Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 27(1): 1-13, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29040558

RESUMO

Fibroblast growth factor receptor 3 (FGFR3) gain-of-function mutations cause dwarfisms, including achondroplasia (ACH) and thanatophoric dysplasia (TD). The constitutive activation of FGFR3 disrupts the normal process of skeletal growth. Bone-growth anomalies have been identified in skeletal ciliopathies, in which primary cilia (PC) function is disrupted. In human ACH and TD, the impact of FGFR3 mutations on PC in growth plate cartilage remains unknown. Here we showed that in chondrocytes from human (ACH, TD) and mouse Fgfr3Y367C/+ cartilage, the constitutively active FGFR3 perturbed PC length and the sorting and trafficking of intraflagellar transport (IFT) 20 to the PC. We demonstrated that inhibiting FGFR3 with FGFR inhibitor, PD173074, rescued both PC length and IFT20 trafficking. We also studied the impact of rapamycin, an inhibitor of mammalian target of rapamycin (mTOR) pathway. Interestingly, mTOR inhibition also rescued PC length and IFT20 trafficking. Together, we provide evidence that the growth plate defects ascribed to FGFR3-related dwarfisms are potentially due to loss of PC function, and these dwarfisms may represent a novel type of skeletal disorders with defective ciliogenesis.


Assuntos
Acondroplasia/metabolismo , Proteínas de Transporte/metabolismo , Condrócitos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Acondroplasia/genética , Acondroplasia/patologia , Animais , Desenvolvimento Ósseo/genética , Proteínas de Transporte/genética , Cartilagem/metabolismo , Cartilagem/patologia , Diferenciação Celular/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Condrócitos/patologia , Cílios/genética , Cílios/metabolismo , Modelos Animais de Doenças , Feminino , Lâmina de Crescimento/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Pirimidinas/farmacologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais
2.
Hum Mol Genet ; 25(14): 2997-3010, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27260401

RESUMO

Activating FGFR3 mutations in human result in achondroplasia (ACH), the most frequent form of dwarfism, where cartilages are severely disturbed causing long bones, cranial base and vertebrae defects. Because mandibular development and growth rely on cartilages that guide or directly participate to the ossification process, we investigated the impact of FGFR3 mutations on mandibular shape, size and position. By using CT scan imaging of ACH children and by analyzing Fgfr3Y367C/+ mice, a model of ACH, we show that FGFR3 gain-of-function mutations lead to structural anomalies of primary (Meckel's) and secondary (condylar) cartilages of the mandible, resulting in mandibular hypoplasia and dysmorphogenesis. These defects are likely related to a defective chondrocyte proliferation and differentiation and pan-FGFR tyrosine kinase inhibitor NVP-BGJ398 corrects Meckel's and condylar cartilages defects ex vivo. Moreover, we show that low dose of NVP-BGJ398 improves in vivo condyle growth and corrects dysmorphologies in Fgfr3Y367C/+ mice, suggesting that postnatal treatment with NVP-BGJ398 mice might offer a new therapeutic strategy to improve mandible anomalies in ACH and others FGFR3-related disorders.


Assuntos
Acondroplasia/genética , Cartilagem/anormalidades , Mandíbula/anormalidades , Côndilo Mandibular/anormalidades , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Acondroplasia/diagnóstico por imagem , Acondroplasia/tratamento farmacológico , Acondroplasia/fisiopatologia , Animais , Cartilagem/crescimento & desenvolvimento , Cartilagem/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Condrócitos/metabolismo , Condrócitos/patologia , Modelos Animais de Doenças , Humanos , Mandíbula/crescimento & desenvolvimento , Mandíbula/fisiopatologia , Côndilo Mandibular/crescimento & desenvolvimento , Côndilo Mandibular/fisiopatologia , Camundongos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Compostos de Fenilureia/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem
3.
J Clin Invest ; 126(5): 1871-84, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27064282

