Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Am J Hum Genet ; 90(3): 550-7, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22387015

RESUMO

Fibroblast growth factor receptor 2 (FGFR2) is a crucial regulator of bone formation during embryonic development. Both gain and loss-of-function studies in mice have shown that FGFR2 maintains a critical balance between the proliferation and differentiation of osteoprogenitor cells. We have identified de novo FGFR2 mutations in a sporadically occurring perinatal lethal skeletal dysplasia characterized by poor mineralization of the calvarium, craniosynostosis, dysmorphic facial features, prenatal teeth, hypoplastic pubis and clavicles, osteopenia, and bent long bones. Histological analysis of the long bones revealed that the growth plate contained smaller hypertrophic chondrocytes and a thickened hypercellular periosteum. Four unrelated affected individuals were found to be heterozygous for missense mutations that introduce a polar amino acid into the hydrophobic transmembrane domain of FGFR2. Using diseased chondrocytes and a cell-based assay, we determined that these mutations selectively reduced plasma-membrane levels of FGFR2 and markedly diminished the receptor's responsiveness to extracellular FGF. All together, these clinical and molecular findings are separate from previously characterized FGFR2 disorders and represent a distinct skeletal dysplasia.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Anormalidades Craniofaciais/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Sequência de Aminoácidos , Doenças do Desenvolvimento Ósseo/metabolismo , Osso e Ossos/anormalidades , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Condrócitos/metabolismo , Anormalidades Craniofaciais/metabolismo , Feto/anormalidades , Feto/metabolismo , Fatores de Crescimento de Fibroblastos/deficiência , Heterozigoto , Humanos , Dados de Sequência Molecular , Mutação , Mutação de Sentido Incorreto , Osteoblastos/metabolismo , Osteogênese/genética , Transdução de Sinais , Esqueleto
2.
Nat Genet ; 26(3): 345-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062477

RESUMO

Proper serum phosphate concentrations are maintained by a complex and poorly understood process. Identification of genes responsible for inherited disorders involving disturbances in phosphate homeostasis may provide insight into the pathways that regulate phosphate balance. Several hereditary disorders of isolated phosphate wasting have been described, including X-linked hypophosphataemic rickets (XLH), hypophosphataemic bone disease (HBD), hereditary hypophosphataemic rickets with hypercalciuria (HHRH) and autosomal dominant hypophosphataemic rickets (ADHR). Inactivating mutations of the gene PHEX, encoding a member of the neutral endopeptidase family of proteins, are responsible for XLH (refs 6,7). ADHR (MIM 193100) is characterized by low serum phosphorus concentrations, rickets, osteomalacia, lower extremity deformities, short stature, bone pain and dental abscesses. Here we describe a positional cloning approach used to identify the ADHR gene which included the annotation of 37 genes within 4 Mb of genomic sequence. We identified missense mutations in a gene encoding a new member of the fibroblast growth factor (FGF) family, FGF23. These mutations in patients with ADHR represent the first mutations found in a human FGF gene.


Assuntos
Cromossomos Humanos Par 12/genética , Fatores de Crescimento de Fibroblastos/genética , Genes Dominantes , Genes , Hipofosfatemia Familiar/genética , Anormalidades Múltiplas/genética , Sequência de Aminoácidos , Análise Mutacional de DNA , Europa (Continente) , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/fisiologia , Heterogeneidade Genética , Humanos , Escore Lod , Masculino , Dados de Sequência Molecular , Linhagem , Mutação Puntual , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Estados Unidos
3.
Trends Genet ; 15(2): 59-65, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10098408

RESUMO

Organs have to develop at precisely determined sites to ensure functionality of the whole organism. Organogenesis is typically regulated by a series of interactions between morphologically distinct tissues. The developing tooth of the mouse is an excellent model to study these processes and we are beginning to understand the networks regulating reciprocal tissue interactions at the molecular level. Synergistic and antagonistic effects of signaling molecules including FGFs and BMPs are recursively used to induce localized responses in the adjacent tissue layer (mesenchyme or epithelium). However, at different phases of odontogenesis these secreted growth factors have distinct effects and at the same time they are regulated by different upstream factors. The mesenchymal transcription factors Msx1 and Pax9 are initially regulated by epithelial FGFs and BMPs, but subsequently they function upstream of these signaling molecules. This cascade provides a molecular model by which reciprocal tissue interactions are controlled.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/fisiologia , Odontogênese , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Indução Embrionária , Epitélio/embriologia , Proteínas do Olho , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/fisiologia , Genes Homeobox , Idade Gestacional , Substâncias de Crescimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Fator de Transcrição MSX1 , Mesoderma/fisiologia , Camundongos , Camundongos Knockout , Morfogênese , Odontogênese/genética , Odontogênese/fisiologia , Fator de Transcrição PAX6 , Fator de Transcrição PAX9 , Fatores de Transcrição Box Pareados , Proteínas Repressoras , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
4.
J Bone Miner Res ; 31(6): 1247-57, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26792657

RESUMO

The transgenic and knockout (KO) animals involving Fgf23 have been highly informative in defining novel aspects of mineral metabolism, but are limited by shortened lifespan, inability of spatial/temporal FGF23 control, and infertility of the global KO. To more finely test the role of systemic and genetic influences in FGF23 production, a mouse was developed that carried a floxed ("f")-Fgf23 allele (exon 2 floxed) which demonstrated in vivo recombination when bred to global-Cre transgenic mice (eIIa-cre). Mice homozygous for the recombined allele ("Δ") had undetectable serum intact FGF23, elevated serum phosphate (p < 0.05), and increased kidney Cyp27b1 mRNA (p < 0.05), similar to global Fgf23-KO mice. To isolate cellular FGF23 responses during phosphate challenge, Fgf23(Δ/f) mice were mated with early osteoblast type Iα1 collagen 2.3-kb promoter-cre mice (Col2.3-cre) and the late osteoblast/early osteocyte Dentin matrix protein-1-cre (Dmp1-cre). Fgf23(Δ/f) /Col2.3-cre(+) and Fgf23(Δ/f) /Dmp1-cre(+) exhibited reduced baseline serum intact FGF23 versus controls. After challenge with high-phosphate diet Cre(-) mice had 2.1-fold to 2.5-fold increased serum FGF23 (p < 0.01), but Col2.3-cre(+) mice had no significant increase, and Dmp1-cre(+) mice had only a 37% increase (p < 0.01) despite prevailing hyperphosphatemia in both models. The Fgf23(Δ/f) /Col2.3-cre was bred onto the Hyp (murine X-linked hypophosphatemia [XLH] model) genetic background to test the contribution of osteoblasts and osteocytes to elevated FGF23 and Hyp disease phenotypes. Whereas Hyp mice maintained inappropriately elevated FGF23 considering their marked hypophosphatemia, Hyp/Fgf23(Δ/f) /Col2.3-cre(+) mice had serum FGF23 <4% of Hyp (p < 0.01), and this targeted restriction normalized serum phosphorus and ricketic bone disease. In summary, deleting FGF23 within early osteoblasts and osteocytes demonstrated that both cell types contribute to baseline circulating FGF23 concentrations, and that targeting osteoblasts/osteocytes for FGF23 production can modify systemic responses to changes in serum phosphate concentrations and rescue the Hyp genetic syndrome. © 2016 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/metabolismo , Fatores de Crescimento de Fibroblastos/deficiência , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Osteócitos/metabolismo , Fosfatos/metabolismo , Animais , Osso e Ossos/patologia , Fator de Crescimento de Fibroblastos 23 , Deleção de Genes , Hipofosfatemia Familiar/patologia , Camundongos , Camundongos Knockout , Osteócitos/patologia
5.
Am J Physiol Endocrinol Metab ; 295(2): E254-61, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18559986

RESUMO

Autosomal recessive hypophosphatemic rickets (ARHR), which is characterized by renal phosphate wasting, aberrant regulation of 1alpha-hydroxylase activity, and rickets/osteomalacia, is caused by inactivating mutations of dentin matrix protein 1 (DMP1). ARHR resembles autosomal dominant hypophosphatemic rickets (ADHR) and X-linked hypophosphatemia (XLH), hereditary disorders respectively caused by cleavage-resistant mutations of the phosphaturic factor FGF23 and inactivating mutations of PHEX that lead to increased production of FGF23 by osteocytes in bone. Circulating levels of FGF23 are increased in ARHR and its Dmp1-null mouse homologue. To determine the causal role of FGF23 in ARHR, we transferred Fgf23 deficient/enhanced green fluorescent protein (eGFP) reporter mice onto Dmp1-null mice to create mice lacking both Fgf23 and Dmp1. Dmp1(-/-) mice displayed decreased serum phosphate concentrations, inappropriately normal 1,25(OH)(2)D levels, severe rickets, and a diffuse form of osteomalacia in association with elevated Fgf23 serum levels and expression in osteocytes. In contrast, Fgf23(-/-) mice had undetectable serum Fgf23 and elevated serum phosphate and 1,25(OH)(2)D levels along with severe growth retardation and focal form of osteomalacia. In combined Dmp1(-/-)/Fgf23(-/-), circulating Fgf23 levels were also undetectable, and the serum levels of phosphate and 1,25(OH)(2)D levels were identical to Fgf23(-/-) mice. Rickets and diffuse osteomalacia in Dmp1-null mice were transformed to severe growth retardation and focal osteomalacia characteristic of Fgf23-null mice. These data suggest that the regulation of extracellular matrix mineralization by DMP1 is coupled to renal phosphate handling and vitamin D metabolism through a DMP1-dependent regulation of FGF23 production by osteocytes.


Assuntos
Proteínas da Matriz Extracelular/deficiência , Raquitismo Hipofosfatêmico Familiar/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X , Osteomalacia/metabolismo , Raquitismo/metabolismo , Animais , Densidade Óssea , Calcificação Fisiológica , Calcitriol/sangue , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Raquitismo Hipofosfatêmico Familiar/sangue , Raquitismo Hipofosfatêmico Familiar/genética , Fêmur/metabolismo , Fêmur/patologia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Masculino , Camundongos , Camundongos Knockout , Osteomalacia/sangue , Osteomalacia/genética , Fosfatos/sangue , RNA/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Raquitismo/sangue , Raquitismo/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA