Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diagn Cytopathol ; 50(10): E275-E279, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35716104

RESUMO

Multifocality in papillary thyroid carcinoma (PTC) is a common finding, but the clonal relationship between individual tumors remains uncertain. While multiple synchronous tumor foci of PTC may develop through permeation of intraglandular lymph vessels of a single malignant clone, they can also arise from independent progenitor clones sustained by different genetic events. We report the case of a 37-year-old man who underwent total thyroidectomy after fine-needle aspiration of two bilateral thyroid nodules that yielded cytological findings consistent with atypia of undetermined significance/follicular lesion of undetermined significance. By next-generation sequencing of a large panel of thyroid carcinoma related genes, we found that the larger tumor harbored a mutation of the NRAS gene, while the contralateral tumor harbored a different mutation in the HRAS gene. Final pathology of the surgical specimen showed two encapsulated follicular variant papillary thyroid carcinomas of 16 and 6 mm in the right and the left lobes, respectively. To the best of our knowledge, this is the fourth case of multifocal PTC showing different HRAS and NRAS mutations, and highlights that mutational heterogeneity is also present in non-BRAF, non-RET genes, supporting the hypothesis that independent progenitor clones may explain multifocality in papillary thyroid carcinoma.


Assuntos
Adenocarcinoma Folicular , Carcinoma Papilar , Neoplasias da Glândula Tireoide , Adenocarcinoma Folicular/patologia , Adulto , Carcinoma Papilar/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Câncer Papilífero da Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
2.
Hum Mutat ; 38(11): 1471-1476, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28730625

RESUMO

Craniosynostosis is commonly caused by mutations in fibroblast growth factor receptors (FGFRs), highlighting the essential role of FGF-mediated signaling in skeletal development. We set out to identify the molecular defect in a family referred for craniosynostosis and in whom no mutation was previously detected. Using next-generation sequencing, we identified a novel missense mutation in FGF9. Modeling based upon the crystal structure and functional studies confirmed its pathogenicity showing that it impaired homodimerization and FGFR3 binding. Only one FGF9 mutation has been previously reported in a multigeneration family with multiple synostoses (SYNS3) but no signs of craniosynostosis. In contrast, our family has a greater phenotypic resemblance to that observed in the Fgf9 spontaneous mouse mutant, elbow-knee-synostosis, Eks, with both multiple synostoses and craniosynostosis. We have demonstrated for the first time that mutations in FGF9 cause craniosynostosis in humans and confirm that FGF9 mutations cause multiple synostoses.


Assuntos
Craniossinostoses/diagnóstico , Craniossinostoses/genética , Fator 9 de Crescimento de Fibroblastos/genética , Mutação , Fenótipo , Sinostose/diagnóstico , Sinostose/genética , Substituição de Aminoácidos , Fator 9 de Crescimento de Fibroblastos/química , Estudos de Associação Genética , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Masculino , Modelos Moleculares , Linhagem , Conformação Proteica , Multimerização Proteica , Radiografia , Transdução de Sinais , Relação Estrutura-Atividade
3.
Eur J Med Genet ; 59(8): 363-6, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27282802

RESUMO

Schimke immunoosseous dysplasia (SIOD) is an autosomal recessive disease characterized by skeletal dysplasia, focal segmental glomerulosclerosis, renal failure and immunodeficiency. In this work, we report the molecular studies undertaken in a severely affected SIOD patient that died at six years old due to nephropathy. The patient was screened for mutations using a targeted skeletal dysplasias panel. A homozygous novel missense mutation was identified, c.1615C > G (p.[Leu539Val]) that was predicted as mildly pathogenic by in silico pathogenicity prediction tools. However, splicing prediction software suggested that this variant may create a new splicing donor site in exon 9, which was subsequently confirmed using a minigene assay in HEK293 cells. Thus, the splicing alteration, c.1615C > G; r.1615c > g, 1615_1644del; (p.[Leu539_Ile548del]), results in the loss of 10 amino acids of the HARP-ATPase catalytic domain and the RPA-binding domain. Several studies have demonstrated a weak genotype-phenotype correlation among such patients. Thus, the molecular characterization has helped us to understand why a predicted weakly pathogenic missense mutation results in severe SIOD and should be considered in similar scenarios.


Assuntos
Arteriosclerose/diagnóstico , Arteriosclerose/genética , DNA Helicases/genética , Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/genética , Mutação de Sentido Incorreto , Síndrome Nefrótica/diagnóstico , Síndrome Nefrótica/genética , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/genética , Splicing de RNA , Sequência de Bases , Análise Mutacional de DNA , Ordem dos Genes , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Masculino , Fenótipo , Doenças da Imunodeficiência Primária , Sítios de Splice de RNA , Índice de Gravidade de Doença
4.
Am J Med Genet A ; 170A(1): 210-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26374189

RESUMO

Primordial dwarfism encompasses rare conditions characterized by severe intrauterine growth retardation and growth deficiency throughout life. Recently, three POC1A mutations have been reported in six families with the primordial dwarfism, SOFT syndrome (Short stature, Onychodysplasia, Facial dysmorphism, and hypoTrichosis). Using a custom-designed Next-generation sequencing skeletal dysplasia panel, we have identified two novel homozygous POC1A mutations in two individuals with primordial dwarfism. The severe growth retardation and the facial profiles are strikingly similar between our patients and those described previously. However, one of our patients was diagnosed with severe foramen magnum stenosis and subglottic tracheal stenosis, malformations not previously associated with this syndrome. Our findings confirm that POC1A mutations cause SOFT syndrome and that mutations in this gene should be considered in patients with severe pre- and postnatal short stature, symmetric shortening of long bones, triangular facies, sparse hair and short, thickened distal phalanges.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Craniofaciais/genética , Nanismo/genética , Hipotricose/genética , Atrofia Muscular/genética , Doenças da Unha/genética , Osteocondrodisplasias/genética , Proteínas/genética , Adolescente , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Humanos , Lactente , Masculino , Doenças da Unha/congênito , Tórax/anormalidades
5.
J Clin Endocrinol Metab ; 100(8): E1133-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26075495

RESUMO

CONTEXT: SHOX mutations have been detected in approximately 70% of Léri-Weill dyschondrosteosis (LWD) and approximately 2.5% of idiopathic short stature (ISS) cases, suggesting the implication of other genes or loci. The recent identification of NPR2 mutations in ISS suggested that NPR2 mutations may also be involved in disproportionate short stature. OBJECTIVE: The objective of the study was to investigate whether NPR2 mutations can account for a proportion of the cases referred for LWD and ISS in whom no SHOX mutation was detected. PATIENTS AND METHODS: We undertook NPR2 mutation screening in 173 individuals referred for suspected LWD and 95 for ISS, with no known defect in SHOX or its enhancers. Intracellular localization and natriuretic peptide precursor C-dependent guanylate cyclase activity were determined for the identified NPR2 variants. RESULTS: Eight NPR2 variants were identified in nine individuals, seven referred for suspected LWD and two for ISS. Six were demonstrated to affect NPR-B cell trafficking and/or its ability to synthesize cyclic GMP (cGMP) under response to natriuretic peptide precursor C/brain natriuretic peptide stimulation. All pathogenic mutations were detected in the suspected LWD referral group (∼3%). Interestingly, one of these patients is currently being treated with recombinant human GH and in contrast to previous reports is showing a positive response to the treatment. CONCLUSIONS: NPR2 mutations account for approximately 3% of patients with disproportionate short stature and/or clinical or radiographic indicators of SHOX deficiency and in whom no SHOX defect has been identified. However, no patient has yet presented with Madelung deformity. Thus, NPR2 should be screened in the SHOX-negative LWD referrals.


Assuntos
Nanismo/genética , Transtornos do Crescimento/genética , Mutação de Sentido Incorreto , Osteocondrodisplasias/genética , Receptores do Fator Natriurético Atrial/genética , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Estudos de Coortes , Nanismo/epidemiologia , Feminino , Frequência do Gene , Transtornos do Crescimento/epidemiologia , Heterozigoto , Humanos , Masculino , Osteocondrodisplasias/epidemiologia , Células Tumorais Cultivadas
6.
PLoS One ; 9(1): e83104, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24421874

RESUMO

SHOX and SHOX2 transcription factors are highly homologous, with even identical homeodomains. Genetic alterations in SHOX result in two skeletal dysplasias; Léri-Weill dyschondrosteosis (LWD) and Langer mesomelic dysplasia (LMD), while no human genetic disease has been linked to date with SHOX2. SHOX2 is, though, involved in skeletal development, as shown by different knockout mice models. Due to the high homology between SHOX and SHOX2, and their functional redundancy during heart development, we postulated that SHOX2 might have the same transcriptional targets and cofactors as SHOX in limb development. We selected two SHOX transcription targets regulated by different mechanisms: 1) the natriuretic peptide precursor B gene (NPPB) involved in the endochondral ossification signalling and directly activated by SHOX; and 2) Aggrecan (ACAN), a major component of cartilage extracellular matrix, regulated by the cooperation of SHOX with the SOX trio (SOX5, SOX6 and SOX9) via the protein interaction between SOX5/SOX6 and SHOX. Using the luciferase assay we have demonstrated that SHOX2, like SHOX, regulates NPPB directly whilst activates ACAN via its cooperation with the SOX trio. Subsequently, we have identified and characterized the protein domains implicated in the SHOX2 dimerization and also its protein interaction with SOX5/SOX6 and SHOX using the yeast-two hybrid and co-immunoprecipitation assays. Immunohistochemistry of human fetal growth plates from different time points demonstrated that SHOX2 is coexpressed with SHOX and the members of the SOX trio. Despite these findings, no mutation was identified in SHOX2 in a cohort of 83 LWD patients with no known molecular defect, suggesting that SHOX2 alterations do not cause LWD. In conclusion, our work has identified the first cofactors and two new transcription targets of SHOX2 in limb development, and we hypothesize a time- and tissue-specific functional redundancy between SHOX and SHOX2.


Assuntos
Agrecanas/genética , Fator Natriurético Atrial/genética , Desenvolvimento Ósseo , Proteínas de Homeodomínio/metabolismo , Peptídeo Natriurético Encefálico/genética , Transcrição Gênica , Agrecanas/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Desenvolvimento Ósseo/genética , Linhagem Celular , Estudos de Coortes , Transtornos do Crescimento/genética , Lâmina de Crescimento/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Mutação/genética , Peptídeo Natriurético Encefálico/metabolismo , Osteocondrodisplasias/genética , Fenótipo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Fatores de Transcrição SOX/metabolismo , Ativação Transcricional/genética
7.
Hum Mol Genet ; 20(8): 1547-59, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21262861

RESUMO

SHOX (short stature homeobox-containing gene) encodes a transcription factor implicated in skeletal development. SHOX haploinsufficiency has been demonstrated in Leri-Weill dyschondrosteosis (LWD), a skeletal dysplasia associated with disproportionate short stature, as well as in a variable proportion of cases with idiopathic short stature (ISS). In order to gain insight into the SHOX signalling pathways, we performed a yeast two-hybrid screen to identify SHOX-interacting proteins. Two transcription factors, SOX5 and SOX6, were identified. Co-immunoprecipitation assays confirmed the existence of the SHOX-SOX5 and SHOX-SOX6 interactions in human cells, whereas immunohistochemical studies demonstrated the coexpression of these proteins in 18- and 32-week human fetal growth plates. The SHOX homeodomain and the SOX6 HMG domain were shown to be implicated in the SHOX-SOX6 interaction. Moreover, different SHOX missense mutations, identified in LWD and ISS patients, disrupted this interaction. The physiological importance of these interactions was investigated by studying the effect of SHOX on a transcriptional target of the SOX trio, Agc1, which encodes one of the main components of cartilage, aggrecan. Our results show that SHOX cooperates with SOX5/SOX6 and SOX9 in the activation of the upstream Agc1 enhancer and that SHOX mutations affect this activation. In conclusion, we have identified SOX5 and SOX6 as the first two SHOX-interacting proteins and have shown that this interaction regulates aggrecan expression, an essential factor in chondrogenesis and skeletal development.


Assuntos
Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição SOXD/metabolismo , Agrecanas/genética , Animais , Condrogênese/genética , Desenvolvimento Fetal/genética , Genes Reporter , Transtornos do Crescimento/genética , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/metabolismo , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Imunoprecipitação , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Camundongos , Complexos Multiproteicos/metabolismo , Mutação de Sentido Incorreto , Osteocondrodisplasias/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Proteína de Homoeobox de Baixa Estatura , Técnicas do Sistema de Duplo-Híbrido
8.
Am J Med Genet A ; 152A(6): 1390-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20503313

RESUMO

Beckwith-Wiedemann syndrome (BWS) is an overgrowth syndrome characterized by macroglossia, macrosomia, and abdominal wall defects. It is a multigenic disorder caused in most patients by alterations in growth regulatory genes. A small number of individuals with BWS (5-10%) have mutations in CDKN1C, a cyclin-dependent kinase inhibitor of G1 cyclin complexes that functions as a negative regulator of cellular growth and proliferation. Here, we report on eight patients with BWS and CDKN1C mutations and review previous reported cases. We analyzed 72 patients (50 BWS, 17 with isolated hemihyperplasia (IH), three with omphalocele, and two with macroglossia) for CDKN1C defects with the aim to search for new mutations and to define genotype-phenotype correlations. Our findings suggest that BWS patients with CDKN1C mutations have a different pattern of clinical malformations than those with other molecular defects. Polydactyly, genital abnormalities, extra nipple, and cleft palate are more frequently observed in BWS with mutations in CDKN1C. The clinical observation of these malformations may help to decide which genetic characterization should be undertaken (i.e., CDKN1C screening), thus optimizing the laboratory evaluation for BWS.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Adulto , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p57/química , Feminino , Genótipo , Humanos , Lactente , Masculino , Mutação , Fenótipo , Polimorfismo Genético , Conformação Proteica
9.
Eur J Med Genet ; 53(4): 204-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20412871

RESUMO

Léri-Weill dyschondrosteosis (LWD, MIM 127300), is a dominantly inherited skeletal dysplasia with disproportionate short stature, mesomelic limb shortening, and the characteristic Madelung deformity. Two regions of the pseudoautosomal region 1 (PAR1) have been shown to be involved in LWD, SHOX (short-stature homeobox-containing gene) and the downstream enhancer region. We report our genetic findings of a young girl clinically diagnosed with LWD. We analyzed the proband and her family using MLPA and microsatellite analysis. We identified a deletion, 726-866 kb in size, of the downstream SHOX enhancer region in the proband. Neither parent carried the deletion. Microsatellite analysis showed that the deleted allele was of paternal origin. The mutation is more likely to have arisen from a de novo event but paternal gonadal mosaicism cannot be excluded. In conclusion, we report the clinical and molecular details of the first case of a de novo deletion of the downstream PAR1 region in an LWD individual. De novo deletions of SHOX and the downstream enhancer region must be therefore considered in cases of isolated LWD.


Assuntos
Proteínas de Homeodomínio/genética , Osteocondrodisplasias/genética , Deleção de Sequência/genética , Criança , Nanismo/diagnóstico , Nanismo/genética , Feminino , Humanos , Masculino , Repetições de Microssatélites/genética , Mutação/genética , Osteocondrodisplasias/diagnóstico , Linhagem , Proteína de Homoeobox de Baixa Estatura , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...