RESUMO

Achondroplasia (ACH) is the most frequent form of dwarfism and is caused by gain-of-function mutations in the fibroblast growth factor receptor 3-encoding (FGFR3-encoding) gene. Although potential therapeutic strategies for ACH, which aim to reduce excessive FGFR3 activation, have emerged over many years, the use of tyrosine kinase inhibitor (TKI) to counteract FGFR3 hyperactivity has yet to be evaluated. Here, we have reported that the pan-FGFR TKI, NVP-BGJ398, reduces FGFR3 phosphorylation and corrects the abnormal femoral growth plate and calvaria in organ cultures from embryos of the Fgfr3Y367C/+ mouse model of ACH. Moreover, we demonstrated that a low dose of NVP-BGJ398, injected subcutaneously, was able to penetrate into the growth plate of Fgfr3Y367C/+ mice and modify its organization. Improvements to the axial and appendicular skeletons were noticeable after 10 days of treatment and were more extensive after 15 days of treatment that started from postnatal day 1. Low-dose NVP-BGJ398 treatment reduced intervertebral disc defects of lumbar vertebrae, loss of synchondroses, and foramen-magnum shape anomalies. NVP-BGJ398 inhibited FGFR3 downstream signaling pathways, including MAPK, SOX9, STAT1, and PLCγ, in the growth plates of Fgfr3Y367C/+ mice and in cultured chondrocyte models of ACH. Together, our data demonstrate that NVP-BGJ398 corrects pathological hallmarks of ACH and support TKIs as a potential therapeutic approach for ACH.


Assuntos
Acondroplasia/tratamento farmacológico , Condrócitos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Pirimidinas/farmacologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Acondroplasia/genética , Acondroplasia/metabolismo , Acondroplasia/patologia , Animais , Linhagem Celular Transformada , Condrócitos/patologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Disco Intervertebral/metabolismo , Disco Intervertebral/patologia , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Mutantes , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
4.
Endocrinology ; 155(8): 3123-35, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24914940

RESUMO

Thyroid hormone (T3) is required for postnatal skeletal growth. It exerts its effect by binding to nuclear receptors, TRs including TRα1 and TRß1, which are present in most cell types. These cell types include chondrocytes and osteoblasts, the interactions of which are known to regulate endochondral bone formation. In order to analyze the respective functions of T3 stimulation in chondrocytes and osteoblasts during postnatal growth, we use Cre/loxP recombination to express a dominant-negative TRα1(L400R) mutant receptor in a cell-specific manner. Phenotype analysis revealed that inhibiting T3 response in chondrocytes is sufficient to reproduce the defects observed in hypothyroid mice, not only for cartilage maturation, but also for ossification and mineralization. TRα1(L400R) in chondrocytes also results in skull deformation. In the meantime, TRα1(L400R) expression in mature osteoblasts has no visible effect. Transcriptome analysis identifies a number of changes in gene expression induced by TRα1(L400R) in cartilage. These changes suggest that T3 normally cross talks with several other signaling pathways to promote chondrocytes proliferation, differentiation, and skeletal growth.


Assuntos
Desenvolvimento Ósseo , Condrócitos/fisiologia , Osteoblastos/fisiologia , Receptores alfa dos Hormônios Tireóideos/fisiologia , Animais , Diferenciação Celular , Condrócitos/citologia , Feminino , Hipotireoidismo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteogênese , Receptor Cross-Talk , Tri-Iodotironina/fisiologia
5.
Hum Mol Genet ; 23(11): 2914-25, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24419316

RESUMO

FGFR3 gain-of-function mutations lead to both chondrodysplasias and craniosynostoses. Achondroplasia (ACH), the most frequent dwarfism, is due to an FGFR3-activating mutation which results in impaired endochondral ossification. The effects of the mutation on membranous ossification are unknown. Fgfr3(Y367C/+) mice mimicking ACH and craniofacial analysis of patients with ACH and FGFR3-related craniosynostoses provide an opportunity to address this issue. Studying the calvaria and skull base, we observed abnormal cartilage and premature fusion of the synchondroses leading to modifications of foramen magnum shape and size in Fgfr3(Y367C/+) mice, ACH and FGFR3-related craniosynostoses patients. Partial premature fusion of the coronal sutures and non-ossified gaps in frontal bones were also present in Fgfr3(Y367C/+) mice and ACH patients. Our data provide strong support that not only endochondral ossification but also membranous ossification is severely affected in ACH. Demonstration of the impact of FGFR3 mutations on craniofacial development should initiate novel pharmacological and surgical therapeutic approaches.


Assuntos
Acondroplasia/enzimologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Acondroplasia/genética , Acondroplasia/patologia , Animais , Condrócitos/citologia , Condrócitos/enzimologia , Feminino , Humanos , Lactente , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Ossificação Heterotópica , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Crânio/anatomia & histologia , Crânio/embriologia , Crânio/enzimologia , Crânio/patologia
6.
Am J Hum Genet ; 91(6): 1108-14, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23200862

RESUMO

Achondroplasia (ACH), the most common form of dwarfism, is an inherited autosomal-dominant chondrodysplasia caused by a gain-of-function mutation in fibroblast-growth-factor-receptor 3 (FGFR3). C-type natriuretic peptide (CNP) antagonizes FGFR3 downstream signaling by inhibiting the pathway of mitogen-activated protein kinase (MAPK). Here, we report the pharmacological activity of a 39 amino acid CNP analog (BMN 111) with an extended plasma half-life due to its resistance to neutral-endopeptidase (NEP) digestion. In ACH human growth-plate chondrocytes, we demonstrated a decrease in the phosphorylation of extracellular-signal-regulated kinases 1 and 2, confirming that this CNP analog inhibits fibroblast-growth-factor-mediated MAPK activation. Concomitantly, we analyzed the phenotype of Fgfr3(Y367C/+) mice and showed the presence of ACH-related clinical features in this mouse model. We found that in Fgfr3(Y367C/+) mice, treatment with this CNP analog led to a significant recovery of bone growth. We observed an increase in the axial and appendicular skeleton lengths, and improvements in dwarfism-related clinical features included flattening of the skull, reduced crossbite, straightening of the tibias and femurs, and correction of the growth-plate defect. Thus, our results provide the proof of concept that BMN 111, a NEP-resistant CNP analog, might benefit individuals with ACH and hypochondroplasia.


Assuntos
Acondroplasia/tratamento farmacológico , Peptídeo Natriurético Tipo C/análogos & derivados , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Acondroplasia/diagnóstico , Acondroplasia/genética , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Modelos Animais de Doenças , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/patologia , Humanos , Camundongos , Mutação , Peptídeo Natriurético Tipo C/química , Peptídeo Natriurético Tipo C/fisiologia , Peptídeo Natriurético Tipo C/uso terapêutico , Tamanho do Órgão/efeitos dos fármacos , Radiografia , Crânio/diagnóstico por imagem , Crânio/efeitos dos fármacos , Crânio/patologia , Resultado do Tratamento
7.
Bone ; 47(5): 905-15, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20673820

RESUMO

Achondroplasia (ACH), the most common form of human dwarfism is caused by a mutation in the Fibroblast Growth Factor Receptor 3 (FGFR3) gene, resulting in constitutive activation of the receptor. Typical radiological features include shortening of the tubular bones and macrocephaly, due to disruption of endochondral ossification. Consequently, FGFR3 has been described as a negative regulator of bone growth. Studying a large cohort of ACH patients, a delay in bone age was observed shortly after birth (for boys p=2.6×10(-9) and for girls p=1.2×10(-8)). This delay was no longer apparent during adolescence. In order to gain further insight into bone formation, bone development was studied in a murine model of chondrodysplasia (Fgfr3(Y367C/+)) from birth to 6weeks of age. Delayed bone age was also observed in Fgfr3(Y367C/+) mice at 1week of age followed by an accelerated secondary ossification center formation. A low level of chondrocyte proliferation was observed in the normal growth plate at birth, which increased with bone growth. In the pathological condition, a significantly high level of proliferative cells was present at birth, but exhibited a transient decrease only to rise again subsequently. Histological and in situ analyses suggested the altered endochondral ossification process may result from delayed chondrocyte differentiation, disruption of vascularization and osteoblast invasion of the femur. All these data provide evidence that FGFR3 regulates normal chondrocyte proliferation and differentiation during bone growth and suggest that constitutive activation of the receptor disrupts both processes. Therefore, the consequences of FGFR3 activation on the physiological process of bone development appear to be dependent on spatial and temporal occurrence. In conclusion, these observations support the notion that FGFR3 has a dual effect, as both a negative and a positive regulator of the endochondral ossification process during post-natal bone development.


Assuntos
Acondroplasia/genética , Acondroplasia/patologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Adolescente , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Criança , Pré-Escolar , Condrócitos/citologia , Feminino , Humanos , Hibridização In Situ , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Mutantes , Mutação , Ossificação Heterotópica
8.
PLoS One ; 4(10): e7633, 2009 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-19898608

RESUMO

Endochondral ossification is the process by which the appendicular skeleton, facial bones, vertebrae and medial clavicles are formed and relies on the tight control of chondrocyte maturation. Fibroblast growth factor receptor (FGFR)3 plays a role in bone development and maintenance and belongs to a family of proteins which differ in their ligand affinities and tissue distribution. Activating mutations of the FGFR3 gene lead to craniosynostosis and multiple types of skeletal dysplasia with varying degrees of severity: thanatophoric dysplasia (TD), achondroplasia and hypochondroplasia. Despite progress in the characterization of FGFR3-mediated regulation of cartilage development, many aspects remain unclear. The aim and the novelty of our study was to examine whole gene expression differences occurring in primary human chondrocytes isolated from normal cartilage or pathological cartilage from TD-affected fetuses, using Affymetrix technology. The phenotype of the primary cells was confirmed by the high expression of chondrocytic markers. Altered expression of genes associated with many cellular processes was observed, including cell growth and proliferation, cell cycle, cell adhesion, cell motility, metabolic pathways, signal transduction, cell cycle process and cell signaling. Most of the cell cycle process genes were down-regulated and consisted of genes involved in cell cycle progression, DNA biosynthesis, spindle dynamics and cytokinesis. About eight percent of all modulated genes were found to impact extracellular matrix (ECM) structure and turnover, especially glycosaminoglycan (GAG) and proteoglycan biosynthesis and sulfation. Altogether, the gene expression analyses provide new insight into the consequences of FGFR3 mutations in cell cycle regulation, onset of pre-hypertrophic differentiation and concomitant metabolism changes. Moreover, impaired motility and ECM properties may also provide clues about growth plate disorganization. These results also suggest that many signaling pathways may be directly or indirectly altered by FGFR3 and confirm the crucial role of FGFR3 in the control of growth plate development.


Assuntos
Cartilagem/embriologia , Condrócitos/metabolismo , Exostose Múltipla Hereditária/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Cartilagem/metabolismo , Proliferação de Células , Condrócitos/citologia , Exostose Múltipla Hereditária/metabolismo , Matriz Extracelular , Heterozigoto , Humanos , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Proteoglicanas/metabolismo , Controle de Qualidade , Transdução de Sinais
9.
FEBS Lett ; 581(14): 2593-8, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17507011

RESUMO

Achondroplasia and thanatophoric dysplasia are human chondrodysplasias caused by mutations in the fibroblast growth factor receptor 3 (FGFR3) gene. We have developed an immortalized human chondrocyte culture model to study the regulation of chondrocyte functions. One control and eight mutant chondrocytic lines expressing different FGFR3 heterozygous mutations were obtained. FGFR3 signaling pathways were modified in the mutant lines as revealed by the constitutive activation of the STAT pathway and an increased level of P21(WAF1/CIP1) protein. This model will be useful for the study of FGFR3 function in cartilage studies and future therapeutic approaches in chondrodysplasias.


Assuntos
Condrócitos/metabolismo , Mutação , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Condrócitos/citologia , Colágeno Tipo II/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Expressão Gênica , Heterozigoto , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Immunoblotting , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9 , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética
10.
Bone ; 39(1): 17-26, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16476576

RESUMO

Multiple hereditary exostoses (MHE) is an autosomal dominant skeletal disorder caused by mutations in one of the two EXT genes and characterized by multiple osteochondromas that generally arise near the ends of growing long bones. Defective endochondral ossification is likely to be involved in the formation of osteochondromas. In order to investigate potential changes in chondrocyte proliferation and/or differentiation during this process, osteochondroma samples from MHE patients were obtained and used for genetic, morphological, immunohistological, and in situ hybridization studies. The expression patterns of IHH (Indian hedgehog) and FGFR3 (Fibroblast Growth Factor Receptor 3) were similar with transcripts expressed throughout osteochondromas. Expression of PTHR1 (Parathyroid Hormone Receptor 1) transcripts was restricted to a narrow zone of prehypertrophic chondrocytes. Numerous cells forming osteochondromas although resembling prehypertrophic chondrocytes, stained positively with an anti-proliferating cell nuclear antigen (PCNA) antibody. In addition, ectopic expression of collagen type I and abnormal presence of osteocalcin (OC), osteopontin (OP), and bone sialoprotein (BSP) were observed in the cartilaginous osteochondromas. These data indicate that most chondrocytes involved in the growth of osteochondromas can proliferate, and that some of them exhibit bone-forming cell characteristics. We conclude that in MHE, defective heparan sulfate biosynthesis caused by EXT mutations maintains the proliferative capacity of chondrocytes and promotes phenotypic modification to bone-forming cells.


Assuntos
Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Proliferação de Células , Condrócitos/patologia , Exostose Múltipla Hereditária/genética , Adolescente , Adulto , Estudos de Casos e Controles , Células Cultivadas , Criança , Pré-Escolar , Condrócitos/ultraestrutura , Colágeno Tipo I/metabolismo , DNA/genética , Análise Mutacional de DNA , Exostose Múltipla Hereditária/diagnóstico , Exostose Múltipla Hereditária/patologia , Feminino , Ligação Genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Sialoproteína de Ligação à Integrina , Perda de Heterozigosidade , Masculino , Mutação , Osteocalcina/metabolismo , Antígeno Nuclear de Célula em Proliferação/análise , Sialoglicoproteínas/metabolismo
11.
Am J Pathol ; 161(4): 1325-35, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12368206

RESUMO

The fibroblast growth factor receptor type 3 (FGFR3) and Indian hedgehog (IHH)/parathyroid hormone (PTH)/PTH-related peptide receptor type 1 (PTHR1) systems are both essential regulators of endochondral ossification. Based on mouse models, activation of the FGFR3 system is suggested to regulate the IHH/PTHR1 pathway. To challenge this possible interaction in humans, we analyzed the femoral growth plates from fetuses carrying activating FGFR3 mutations (9 achondroplasia, 21 and 8 thanatophoric dysplasia types 1 and 2, respectively) and 14 age-matched controls by histological techniques and in situ hybridization using riboprobes for human IHH, PTHR1, type 10 and type 1 collagen transcripts. We show that bone-perichondrial ring enlargement and growth plate increased vascularization in FGFR3-mutated fetuses correlate with the phenotypic severity of the disease. PTHR1 and IHH expression in growth plates, bone-perichondrial rings and vascular canals is not affected by FGFR3 mutations, irrespective of the mutant genotype and age, and is in keeping with cell phenotypes. These results indicate that in humans, FGFR3 signaling does not down-regulate the main players of the IHH/PTHR1 pathway. Furthermore, we show that cells within the bone-perichondrial ring in controls and patients express IHH, PTHR1, and type 10 and type 1 collagen transcripts, suggesting that bone-perichondrial ring formation involves cells of both chondrocytic and osteoblastic phenotypes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/embriologia , Mutação , Proteínas Tirosina Quinases , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Hormônios Paratireóideos/genética , Transativadores/genética , Acondroplasia/embriologia , Acondroplasia/genética , Acondroplasia/patologia , Divisão Celular , Desenvolvimento Embrionário e Fetal , Fêmur/embriologia , Genótipo , Lâmina de Crescimento/anormalidades , Lâmina de Crescimento/patologia , Proteínas Hedgehog , Humanos , Fenótipo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Displasia Tanatofórica/embriologia , Displasia Tanatofórica/genética , Displasia Tanatofórica/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA