutations in FLNA, although how these mutations result in a hyperostotic phenotype remains unknown. Approximately one half of individuals with FMD have no identified mutation in FLNA and are phenotypically very similar to individuals with FLNA mutations, except for an increased tendency to form keloid scars. Using whole-exome sequencing and targeted Sanger sequencing in 19 FMD-affected individuals with no identifiable FLNA mutation, we identified mutations in two genes-MAP3K7, encoding transforming growth factor ß (TGF-ß)-activated kinase (TAK1), and TAB2, encoding TAK1-associated binding protein 2 (TAB2). Four mutations were found in MAP3K7, including one highly recurrent (n = 15) de novo mutation (c.1454C>T [ p.Pro485Leu]) proximal to the coiled-coil domain of TAK1 and three missense mutations affecting the kinase domain (c.208G>C [p.Glu70Gln], c.299T>A [p.Val100Glu], and c.502G>C [p.Gly168Arg]). Notably, the subjects with the latter three mutations had a milder FMD phenotype. An additional de novo mutation was found in TAB2 (c.1705G>A, p.Glu569Lys). The recurrent mutation does not destabilize TAK1, or impair its ability to homodimerize or bind TAB2, but it does increase TAK1 autophosphorylation and alter the activity of more than one signaling pathway regulated by the TAK1 kinase complex. These findings show that dysregulation of the TAK1 complex produces a close phenocopy of FMD caused by FLNA mutations. Furthermore, they suggest that the pathogenesis of some of the filaminopathies caused by FLNA mutations might be mediated by misregulation of signaling coordinated through the TAK1 signaling complex.


Assuntos
Testa/anormalidades , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Mutação/genética , Osteocondrodisplasias/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Feminino , Filaminas/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , NF-kappa B/metabolismo , Osteocondrodisplasias/metabolismo , Fosforilação , Ligação Proteica , Multimerização Proteica
13.
Am J Hum Genet ; 97(6): 837-47, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26637977

RESUMO

The periosteum contributes to bone repair and maintenance of cortical bone mass. In contrast to the understanding of bone development within the epiphyseal growth plate, factors that regulate periosteal osteogenesis have not been studied as intensively. Osteofibrous dysplasia (OFD) is a congenital disorder of osteogenesis and is typically sporadic and characterized by radiolucent lesions affecting the cortical bone immediately under the periosteum of the tibia and fibula. We identified germline mutations in MET, encoding a receptor tyrosine kinase, that segregate with an autosomal-dominant form of OFD in three families and a mutation in a fourth affected subject from a simplex family and with bilateral disease. Mutations identified in all families with dominant inheritance and in the one simplex subject with bilateral disease abolished the splice inclusion of exon 14 in MET transcripts, which resulted in a MET receptor (MET(Δ14)) lacking a cytoplasmic juxtamembrane domain. Splice exclusion of this domain occurs during normal embryonic development, and forced induction of this exon-exclusion event retarded osteoblastic differentiation in vitro and inhibited bone-matrix mineralization. In an additional subject with unilateral OFD, we identified a somatic MET mutation, also affecting exon 14, that substituted a tyrosine residue critical for MET receptor turnover and, as in the case of the MET(Δ14) mutations, had a stabilizing effect on the mature protein. Taken together, these data show that aberrant MET regulation via the juxtamembrane domain subverts core MET receptor functions that regulate osteogenesis within cortical diaphyseal bone.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Éxons , Mutação em Linhagem Germinativa , Osteogênese/genética , Periósteo/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Adulto , Sequência de Bases , Doenças do Desenvolvimento Ósseo/metabolismo , Doenças do Desenvolvimento Ósseo/patologia , Diferenciação Celular , Criança , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Dominantes , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Osteoblastos/metabolismo , Osteoblastos/patologia , Linhagem , Periósteo/crescimento & desenvolvimento , Periósteo/patologia , Cultura Primária de Células , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-met/metabolismo , Splicing de RNA
14.
J Neurol Sci ; 351(1-2): 140-145, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25813273

RESUMO

Congenital mirror movements (CMM) is a disorder characterized by unintentional mirroring in homologous motor systems of voluntary movements on the opposite side, usually affecting the distal upper extremities. Genetic analyses have revealed involvement of three genes (DCC, RAD51, and DNAL4). We sought to distinguish whether different phenotypes of CMM exist, and if so, whether they might map to different causative genes. We studied 14 individuals across five families with dominantly-inherited CMM. We used accelerometer gloves to analyse the fine detail of index finger tapping movements, and applied standard genetic methodology to analyse DNA samples. Two forms of mirroring were distinguished: 'actual' in which the mirroring followed precisely the movements of the voluntary hand, and 'fractionated' in which the mirroring was saccadic. We found that actual mirroring was characteristic of individuals in a family with a RAD51 mutation, and fractionated more characteristic of a family with a DCC mutation. These findings are suggestive of specific genotype-phenotype correlations in CMM. Three heterozygous individuals (one RAD51; two DCC) showed no apparent mirroring on visual inspection, although mirroring was detectable with the accelerometer gloves. Thus, subclinical mirroring may be present even when undetectable on clinical observation.


Assuntos
Transtornos dos Movimentos/genética , Rad51 Recombinase/genética , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Idoso , Dineínas do Axonema/genética , Receptor DCC , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Adulto Jovem
15.
Nat Genet ; 45(11): 1300-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24056717

RESUMO

The regulated proliferation and differentiation of neural stem cells before the generation and migration of neurons in the cerebral cortex are central aspects of mammalian development. Periventricular neuronal heterotopia, a specific form of mislocalization of cortical neurons, can arise from neuronal progenitors that fail to negotiate aspects of these developmental processes. Here we show that mutations in genes encoding the receptor-ligand cadherin pair DCHS1 and FAT4 lead to a recessive syndrome in humans that includes periventricular neuronal heterotopia. Reducing the expression of Dchs1 or Fat4 within mouse embryonic neuroepithelium increased progenitor cell numbers and reduced their differentiation into neurons, resulting in the heterotopic accumulation of cells below the neuronal layers in the neocortex, reminiscent of the human phenotype. These effects were countered by concurrent knockdown of Yap, a transcriptional effector of the Hippo signaling pathway. These findings implicate Dchs1 and Fat4 upstream of Yap as key regulators of mammalian neurogenesis.


Assuntos
Caderinas/genética , Córtex Cerebral/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Proteínas Supressoras de Tumor/genética , Anormalidades Múltiplas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas Relacionadas a Caderinas , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Anormalidades Craniofaciais/genética , Deformidades Congênitas do Pé/genética , Técnicas de Silenciamento de Genes , Deformidades Congênitas da Mão/genética , Humanos , Deficiência Intelectual/genética , Instabilidade Articular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Heterotopia Nodular Periventricular/genética , Fosfoproteínas/genética , Análise de Sequência de DNA , Transdução de Sinais/genética , Proteínas de Sinalização YAP
17.
Mol Vis ; 15: 1700-8, 2009 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-19710953

RESUMO

PURPOSE: With advances in phenotyping tools and availability of molecular characterization, an increasing number of phenotypically and genotypically diverse inherited corneal dystrophies are described. We aimed to determine the underlying causative genetic mechanism in a three-generation pedigree affected with a unique anterior membrane corneal dystrophy characterized by early onset recurrent corneal erosions, small discrete focal opacities at the level of Bowman layer and anterior stroma, anterior stromal flecks, and prominent corneal nerves. METHODS: Twenty affected and unaffected members of a three-generation family were examined and extensively clinically characterized including corneal topography and in vivo confocal microscopy, and biological specimens were collected for DNA extraction. Mutational analysis of two corneal genes (TGFBI [Transforming Growth factor-beta induced] and ZEB1 [zinc finger E box-binding homeobox 1]) was undertaken, in addition to testing with the Asper Corneal Dystrophy gene chip (Asper Ophthalmics, Tartu, Estonia). Subsequent Genotyping To 11 Known Corneal Gene Loci (COL8A2 [Collagen, Type VIII, Alpha-2], TACSTD2 [Tumor-Associated Calcium Signal Transducer 2], PIP5K3 [Phosphatidylinositol-3-Phosphate 5-Kinase, Type III], GSN [Gelsolin], KERA [Keratocan], VSX1 [Visual System Homeobox Gene 1], COL6A1 [Collagen, Type VI, Alpha-1], MMP9 [Matrix Metalloproteinase 9], KRT3 [Keratin 3]), and two putative loci, 3p14-q13 and 15q22.33-24) was undertaken using polymorphic markers, and haplotypes constructed. Multipoint linkage analysis was performed to generate LOD scores and produce LOD plots across the candidate intervals. RESULTS: No pathogenic sequence variations were detected in TGFBI or ZEB1 of the proband nor on the Asper Corneal Dystrophy gene chip (302 mutations in 12 genes). Multipoint linkage analysis of 11 known corneal genes and loci generated negative LOD plots and was able to exclude all genes tested including PIP5K3. CONCLUSIONS: Exclusion of linkage to candidate corneal loci combined with an absence of pathogenic mutations in known corneal genes in this pedigree suggest a different genetic causative mechanism in this dystrophy than the previously documented corneal genes. This unique phenotype of an anterior membrane dystrophy may therefore provide an opportunity to identify a new gene responsible for corneal disease.


Assuntos
Distrofias Hereditárias da Córnea/genética , Distrofias Hereditárias da Córnea/patologia , Genes Dominantes , Linhagem , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Córnea/patologia , Demografia , Feminino , Humanos , Escore Lod , Masculino , Membranas/patologia , Microscopia Confocal , Pessoa de Meia-Idade
18.
Am J Med Genet A ; 146A(18): 2376-84, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18698629

RESUMO

Spondyloepiphyseal dysplasia (SED), Omani type (OMIM 608637) is a recessively inherited skeletal dysplasia previously described in two distantly related families from the Republic of Oman. The phenotype consists of short stature, severe kyphoscoliosis, arthritic joints (elbows, wrists, knees), secondary large joint dislocations, rhizomelia, fusion of carpal bones and mild brachydactyly. Affected individuals were homozygous for a missense mutation, R304Q in CHST3 that encodes the enzyme chondroitin 6-O-sulfotransferase-1 (C6ST-1). This enzyme mediates the sulfation of proteoglycans, particularly chondroitin sulfate (CS), in the extracellular matrix of cartilage. Here we describe the identification of a mutation (857T > C predicting the substitution L286P) in CHST3 in a Turkish family and extend the clinical phenotype of SED-Omani type to include congenital joint dislocation, club feet, ventricular septal defect, deafness, metacarpal shortening and accessory carpal ossification centers. Fibroblasts and urine obtained from affected patients demonstrated negligible levels of 6-O-sulfated GalNAc residue in CS. Furthermore, the 6-O-sulfotransferase activity of cloned C6ST-1 into which the L286P mutation had been introduced was dramatically reduced, confirming the pathogenicity of this substitution. These results indicate that the clinical consequences of a deficiency of 6-O-sulfation in CS can be varied and that a clinical spectrum may exist similar to that seen in other skeletal dysplasias characterized by disorders of proteoglycan sulfation.


Assuntos
Osteocondrodisplasias/genética , Mutação Puntual , Sulfotransferases/genética , Substituição de Aminoácidos , Criança , Pré-Escolar , Clonagem Molecular , Consanguinidade , Dissacarídeos/urina , Feminino , Fibroblastos/enzimologia , Humanos , Omã , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/enzimologia , Linhagem , Fenótipo , Radiografia , Coluna Vertebral/anormalidades , Coluna Vertebral/diagnóstico por imagem , Sulfotransferases/urina , Carboidrato Sulfotransferases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA