Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Osteoarthritis Cartilage ; 23(3): 443-53, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25464167

RESUMO

OBJECTIVE: To test the effects of sequential exposure to FGF2, 9 and 18 on human Mesenchymal Stem Cells (hMSC) differentiation during in vitro chondrogenesis. DESIGN: Control and FGF2-expanded hMSC were cultured in aggregates in the presence of rhFGF9, rhFGF18 or rhFGFR3-specific signaling FGF variants, starting at different times during the chondroinductive program. Quantitative real time polymerase chain reaction (qRT-PCR) and immunocytochemistry were performed at different stages. The aggregate cultures were switched to a hypertrophy-inducing medium along with rhFGFs and neutralizing antibodies against FGFR1 and FGFR3. Histological/immunohistochemical/biochemical analyses were performed. RESULTS: FGF2-exposed hMSC during expansion up-regulated Sox9 suggesting an early activation of the chondrogenic machinery. FGF2, FGF9 and 18 modulated the expression profile of FGFR1 and FGFR3 in hMSC during expansion and chondrogenesis. In combination with transforming growth factor-beta (TGF-ß), FGF9 and FGF18 inhibited chondrogenesis when added at the beginning of the program (≤ d7), while exhibiting an anabolic effect when added later (≥d14), an effect mediated by FGFR3. Finally, FGFR3 signaling induced by either FGF9 or FGF18 delayed the appearance of spontaneous and induced hypertrophy-related changes. CONCLUSIONS: The stage of hMSC-dependent chondrogenesis at which the growth factors are added impacts the progression of the differentiation program: increased cell proliferation and priming (FGF2); stimulated early chondrogenic differentiation (TGF-ß, FGF9/FGF18) by shifting the chondrogenic program earlier; augmented extracellular matrix (ECM) production (FGF9/FGF18); and delayed terminal hypertrophy (FGF9/FGF18). Collectively, these factors could be used to optimize pre-implantation conditions of hMSC when used to engineer cartilage grafts.


Assuntos
Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 9 de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/metabolismo , Humanos , Hipertrofia , Técnicas In Vitro , Células-Tronco Mesenquimais/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo
2.
Acta Biomater ; 101: 293-303, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726249

RESUMO

Biomaterials play a pivotal role in cell-free cartilage repair approaches, where cells must migrate through the scaffold, fill the defect, and then proliferate and differentiate facilitating tissue remodeling. Here we used multiple assays to test the influence of chemokines and growth factors on cell migration and cartilage repair in two different hyaluronan (HA)-based hydrogels. We first investigated bone marrow Mesenchymal Stromal Cells (BMSC) migration in vitro, in response to different concentrations of platelet-derived growth factor-BB (PDGF-BB), chemokine ligand 5 (CCL5/RANTES) and stromal cell-derived factor 1 (SDF-1), using a 3D spheroid-based assay. PDGF-BB was selected as most favourable chemotactic agent, and MSC migration was assessed in the context of physical impediment to cell recruitment by testing Fibrin-HA and HA-Tyramine hydrogels of different cross-linking densities. Supplementation of PDGF-BB stimulated progressive migration of MSC through the gels over time. We then investigated in situ cell migration into the hydrogels with and without PDGF-BB, using a cartilage-bone explant model implanted subcutaneously in athymic mice. In vivo studies show that when placed into an osteochondral defect, both hydrogels supported endogenous cell infiltration and provided an amenable microenvironment for cartilage production. These processes were best supported in Fibrin-HA hydrogel in the absence of PDGF-BB. This study used an advanced preclinical testing platform to select an appropriate microenvironment provided by implanted hydrogels, demonstrating that HA-based hydrogels can promote the initial and critical step of endogenous cell recruitment and circumvent some of the clinical challenges in cartilage tissue repair. STATEMENT OF SIGNIFICANCE: The challenge of articular cartilage repair arises from its complex structure and architecture, which confers the unique mechanical behavior of the extracellular matrix. The aim of our research is to identify biomaterials for implants that can support migration of endogenous stem and progenitor cell populations from cartilage and bone tissue, in order to permanently replace damaged cartilage with the original hyaline structure. Here, we present an in vitro 3D spheroid-based migration assay and an osteochondral defect model, which provide the opportunity to assess biomaterials and biomolecules, and to get stronger experimental evidence of the not well-characterized dynamic process of endogenous cells colonization in an osteochondral defect. Furthermore, the delicate step of early cell migration into biomaterials towards functional tissue engineering is reproduced. These tests can be used for pre-clinical testing of newly developed material designs in the field of scaffold engineering.


Assuntos
Materiais Biomiméticos/farmacologia , Cartilagem Articular/metabolismo , Matriz Extracelular/metabolismo , Ácido Hialurônico/farmacologia , Hidrogéis/farmacologia , Adolescente , Idoso , Animais , Becaplermina/farmacologia , Cartilagem Articular/efeitos dos fármacos , Bovinos , Movimento Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Reagentes de Ligações Cruzadas/farmacologia , Matriz Extracelular/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Esferoides Celulares/efeitos dos fármacos , Tiramina/farmacologia , Cicatrização/efeitos dos fármacos
3.
Clin Orthop Relat Res ; 466(8): 1849-55, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18528742

RESUMO

UNLABELLED: Fibrin, a homologous polymer, is the natural scaffold of wound healing and therefore a candidate as a carrier for cell transplantation. We explored a novel matrix-based implant cartilage repair composed of both fibrin and hyaluronan in a defined ratio that takes advantage of the biological and mechanical properties of these two elements. The matrix was seeded with autologous chondrocytes expanded in the presence of a proprietary growth factor variant designed to preserve their chondrogenic potential. We prospectively followed eight patients with symptomatic-chronic cartilage defects treated with this carrier. Patients had arthroscopy to harvest autologous chondrocytes then grown in autologous serum. Chondrocytes were cultured in the presence of the FGF variant and then seeded on the fibrin-hyaluronan matrix. About 4 weeks following biopsy, the patients underwent implantation of the constructs by miniarthrotomy. Three of the eight patients had transient effusion. Clinical performance was measured by Lysholm and IKDC scores, MRI, and the need for secondary surgery. The clinical outcome of a 1-year followup demonstrated increase of clinical scores. The MRI followup showed good filling of the defect with tissue having the imaging appearance of cartilage in all patients. Apart from the transient effusion in three patients we observed no other adverse events during the followup. LEVEL OF EVIDENCE: Level IV, therapeutic study.


Assuntos
Condrócitos/transplante , Fêmur/lesões , Osteocondrite Dissecante/cirurgia , Alicerces Teciduais , Adulto , Cartilagem/cirurgia , Colagenases/metabolismo , Feminino , Fêmur/cirurgia , Fibrina , Humanos , Imageamento por Ressonância Magnética , Masculino , Projetos Piloto , Alicerces Teciduais/química
4.
J Clin Invest ; 104(11): 1517-25, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10587515

RESUMO

Missense mutations in fibroblast growth factor receptor 3 (FGFR3) result in several human skeletal dysplasias, including the most common form of dwarfism, achondroplasia. Here we show that a glycine-to-cysteine substitution at position 375 (Gly375Cys) in human FGFR3 causes ligand-independent dimerization and phosphorylation of FGFR3 and that the equivalent substitution at position 369 (Gly369Cys) in mouse FGFR3 causes dwarfism with features mimicking human achondroplasia. Accordingly, homozygous mice were more severely affected than heterozygotes. The resulting mutant mice exhibited macrocephaly and shortened limbs due to retarded endochondral bone growth and premature closure of cranial base synchondroses. Compared with their wild-type littermates, mutant mice growth plates shared an expanded resting zone and narrowed proliferating and hypertrophic zones, which is correlated with the activation of Stat proteins and upregulation of cell-cycle inhibitors. Reduced bone density is accompanied by increased activity of osteoclasts and upregulation of genes that are related to osteoblast differentiation, including osteopontin, osteonectin, and osteocalcin. These data reveal an essential role for FGF/FGFR3 signals in both chondrogenesis and osteogenesis during endochondral ossification.


Assuntos
Acondroplasia/genética , Condrogênese/genética , Osteogênese/genética , Proteínas Tirosina Quinases , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Linhagem Celular , Dimerização , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/farmacologia , Marcação de Genes , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Mutação , Osteocalcina/metabolismo , Osteonectina/metabolismo , Osteopontina , Fosforilação , Radiografia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Sialoglicoproteínas/metabolismo , Transfecção
5.
Mol Cell Biol ; 17(4): 1938-46, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9121441

RESUMO

Heparan sulfate proteoglycans (HSPG) play a critical role in the formation of distinct fibroblast growth factor (FGF)-HS complexes, augmenting high-affinity binding and receptor activation. Perlecan, a secreted HSPG abundant in proliferating cells, is capable of inducing FGF-receptor interactions in vitro and angiogenesis in vivo. Stable and specific reduction of perlecan levels in mouse NIH 3T3 fibroblasts and human metastatic melanoma cells has been achieved by expression of antisense cDNA corresponding to the N-terminal and HS attachment domains of perlecan. Long-term perlecan downregulation is evidenced by reduced levels of perlecan mRNA and core protein as indicated by Northern blot analysis, immunoblots, and immunohistochemistry, using DNA probes and antibodies specific to mouse or human perlecan. The response of antisense perlecan-expressing cells to increasing concentrations of basic FGF (bFGF) is dramatically reduced in comparison to that in wild-type or vector-transfected cells, as measured by thymidine incorporation and rate of proliferation. Furthermore, receptor binding and affinity labeling of antisense perlecan-transfected cells with 125I-bFGF is markedly inhibited, indicating that eliminating perlecan expression results in reduced high-affinity bFGF binding. Both the binding and mitogenic response of antisense-perlecan-expressing clones to bFGF can be rescued by exogenous heparin or perlecan. These results support the notion that perlecan is a major accessory receptor for bFGF in mouse fibroblasts and human melanomas and point to the possible use of perlecan antisense constructs as specific modulators of bFGF-mediated responses.


Assuntos
DNA Antissenso/farmacologia , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Proteoglicanas de Heparan Sulfato , Heparitina Sulfato/genética , Proteoglicanas/genética , Células 3T3 , Animais , Divisão Celular/efeitos dos fármacos , DNA Antissenso/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Heparina/farmacologia , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Proteoglicanas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
6.
Mol Cell Biol ; 20(2): 516-22, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10611230

RESUMO

A point mutation, Gly380Arg, in the transmembrane domain of fibroblast growth factor receptor 3 (FGFR3) leads to achondroplasia, the most common form of genetic dwarfism in humans. This substitution was suggested to enhance mutant receptor dimerization, leading to constitutive, ligand-independent activation. We found that dimerization and activation of the G380R mutant receptor are predominantly ligand dependent. However, using both transient and stable transfections, we found significant overexpression only of the mutant receptor protein. Metabolic pulse-chase experiments, cell surface labeling, and kinetics of uptake of radiolabeled ligand demonstrated a selective delay in the down-regulation of the mutant receptor. Moreover, this receptor was now resistant to ligand-mediated internalization, even at saturating ligand concentrations. Finally, transgenic mice expressing the human G380R mutant receptor under the mouse receptor transcriptional control demonstrated a markedly expanded area of FGFR3 immunoreactivity within their epiphyseal growth plates, compatible with an in vivo defect in receptor down-regulation. We propose that the achondroplasia mutation G380R uncouples ligand-mediated receptor activation from down-regulation at a site where the levels and kinetics of FGFR3 signals are crucial for chondrocyte maturation and bone formation.


Assuntos
Substituição de Aminoácidos/genética , Regulação para Baixo , Proteínas Tirosina Quinases , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Desenvolvimento Ósseo , Linhagem Celular , Membrana Celular/metabolismo , Condrócitos/citologia , Condrócitos/metabolismo , Dimerização , Endocitose , Lâmina de Crescimento/metabolismo , Humanos , Cinética , Ligantes , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peso Molecular , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-fos/metabolismo , Agregação de Receptores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/química , Transdução de Sinais
7.
Mol Cell Biol ; 12(1): 240-7, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1309590

RESUMO

Heparin is required for the binding of basic fibroblast growth factor (bFGF) to high-affinity receptors on cells deficient in cell surface heparan sulfate proteoglycan. So that this heparin requirement could be evaluated in the absence of other cell surface molecules, we designed a simple assay based on a genetically engineered soluble form of murine FGF receptor 1 (mFR1) tagged with placental alkaline phosphatase. Using this assay, we showed that FGF-receptor binding has an absolute requirement for heparin. By using a cytokine-dependent lymphoid cell line engineered to express mFR1, we also showed that FGF-induced mitogenic activity is heparin dependent. Furthermore, we tested a series of small heparin oligosaccharides of defined lengths for their abilities to support bFGF-receptor binding and biologic activity. We found that a heparin oligosaccharide with as few as eight sugar residues is sufficient to support these activities. We also demonstrated that heparin facilitates FGF dimerization, a property that may be important for receptor activation.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Heparina/fisiologia , Receptores de Superfície Celular/metabolismo , Células 3T3 , Fosfatase Alcalina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Divisão Celular/fisiologia , Sistema Livre de Células , DNA , Heparina/química , Humanos , Camundongos , Dados de Sequência Molecular , Oligossacarídeos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos , Solubilidade
8.
J Natl Cancer Inst ; 85(2): 121-31, 1993 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-8418301

RESUMO

BACKGROUND: Recombinant human basic fibroblast growth factor (rHu-bFGF) is known to stimulate proliferation in some tumor cells and to modulate tumor vascularization. PURPOSE: The purpose of this study was to examine the possible role of this agent in the development of tumors. The study was designed to determine the effects of modulating bFGF activity in vivo in tumor models from cell lines with different responses to bFGF and with different content and receptor levels of bFGF. METHODS: Two tumor cell lines (human DLD-2 colon carcinoma and rat C6 glioma) were characterized for bFGF content and bFGF receptor levels by Western blot analysis in cultured cells and by studies of [125I]rHu-bFGF binding to sections from xenografts grown in nude mice. Tumor cell proliferation was monitored after treatment with rHu-bFGF or the DG2 or DE6 IgG monoclonal antibody to rHu-bFGF in culture and in vivo. RESULTS: C6 cells exhibited 7800 high-affinity receptors for rHu-bFGF per cell (dissociation constant [Kd] = 46 pM), while DLD-2 cells lacked high-affinity receptors. rHu-bFGF stimulated [3H]thymidine uptake by C6 cells, but the addition of DG2 IgG prevented this stimulation; rHu-bFGF had no effect on [3H]thymidine incorporation by DLD-2 cells. C6 cells had higher levels of immunoreactive bFGF than did DLD-2 cells. The xenografts from both cell lines exhibited high-affinity [125I]rHu-bFGF binding that was concentrated on vascular-like structures. rHu-bFGF at a dosage of 0.25 mg/kg given intraperitoneally daily for 18 days caused a twofold increase in DLD-2 tumor weight but had little effect on the growth of C6 xenografts. In contrast, daily intravenous injections of DG2 IgG given to mice had no effect on DLD-2 tumor growth but reduced growth of C6 tumors by approximately 30%--a statistically significant difference. CONCLUSIONS: The addition of exogenous rHu-bFGF or of a neutralizing antibody resulted in significant alterations in tumor growth in vivo, which were specific for tumor type and bFGF characteristics. While some of these effects may be mediated by the bFGF-responsive endothelial cells of the tumor vasculature (DLD-2 colon carcinoma), others may result from inhibition of bFGF-dependent tumor cell proliferation (C6 glioma). IMPLICATIONS: Studies that measure tumor blood flow are necessary to confirm that these effects are mediated by changes in tumor vasculature.


Assuntos
Neoplasias do Colo/patologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glioma/patologia , Animais , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Autorradiografia , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/terapia , Citosol/ultraestrutura , Relação Dose-Resposta a Droga , Fator 2 de Crescimento de Fibroblastos/farmacocinética , Glioma/terapia , Humanos , Imunoglobulina G , Radioisótopos do Iodo , Camundongos , Camundongos Nus , Testes de Neutralização , Ratos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas
9.
Cancer Res ; 52(20): 5656-62, 1992 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-1394189

RESUMO

NIH-3T3 cells transfected with basic fibroblast growth factor (bFGF) fused to a signal peptide sequence (spbFGF cells) are transformed in vitro and tumorigenic in vivo. Treatment of spbFGF cells with low and nontoxic concentrations (0.5-2.5 micrograms/ml) of negatively charged, nonsulfated aromatic compounds (e.g., aurin tricarboxylic acid, 4-hydroxyphenoxyacetic acid) resulted in restoration of their normal proliferative rate, morphological appearance, and adhesion properties. Binding and cross-linking experiments using 125I-labeled bFGF revealed that these alterations were associated with an up-regulation of high affinity receptors bFGF receptors was induced by these compounds in spbFGF cells that were seeded on fibronectin to enforce a firm cell attachment and flattening. Thus, induction of spbFGF cell adhesion and spreading may not be related to restoration of normal bFGF-receptor interactions. Although the negatively charged aromatic compounds mimic many of the effects of heparin in other systems (e.g., release of heparin- and heparan sulfate-bound proteins, inhibition of heparanase), heparin, heparan sulfate, and dextran sulfate were not effective at the low concentrations of the anionic compounds used in the present study. Likewise, suramin, a sulfated aromatic molecule, was effective at toxic concentrations, 400-600-fold higher than the nonsulfated aromatic compounds. The development of defined, nontoxic anionic compounds may provide a new strategy to interfere with the autonomous and anchorage independent mode of cell growth involved in autocrine cell transformation and cancer.


Assuntos
Fator 2 de Crescimento de Fibroblastos/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Inibidores do Crescimento/farmacologia , Hidrocarbonetos/farmacologia , Células 3T3 , Animais , Ânions/farmacologia , Ácido Aurintricarboxílico/farmacologia , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Camundongos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Cancer Res ; 60(11): 2973-80, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10850445

RESUMO

Fibroblast growth factors (FGFs) and vascular endothelial growth factor (VEGF) play a pivotal role in the multistep pathway of tumor progression, metastasis, and angiogenesis. We have identified a porphyrin analogue, 5,10,15,20-tetrakis(methyl-4-pyridyl)-21H,23H-porphine-tetra -p-tosylate salt (TMPP), as a potent inhibitor of FGF2 and VEGF receptor binding and activation. TMPP demonstrated potent inhibition of binding of soluble FGF receptor 1 (FGFR1) to FGF2 immobilized on heparin at submicromolar concentrations. TMPP inhibits binding of radiolabeled FGF2 to FGFR in a cell-free system as well as to cells genetically engineered to express FGFR1. Furthermore, TMPP also inhibits the binding of VEGF to its tyrosine kinase receptor in a dose-dependent manner. In an in vitro angiogenic assay measuring the extent of endothelial cell growth, tube formation, and sprouting, TMPP dramatically reduced the extent of the FGF2-induced endothelial cell outgrowth and differentiation. In a Lewis lung carcinoma model, mice receiving TMPP showed a marked inhibition of both primary tumor progression and lung metastases development, with nearly total inhibition of the metastatic phenotype upon alternate daily injections of TMPP at 25 microg/g of body mass. Finally, novel meso-pyridylium-substituted, nonsymmetric porphyrins, as well as a novel corrole-based derivative, with >50-fold increase in activity in vitro, had a significantly improved efficacy in blocking tumor progression and metastasis in vivo.


Assuntos
Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Porfirinas/química , Porfirinas/metabolismo , Porfirinas/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento/antagonistas & inibidores , Animais , Aorta/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Progressão da Doença , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Fenótipo , Porfirinas/síntese química , Ligação Proteica/efeitos dos fármacos , Ratos , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
11.
Oncogene ; 7(10): 1957-62, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1408137

RESUMO

Fibroblast growth factors (FGFs) and their receptors play an important role in cell growth, angiogenesis and embryonal development. Four distinct genes encoding fibroblast growth factor receptors (FGFRs) were identified: flg, encoding FGFR1, bek encoding FGFR2, and the genes for FGFR3 and FGFR4. Both FGFR2 and keratinocyte growth factor receptor (KGFR) are encoded by the same gene, bek. To study the regulation of expression of the FGF receptors we analysed the DNA sequence flanking the 5' region of the cDNA of murine FGFR2 to seek elements that control its transcription. A 5-kbp fragment containing the 5' end of the cDNA was isolated from mouse genomic library and used to map the promoter region. We found that the sequence encoding the 5' non-translated region of the FGFR2/KGFR cDNA contains an intron located 210 bp upstream from the translation start site. Using RNAase protection and primer extension, we identified the mRNA start 37 bp upstream from the beginning of the bek cDNA. The promoter activity was found to reside in a 1.3-kbp fragment upstream from the cDNA, and deletion mapping further localized the promoter to a 0.7-kbp fragment. The sequence of this region shows high G+C content (62%), which is particularly emphasized in the 200 bp upstream from the mRNA start (80% G+C). This region contains the CCGCCC, GGGCGG AND GGAGG motifs also found in promoters of other growth factor receptors. Neither TATA nor CAAT boxes were found near the RNA start site. The characterization of this promoter will allow studies of the regulation of expression of the FGFR2 during development and in pathophysiological states. The differences between the promoter sequence of the gene for FGFR2 (bek) and FGFR1 (flg) may explain their differential expression during development.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Regiões Promotoras Genéticas , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Sequência de Bases , DNA/química , Camundongos , Dados de Sequência Molecular , Transcrição Gênica
12.
Oncogene ; 14(25): 2999-3009, 1997 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-9223663

RESUMO

Basic Fibroblast Growth Factor (bFGF/FGF2) is thought to play a decisive role in malignant progression. Aberrant expression of bFGF causes constitutive autocrine activation of its cognate receptor and autonomous growth of human melanoma cells or bFGF transformed fibroblasts in culture. It remains to be determined, however, whether the endogenous bFGF confers growth advantage to tumors and what are the downstream targets of the activated FGF receptor critical for its transforming capacity. We therefore transfected metastatic melanoma cells and bFGF transformed mouse fibroblasts with a dominant-negative mutant of the murine FGF receptor 1 (fgfr1/flg), comprising the extracellular and transmembrane domains but lacking the intracellular kinase domain (dnflg). Reverse transcriptase-PCR, 125I-bFGF binding and affinity labeling analyses show that the truncated receptor is targeted to the membrane and is expressed at much higher levels than the endogenous receptor in all of the selected clones. Expression of the dnflg dramatically reduces the basal as well as bFGF induced growth of these cells in vitro and also suppresses their tumorigenic potential in nude mice. The expression of the dnflg does not significantly alter the general level of tyrosyl-phosphorylated proteins in the trunsduced melanoma cells. Rather, a major downstream affected target is a Src-family kinase, whose activity, determined by an in vitro immune kinase assay, is stimulated in normal melanocytes by exogenous bFGF, and is markedly reduced in the dnflg-expressing melanoma cells. The present study demonstrates that direct interference with the activity of FGF receptors has a deleterious effect on cell proliferation and survival in vitro and in vivo leading to the suppression of melanoma tumor progression possibly through the inactivation of a Src-family kinase.


Assuntos
Fator 2 de Crescimento de Fibroblastos/genética , Melanoma/genética , Melanoma/patologia , Proteínas Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases , Receptores de Fatores de Crescimento de Fibroblastos/genética , Células 3T3/patologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Linhagem Celular Transformada , Fibroblastos/patologia , Proteínas Filagrinas , Genes Dominantes , Humanos , Melanoma/enzimologia , Camundongos , Fenótipo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas , Domínios de Homologia de src
13.
Oncogene ; 6(6): 1089-92, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1648703

RESUMO

Two genes, flg and bek, have been recently shown to encode receptors for fibroblast growth factors (FGF). Here we report the molecular cloning and sequence of a new member of the FGF receptor family, denoted flg-2, which was isolated from a human keratinocyte cDNA library. The cDNA sequence predicts an extracellular region possessing three immunoglobulin-like domains, a single transmembrane region and a cytoplasmic portion containing the tyrosine kinase domain split by a short inter-kinase segment. The amino acid sequence of flg-2 shows 68% and 64% identity with bek and flg, respectively. The most variable domain among the three genes is the amino-terminal immunoglobulin-like domain. Comparison with the chicken FGF receptor genes suggests that flg-2 is homologous to cek-2, whereas flg and bek are homologous to cek-1 and cek-3, respectively. Analysis of mRNA from various tissues shows that flg-2 is expressed predominantly in skin, brain and lung.


Assuntos
Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Sequência de Bases , DNA/genética , Proteínas Filagrinas , Genes/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/ultraestrutura , Dados de Sequência Molecular , Receptores de Superfície Celular/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos
14.
Oncogene ; 15(22): 2717-25, 1997 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-9400998

RESUMO

Basic fibroblast growth factor (bFGF) can exert mitogenic and viability-promoting effects in a wide range of biological systems. The biochemical activities mediating the cell survival function of bFGF are largely unknown. We report here that exposure of fibroblasts to bFGF, which confers upon them increased survival, also causes at the same time an increase in cellular levels of the Mdm2 oncoprotein. Cells constitutively exposed to a bFGF autocrine loop are more refractory to killing by cisplatin. This increased chemoresistance coincides with elevated Mdm2 and reduced activation of the endogenous p53, resulting in inefficient transcriptional activation of the bax gene promoter. Importantly, unlike Mdm2 accumulation in fibroblasts exposed to DNA damage, induction of Mdm2 by bFGF does not occur through a p53-mediated pathway. The role of p53 in DNA damage-induced apoptosis and the ability of Mdm2 to block p53-mediated cell death are well established. These findings therefore suggest that induction of Mdm2 and the subsequent inhibition of p53 function may contribute, at least partially, to the anti-apoptotic effects of bFGF and possibly some other survival factors.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/metabolismo , Células 3T3/efeitos dos fármacos , Animais , Antineoplásicos , Apoptose/efeitos dos fármacos , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/farmacologia , Meios de Cultura , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/efeitos dos fármacos , Ciclinas/genética , Dano ao DNA/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Ratos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2
15.
Biochim Biophys Acta ; 686(2): 197-203, 1982 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-7082662

RESUMO

The kinetic properties of the mediated transport of chloroquine in human erythrocytes are investigated. The high rates of translocation across the cell membrane and high adsorbance properties to glass surfaces have led to the development of new techniques for measuring initial rates of transport. Three different methodological procedures are used to accomplish a complete kinetic characterization of the system. All measurements were done at 25 degrees C. Under zero-trans conditions the system displays complete symmetry, the Michaelis constants being 39.2 +/- 2.4 microM for influx and 36.6 +/- 5.6 microM for efflux. The respective maximal velocities are 206.4 +/- 36.0 microM . min-1 and 190.0 +/- 7.8 microM . min-1. Under equilibrium-exchange conditions the Michaelis constant is 108.6 +/- 15.6 microM and the maximal velocity is 630.3 +/- 50.4 microM . min-1. This 3-fold increase in both K and V over the zero-trans values indicates that the rate-limiting step in the transport of chloroquine is the movement of the unloaded carrier. The kinetic data are consistent with the prediction of a simple carrier model.


Assuntos
Cloroquina/sangue , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Receptores de Droga/metabolismo , Transporte Biológico , Humanos , Cinética , Matemática
16.
Matrix Biol ; 24(8): 520-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16253490

RESUMO

Fibroblast growth factor receptor 3 (FGFR3) signaling pathways are essential for normal longitudinal bone growth. Mutations in this receptor lead to various human growth disorders, including Achondroplasia, disproportionately short-limbed dwarfism, characterized by narrowing of the hypertrophic region of the epiphyseal growth plates. Here we find that FGF9, a preferred ligand for FGFR3 rapidly induces the upregulation and secretion of the matrix resident phosphoprotein, osteopontin (OPN) in cultured chicken chondrocytes. This effect was observed as early as two hours post stimulation and at FGF9 concentrations as low as 1.25 ng/ml at both mRNA and protein levels. OPN expression is known to be associated with chondrocyte and osteoblast differentiation and osteoclast activation. Unexpectedly, FGF9 induced OPN was accompanied by inhibition of differentiation and increased proliferation of the treated chondrocytes. Moreover, FGF9 stimulated OPN expression irrespective of the differentiation stage of the cells or culture conditions. In situ hybridization analysis of epiphyseal growth plates from chicken or mice homozygous for the Achondroplasia, G369C/mFGFR3 mutation demonstrated co-localization of OPN expression and osteoclast activity, as evidenced by tartarate resistant acid phosphatase positive cells in the osteochondral junction. We propose that FGF signaling directly activates OPN expression independent of chondrocytes differentiation. This may enhance the recruitment and activation of osteoclasts, and increase in cartilage resorption and remodeling in the chondro-osseus border.


Assuntos
Condrócitos/efeitos dos fármacos , Fator 9 de Crescimento de Fibroblastos/farmacologia , Lâmina de Crescimento/citologia , Lâmina de Crescimento/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Sialoglicoproteínas/metabolismo , Regulação para Cima/efeitos dos fármacos , Acondroplasia/genética , Acondroplasia/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Galinhas , Condrócitos/metabolismo , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicina/genética , Glicina/metabolismo , Lâmina de Crescimento/metabolismo , Camundongos , Mutação/genética , Osteopontina , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo
17.
J Bone Miner Res ; 14(11): 1909-15, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10571691

RESUMO

Mutations in fibroblast growth factor receptor 3 (FGFR3) cause several human chondrodysplasias, including achondroplasia, the most common form of dwarfism in humans. From in vitro studies, the skeletal defects observed in these disorders have been attributed to constitutive activation of FGFR3. Here we show that FGF9 and FGFR3, a high-affinity receptor for this ligand, have similar developmental expression patterns, particularly in areas of active chondrogenesis. Targeted overexpression of FGF9 to cartilage of transgenic mice disturbs postnatal skeletal development and linear bone growth. The growth plate of these mice exhibits reduced proliferation and terminal differentiation of chondrocytes similar to that observed in the human disorders. The observations provide evidence that targeted, in vivo activation of endogenous FGFR3 inhibits bone growth and demonstrate that signals derived from FGF9-FGFR3 interactions can physiologically block endochondral ossification to produce a phenotype characteristic of the achondroplasia group of human chondrodysplasias.


Assuntos
Condrócitos/metabolismo , Fatores de Crescimento de Fibroblastos , Substâncias de Crescimento/biossíntese , Osteocondrodisplasias/metabolismo , Proteínas Tirosina Quinases , Animais , Desenvolvimento Ósseo , Diferenciação Celular , Linhagem Celular , Fator 9 de Crescimento de Fibroblastos , Expressão Gênica , Substâncias de Crescimento/genética , Camundongos , Camundongos Transgênicos , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética
18.
Hum Gene Ther ; 9(6): 823-33, 1998 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-9581905

RESUMO

Vascular endothelial growth factor (VEGF), a potent angiogenic mediator, is overexpressed in most solid tumors. On the basis of the knowledge that solid tumor growth beyond a small volume is critically dependent on angiogenesis, and that adenovirus (Ad) vectors can mediate efficient in vivo gene transfer and expression, we hypothesized that Ad-mediated transfer of a secreted form of the extracellular domain of the flt-1 VEGF receptor (Adsflt) would suppress tumor growth on a regional basis. To evaluate this concept, three tumor models were examined using a murine colon carcinoma cell line and syngeneic BALB/c mice. First, mice with preestablished splenic CT26.CL25 tumors and liver metastases were given Adsflt on AdNull intravenously and, after 15 days, spleens and livers were harvested to quantify tumor burden. Adslft-treated animals had minimal residual splenic tumors and liver metastases; in contrast, control animals had bulky splenic tumors and extensive liver metastases (p < 0.003). Second, mice with preestablished lung metastases showed a significant reduction in pulmonary metastases with regionally administered Adslft (intratracheal, p < 0.02) but not when the vector was systemically administered (intravenous, p > 0.9). Finally, mice with primary subcutaneous tumors treated with intratumoral administration of Adslft showed significant tumor suppression (p < 0.05) not observed in AdNull-treated mice or mice given Adslft intravenously (p > 0.3). We conclude that Ad-mediated in vivo regional delivery of a secreted form of the extracellular domain of the flt-1 VEGF receptor can effectively inhibit regional tumor growth, a strategy that may provide a means to control tumor growth within the treated organ without the risk of systemic antiangiogenesis.


Assuntos
Terapia Genética , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Adenoviridae , Animais , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , DNA Complementar/administração & dosagem , Técnicas de Transferência de Genes , Vetores Genéticos , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/secundário , Neoplasias Cutâneas/terapia , Células Tumorais Cultivadas , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
19.
FEBS Lett ; 330(3): 249-52, 1993 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-8375495

RESUMO

Four distinct FGF receptors were cloned and characterized and it was demonstrated that the ligand binding site of FGF receptors is confined to the extracellular immunoglobulin-like (Ig)-domain 2 and 3. The Ig-domain 3 is encoded by two separate exons: exon IIIa encodes the N-terminal half, and the C-terminal half is encoded by either exon IIIb or IIIc in FGFR1 and FGFR2, whereas FGFR4 is devoid of exon IIIb. Alternative usage of exons IIIb and IIIc determine the ligand binding specificity of the receptor. To analyze the arrangement of these exons in FGFR3 we cloned the genomic sequence between exon IIIa and IIIc of FGFR3 and identified an alternative exon, corresponding to exon IIIb of the FGFR1 and FGFR2. The sequence of this exon shows Ig-domain hallmarks, 44% identity with exon IIIb of other FGF receptors and 36% identity with exon IIIc of FGFR3. Using this exon as a probe for mouse RNA as well as PCR analysis, demonstrated that exon IIIb encodes an authentic form of FGFR3 that is expressed in mouse embryo, mouse skin and mouse epidermal keratinocytes. The results demonstrate that the presence of alternative exons for Ig-domain 3 is a general phenomena in FGFR1, 2 and 3, and represents a novel genetic mechanism for the generation of receptor diversity.


Assuntos
Éxons , Imunoglobulinas/química , Receptores de Fatores de Crescimento de Fibroblastos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , DNA , Embrião de Mamíferos/citologia , Queratinócitos/química , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Receptores de Fatores de Crescimento de Fibroblastos/química , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Homologia de Sequência de Aminoácidos , Pele/citologia
20.
FEBS Lett ; 528(1-3): 83-9, 2002 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-12297284

RESUMO

A crucial aspect of ligand-mediated receptor activation and shut-down is receptor internalization and degradation. Here we compared the ubiquitylation of either wild type or a K508A 'kinase-dead' mutant of fibroblast growth factor receptor 3 (FGFR3) with that of its naturally occurring overactive mutants, G380R as in achondroplasia, or K650E involved in thanatophoric dysplasia. Fibroblast growth factor receptors ubiquitylation was found to be directly proportional to their intrinsic tyrosine kinase activity, both of which could be blocked using kinase inhibitors. Despite excessive ubiquitylation, both overactive mutants failed to be efficiently degraded, even when challenged with ligand or overexpression of c-Cbl, a putative E3 ligase. We conclude that phosphorylation is essential for FGFR3 ubiquitylation, but is not sufficient to induce downregulation of its internalization resistant mutants.


Assuntos
Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/química , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Acondroplasia/genética , Acondroplasia/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Regulação para Baixo , Humanos , Lisossomos/metabolismo , Complexos Multienzimáticos/metabolismo , Fosforilação , Mutação Puntual , Complexo de Endopeptidases do Proteassoma , Proteínas Tirosina Quinases/genética , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Displasia Tanatofórica/genética , Displasia Tanatofórica/metabolismo , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA