Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Endocrinol Metab ; 90(2): 864-70, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15572418

RESUMO

Familial benign hypocalciuric hypercalcemia (FBHH), in which calcium homeostasis is disordered, can be distinguished from mild primary hyperparathyroidism by the finding of a heterozygous loss-of-function mutation in the calcium-sensing receptor (CaSR). Here, we report a Polish kindred with FBHH, the proband of which had undergone an unsuccessful parathyroidectomy. Direct sequence analysis of exon 4 of her CASR gene identified a heterozygous R227Q mutation in the extracellular domain of the receptor. This mutation segregated with other affected family members. A de novo heterozygous R227L mutation had previously been identified in a case of neonatal hyperparathyroidism. We performed a functional analysis by transiently transfecting wild-type and mutant (R227Q, R227L) CaSRs in human embryonic kidney (HEK293) cells. Both mutant receptors were expressed at a similar level to that of the wild-type, demonstrated a 160-kDa molecular species consistent with having undergone full maturation, and were visualized on the cell surface. Although both mutants were impaired in their MAPK responses to increasing extracellular calcium concentrations relative to wild type, this was more marked for R227L (EC(50) = 9.7 mM) than R227Q (EC(50) = 7.9 mM) relative to wild type (EC(50) = 3.7 mM). When cotransfected with wild-type CaSR to mimic the heterozygous state, the curves for both R227Q and R227L were right shifted intermediate to the curves for wild type and the respective mutant. This differential responsiveness may account, in part, for the markedly different clinical presentation of the R227Q mutation, classic FBHH, vs. the neonatal hyperparathyroidism of the R227L mutation.


Assuntos
Códon/genética , Hipercalcemia/genética , Hiperparatireoidismo/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores de Detecção de Cálcio/genética , Substituição de Aminoácidos , Sequência de Bases , Cálcio/sangue , DNA Complementar/genética , Feminino , Humanos , Magnésio/sangue , Masculino , Pessoa de Meia-Idade , Linhagem , Fosfatos/sangue
2.
Hum Mutat ; 24(2): 107-11, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15241791

RESUMO

Familial hypocalciuric hypercalcemia (FHH) is caused by heterozygous loss-of-function mutations in the calcium-sensing receptor (CASR), in which the lifelong hypercalcemia is generally asymptomatic. Homozygous loss-of-function CASR mutations manifest as neonatal severe hyperparathyroidism (NSHPT), a rare disorder characterized by extreme hypercalcemia and the bony changes of hyperparathyroidism, which occur in infancy. Activating mutations in the CASR gene have been identified in several families with autosomal dominant hypocalcemia (ADH), autosomal dominant hypoparathyroidism, or hypocalcemic hypercalciuria. Individuals with ADH may have mild hypocalcemia and relatively few symptoms. However, in some cases seizures can occur, especially in younger patients, and these often happen during febrile episodes due to intercurrent infection. Thus far, 112 naturally-occurring mutations in the human CASR gene have been reported, of which 80 are unique and 32 are recurrent. To better understand the mutations causing defects in the CASR gene and to define specific regions relevant for ligand-receptor interaction and other receptor functions, the data on mutations were collected and the information was centralized in the CASRdb (www.casrdb.mcgill.ca), which is easily and quickly accessible by search engines for retrieval of specific information. The information can be searched by mutation, genotype-phenotype, clinical data, in vitro analyses, and authors of publications describing the mutations. CASRdb is regularly updated for new mutations and it also provides a mutation submission form to ensure up-to-date information. The home page of this database provides links to different web pages that are relevant to the CASR, as well as disease clinical pages, sequence of the CASR gene exons, and position of mutations in the CASR. The CASRdb will help researchers to better understand and analyze the mutations, and aid in structure-function analyses.


Assuntos
Bases de Dados Genéticas , Genes Dominantes/genética , Marcadores Genéticos/genética , Mutação/genética , Receptores de Detecção de Cálcio/genética , Sistemas de Gerenciamento de Base de Dados , Previsões , Genótipo , Humanos , Recém-Nascido , Internet , Fenótipo , Software
3.
J Clin Endocrinol Metab ; 88(8): 3674-81, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12915654

RESUMO

De novo activating mutations in the calcium-sensing receptor (CASR) gene are a common cause of sporadic isolated hypoparathyroidism. Here, we describe a family in which two affected siblings were found to be heterozygous for a novel F788L mutation in the fifth transmembrane domain encoded by exon 7 of the CASR. Both parents and the third sibling were clinically unaffected and genotypically normal by direct sequencing of their leukocyte exon 7 PCR amplicons. However, the mother was revealed to be a mosaic for the mutation by sequence analysis of multiple subclones as well as denaturing HPLC of the CASR exon 7 leukocyte PCR product. A functional analysis of the mutation was performed by transiently transfecting wild-type and mutant CASRs tagged with a c-Myc epitope in human embryonic kidney (HEK293) cells. The mutant CASR was expressed at a similar level as the wild type. The F788L mutant produced a significant shift to the left relative to the wild-type CASR in the MAPK response to increasing extracellular calcium concentrations. This is the first report of mosaicism for an activating CASR mutation and suggests that care should be exercised in counseling for risks of recurrence in a situation where a de novo mutation appears likely.


Assuntos
Hipocalcemia/genética , Mosaicismo/genética , Mutação/fisiologia , Receptores de Superfície Celular/genética , Adolescente , Adulto , Cálcio/sangue , Cálcio/fisiologia , Criança , Cromatografia Líquida de Alta Pressão , Impressões Digitais de DNA , Éxons/genética , Feminino , Haplótipos , Humanos , Hipocalcemia/etiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Linhagem , Desnaturação Proteica , Receptores de Detecção de Cálcio , Recidiva , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
PLoS One ; 6(10): e25038, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022372

RESUMO

Genome-wide association studies (GWAS) in several populations have demonstrated significant association of the IL23R gene with IBD (Crohn's disease (CD) and ulcerative colitis (UC)) and psoriasis, suggesting that perturbation of the IL-23 signaling pathway is relevant to the pathophysiology of these diseases. One particular variant, R381Q (rs11209026), confers strong protection against development of CD. We investigated the effects of this variant in primary T cells from healthy donors carrying IL23R(R381) and IL23R(Q381) haplotypes. Using a proprietary anti-IL23R antibody, ELISA, flow cytometry, phosphoflow and real-time RT-PCR methods, we examined IL23R expression and STAT3 phosphorylation and activation in response to IL-23. IL23R(Q381) was associated with reduced STAT3 phosphorylation upon stimulation with IL-23 and decreased number of IL-23 responsive T-cells. We also observed slightly reduced levels of proinflammatory cytokine secretion in IL23R(Q381) positive donors. Our study shows conclusively that IL23R(Q381) is a loss-of-function allele, further strengthening the implication from GWAS results that the IL-23 pathway is pathogenic in human disease. This data provides an explanation for the protective role of R381Q in CD and may lead to the development of improved therapeutics for autoimmune disorders like CD.


Assuntos
Substituição de Aminoácidos/genética , Predisposição Genética para Doença , Doenças Inflamatórias Intestinais/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores de Interleucina/genética , Sequência de Aminoácidos , Arginina/genética , Linhagem Celular Transformada , Células Clonais , Sequência Conservada/genética , Humanos , Interleucina-23/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Contagem de Linfócitos , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação/efeitos dos fármacos , Receptores de Interleucina/química , Fatores de Transcrição STAT/metabolismo , Especificidade da Espécie , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Doadores de Tecidos
5.
J Mol Endocrinol ; 42(4): 331-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19179454

RESUMO

The calcium-sensing receptor (CASR), a plasma membrane G-protein-coupled receptor, is expressed in parathyroid gland and kidney, and controls systemic calcium homeostasis. Inactivating CASR mutations are associated with familial hypocalciuric hypercalcemia (FHH) and neonatal severe hyperparathyroidism, and activating mutations cause autosomal dominant hypocalcemia (ADH). CASR mutation identification plays an important role in the clinical management of mineral metabolism disorders. We describe here a high-throughput method using screening with denaturing high performance liquid chromatography (DHPLC) to initially interrogate 12 amplicons covering translated exons and exon/intron boundaries, followed by sequencing of any amplicon with a modified melting curve relative to wild type, and direct sequencing of a 13th amplicon encoding the COOH-terminal tail to distinguish causative mutations from three common missense single nucleotide polymorphisms. A blinded analysis of 32 positive controls representing mutations throughout the CASR sequence, as well as 22 negative controls, yielded a concordance rate of 100%. We report eight novel and five recurrent FHH mutations, along with six novel and two recurrent ADH mutations. Thus, DHPLC provides a rapid and effective means to screen for CASR mutations.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Mutação/genética , Receptores de Detecção de Cálcio/genética , Biologia Computacional , Análise Mutacional de DNA , Éxons/genética , Genoma Humano/genética , Humanos , Proteínas Mutantes/genética , Mutação de Sentido Incorreto/genética , Reprodutibilidade dos Testes
6.
Hum Mol Genet ; 15(14): 2200-9, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16740594

RESUMO

Calcium-sensing receptor (CASR), expressed in parathyroid gland and kidney, is a critical regulator of extracellular calcium homeostasis. This G protein-coupled receptor exists at the plasma membrane as a homodimer, although it is unclear at which point in the biosynthetic pathway dimerization occurs. To address this issue, we have analyzed wild-type and mutant CASRs harboring R66H, R66C or N583X-inactivating mutations identified in familial hypocalciuric hypercalcemia/neonatal severe hyperparathyroid patients, which were transiently expressed in kidney cells. All mutants were deficient in cell signaling responses to extracellular CASR ligands relative to wild-type. All mutants, although as well expressed as wild-type, lacked mature glycosylation, indicating impaired trafficking from the endoplasmic reticulum (ER). Dimerized forms of wild-type, R66H and R66C mutants were present, but not of the N583X mutant. By immunofluorescence confocal microscopy of non-permeabilized cells, although cell surface expression was observed for the wild-type, little or none was seen for the mutants. In permeabilized cells, perinuclear staining was observed for both wild-type and mutants. By colocalization fluorescence confocal microscopy, the mutant CASRs were localized within the ER but not within the Golgi apparatus. By the use of photobleaching fluorescence resonance energy transfer microscopy, it was demonstrated that the wild-type, R66H and R66C mutants were dimerized in the ER, whereas the N583X mutant was not. Hence, constitutive CASR dimerization occurs in the ER and is likely to be necessary, but is not sufficient, for exit of the receptor from the ER and trafficking to the cell surface.


Assuntos
Retículo Endoplasmático/metabolismo , Mutação Puntual , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/genética , Fenômenos Biofísicos , Biofísica , Linhagem Celular , Membrana Celular/metabolismo , Dimerização , Feminino , Transferência Ressonante de Energia de Fluorescência , Glicosilação , Complexo de Golgi/metabolismo , Humanos , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hiperparatireoidismo/genética , Hiperparatireoidismo/metabolismo , Lactente , Recém-Nascido , Sistema de Sinalização das MAP Quinases , Masculino , Linhagem , Estrutura Quaternária de Proteína , Receptores de Detecção de Cálcio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção
7.
Hum Mol Genet ; 14(12): 1679-90, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15879434

RESUMO

The CASR, a cell surface glycoprotein expressed in parathyroid gland and kidney, is critical for maintaining extracellular calcium homeostasis. The inherited disorders, familial hypocalciuric hypercalcemia (FHH) and neonatal severe hyperparathyroidism (NSHPT), are caused by inactivating mutations in the CASR gene. The CASR has an N-terminal, 19 amino acid signal peptide that is predicted to direct the nascent polypeptide chain, as it emerges from the ribosome, into the endoplasmic reticulum (ER). Here, we report the functional characterization of three CASR mutations identified in hypercalcemic/hyperparathyroid patients. The mutations, L11S, L13P and T14A, lie within the signal peptide hydrophobic core. When transiently transfected into kidney cells, L11S and L13P mutants demonstrated reduced intracellular and plasma membrane expression and signaling to the mitogen-activated protein kinase pathway in response to extracellular calcium relative to wild-type CASR and the T14A mutant. All mutant CASR RNAs translated into protein normally. In cotranslational processing assays, which test the functionality of the signal peptide in the early secretory pathway, the wild-type CASR and mutant T14A nascent polypeptides were targeted to microsomal vesicles, representing the ER, translocated into the vesicular lumen and underwent core N-glycosylation. In contrast, the L11S and L13P mutants failed to be inserted in the microsomes and undergo glycosylation. This is the first study examining the function of the CASR signal sequence and reveals that both L11S and L13P mutants are markedly impaired with respect to cotranslational processing, accounting for the observed parathyroid dysfunction.


Assuntos
Hipercalcemia/genética , Hipercalcemia/metabolismo , Hiperparatireoidismo/genética , Hiperparatireoidismo/metabolismo , Mutação de Sentido Incorreto/genética , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas , Receptores de Detecção de Cálcio/genética , Adulto , Sequência de Aminoácidos , Cálcio/metabolismo , Criança , Feminino , Glicosilação , Humanos , Rim/metabolismo , Masculino , Microssomos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Linhagem , Transporte Proteico , Receptores de Detecção de Cálcio/metabolismo , Homologia de Sequência de Aminoácidos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA