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.
Cells ; 8(9)2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31527546

RESUMO

Even though distinctive advances in the field of esophageal cancer therapy have occurred over the last few years, patients' survival rates remain poor. FGF8, FGF18, and FGFR4 have been identified as promising biomarkers in a number of cancers; however no data exist on expression of FGF8, FGF18, and FGFR4 in adenocarcinomas of the esophago-gastric junction (AEG). A preliminary analysis of the Cancer Genome Atlas (TCGA) database on FGF8, FGF18, and FGFR4 mRNA expression data of patients with AEG was performed. Furthermore, protein levels of FGF8, FGF18, and FGFR4 in diagnostic biopsies and post-operative specimens in neoadjuvantly treated and primarily resected patients using immunohistochemistry were investigated. A total of 242 patients was analyzed in this study: 87 patients were investigated in the TCGA data set analysis and 155 patients in the analysis of protein expression using immunohistochemistry. High protein levels of FGF8, FGF18, and FGFR4 were detected in 94 (60.7%), 49 (31.6%) and 84 (54.2%) patients, respectively. Multivariable Cox proportional hazard regression models revealed that high expression of FGF8 was an independent prognostic factor for diminished overall survival for all patients and for neoadjuvantly treated patients. By contrast, FGF18 overexpression was significantly associated with longer survival rates in neoadjuvantly treated patients. In addition, FGF8 protein level correlated with Mandard regression due to neoadjuvant therapy, indicating potential as a predictive marker. In summary, FGF8 and FGF18 are promising candidates for prognostic factors in adenocarcinomas of the esophago-gastric junction and new potential targets for new anti-cancer therapies.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fatores de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/biossíntese , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Idoso , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/patologia , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Fatores de Crescimento de Fibroblastos/genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Taxa de Sobrevida
2.
J Biol Chem ; 292(25): 10520-10533, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28461333

RESUMO

Amputation of the proximal region in mammals is not followed by regeneration because blastema cells (BCs) and expression of regenerative genes, such as Msh homeobox (Msx) genes, are absent in this animal group. The lack of BCs and positional information in other cells is therefore the main obstacle to therapeutic approaches for limb regeneration. Hence, this study aimed to create blastema-like cells (BlCs) by overexpressing Msx1 and Msx2 genes in mouse bone marrow-derived mesenchymal stem cells (mBMSCs) to regenerate a proximally amputated digit tip. We transduced mBMSCs with Msx1 and Msx2 genes and compared osteogenic activity and expression levels of several Msx-regulated genes (Bmp4, Fgf8, and keratin 14 (K14)) in BlC groups, including MSX1, MSX2, and MSX1/2 (in a 1:1 ratio) with those in mBMSCs and BCs in vitro and in vivo following injection into the amputation site. We found that Msx gene overexpression increased expression of specific blastemal markers and enhanced the proliferation rate and osteogenesis of BlCs compared with mBMSCs and BCs via activation of Fgf8 and Bmp4 Histological analyses indicated full regrowth of digit tips in the Msx-overexpressing groups, particularly in MSX1/2, through endochondral ossification 6 weeks post-injection. In contrast, mBMSCs and BCs formed abnormal bone and nail. Full digit tip was regenerated only in the MSX1/2 group and was related to boosted Bmp4, Fgf8, and K14 gene expression and to limb-patterning properties resulting from Msx1 and Msx2 overexpression. We propose that Msx-transduced cells that can regenerate epithelial and mesenchymal tissues may potentially be utilized in limb regeneration.


Assuntos
Células da Medula Óssea/metabolismo , Membro Posterior/fisiologia , Proteínas de Homeodomínio/biossíntese , Fator de Transcrição MSX1/biossíntese , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Regeneração , Aloenxertos , Animais , Proteína Morfogenética Óssea 4/biossíntese , Proteína Morfogenética Óssea 4/genética , Proliferação de Células/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/genética , Proteínas de Homeodomínio/genética , Queratina-14/biossíntese , Queratina-14/genética , Fator de Transcrição MSX1/genética , Camundongos , Transdução Genética
3.
Appl Microbiol Biotechnol ; 100(2): 625-35, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26411459

RESUMO

Human fibroblast growth factor 8b (FGF8b) was expressed based on a baculovirus expression vector system (BEVS) and identified as having a protective effect on Parkinson's disease. Immunoblotting demonstrated that rhFGF8b proteins were recognized by a human anti-FGF8b antibody. The multiplicity of infection and timing of harvest had a significant effect on protein yield and protein quality. Our results indicated that the rhFGF8b was first detectable at 36 h postinfection and reached a maximum at 60 h. A multiplicity of infection (MOI) of 8 pfu/mL was suitable for harvest. The target protein was purified by heparin-affinity chromatography. In vitro methylthiazol tetrazolium (MTT) assays demonstrated that the purified rhFGF8b could significantly stimulate proliferation of NIH3T3 cells. Furthermore, to elucidate the effect of rhFGF8b on Parkinson's disease, we used FGF8b pretreatment on a cell model of Parkinson's disease. The results indicated that rhFGF8b prevented necrosis and apoptosis of 1-METHYL-4-phenyl pyridine (MPP(+)) treated PC12 cells. Moreover, the effect of FGF8b on messenger RNA (mRNA) levels of apoptosis and ERS genes was investigated to clarify the molecular mechanisms of FGF8b. The results suggest that FGF8b exerts neuroprotective effects by alleviating endoplasmic reticulum (ER) stress during PD. These results suggest that FGF8b may be a promising candidate therapeutic drug for neurodegenerative diseases related to ER stress.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Baculoviridae/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromatografia de Afinidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/isolamento & purificação , Humanos , Camundongos , Células NIH 3T3 , Fármacos Neuroprotetores/isolamento & purificação , Células PC12 , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Sais de Tetrazólio , Tiazóis
4.
Hum Mol Genet ; 24(15): 4185-97, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25911675

RESUMO

The p63 transcription factor, homolog to the p53 tumor suppressor gene, plays a crucial role in epidermal and limb development, as its mutations are associated to human congenital syndromes characterized by skin, craniofacial and limb defects. While limb and skin-specific p63 transcriptional targets are being discovered, little is known of the post-translation modifications controlling ΔNp63α functions. Here we show that the p300 acetyl-transferase physically interacts in vivo with ΔNp63α and catalyzes its acetylation on lysine 193 (K193) inducing ΔNp63α stabilization and activating specific transcriptional functions. Furthermore we show that Fibroblast Growth Factor-8 (FGF8), a morphogenetic signaling molecule essential for embryonic limb development, increases the binding of ΔNp63α to the tyrosine kinase c-Abl as well as the levels of ΔNp63α acetylation. Notably, the natural mutant ΔNp63α-K193E, associated to the Split-Hand/Foot Malformation-IV syndrome, cannot be acetylated by this pathway. This mutant ΔNp63α protein displays promoter-specific loss of DNA binding activity and consequent altered expression of development-associated ΔNp63α target genes. Our results link FGF8, c-Abl and p300 in a regulatory pathway that controls ΔNp63α protein stability and transcriptional activity. Hence, limb malformation-causing p63 mutations, such as the K193E mutation, are likely to result in aberrant limb development via the combined action of altered protein stability and altered promoter occupancy.


Assuntos
Anormalidades Congênitas/genética , Fator 8 de Crescimento de Fibroblasto/genética , Proteínas Proto-Oncogênicas c-abl/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Fatores de Transcrição de p300-CBP/genética , Animais , Linhagem Celular , Anormalidades Congênitas/embriologia , Anormalidades Congênitas/patologia , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/patologia , Camundongos , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-abl/biossíntese , Proteínas Proto-Oncogênicas c-abl/metabolismo , Transdução de Sinais , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/metabolismo , Fatores de Transcrição de p300-CBP/biossíntese , Fatores de Transcrição de p300-CBP/metabolismo
5.
Dev Biol ; 387(1): 37-48, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24424161

RESUMO

Isl1 expression marks progenitor populations in developing embryos. In this study, we investigated the contribution of Isl1-expressing cells that utilize the ß-catenin pathway to skeletal development. Inactivation of ß-catenin in Isl1-expressing cells caused agenesis of the hindlimb skeleton and absence of the lower jaw (agnathia). In the hindlimb, Isl1-lineages broadly contributed to the mesenchyme; however, deletion of ß-catenin in the Isl1-lineage caused cell death only in a discrete posterior domain of nascent hindlimb bud mesenchyme. We found that the loss of posterior mesenchyme, which gives rise to Shh-expressing posterior organizer tissue, caused loss of posterior gene expression and failure to expand chondrogenic precursor cells, leading to severe truncation of the hindlimb. In facial tissues, Isl1-expressing cells broadly contributed to facial epithelium. We found reduced nuclear ß-catenin accumulation and loss of Fgf8 expression in mandibular epithelium of Isl1(-/-) embryos. Inactivating ß-catenin in Isl1-expressing epithelium caused both loss of epithelial Fgf8 expression and death of mesenchymal cells in the mandibular arch without affecting epithelial proliferation and survival. These results suggest a Isl1→ß-catenin→Fgf8 pathway that regulates mesenchymal survival and development of the lower jaw in the mandibular epithelium. By contrast, activating ß-catenin signaling in Isl1-lineages caused activation of Fgf8 broadly in facial epithelium. Our results provide evidence that, despite its broad contribution to hindlimb mesenchyme and facial epithelium, the Isl1-ß-catenin pathway regulates skeletal development of the hindlimb and lower jaw through discrete populations of cells that give rise to Shh-expressing posterior hindlimb mesenchyme and Fgf8-expressing mandibular epithelium.


Assuntos
Membro Posterior/embriologia , Anormalidades Maxilomandibulares/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Osteogênese/genética , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Região Branquial/embriologia , Linhagem da Célula/genética , Proliferação de Células , Sobrevivência Celular , Regulação para Baixo , Fosfatase 6 de Especificidade Dupla/biossíntese , Embrião de Mamíferos/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/deficiência , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Membro Posterior/anormalidades , Proteínas de Homeodomínio/biossíntese , Anormalidades Maxilomandibulares/genética , Fatores de Transcrição Kruppel-Like/biossíntese , Proteínas com Homeodomínio LIM/genética , Mandíbula/embriologia , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese , Transdução de Sinais/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Regulação para Cima , Proteína Gli3 com Dedos de Zinco , beta Catenina/genética
6.
Development ; 141(1): 148-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24284205

RESUMO

Non-canonical Wnt/planar cell polarity (PCP) signaling plays a primary role in the convergent extension that drives neural tube closure and body axis elongation. PCP signaling gene mutations cause severe neural tube defects (NTDs). However, the role of canonical Wnt/ß-catenin signaling in neural tube closure and NTDs remains poorly understood. This study shows that conditional gene targeting of ß-catenin in the dorsal neural folds of mouse embryos represses the expression of the homeobox-containing genes Pax3 and Cdx2 at the dorsal posterior neuropore (PNP), and subsequently diminishes the expression of the Wnt/ß-catenin signaling target genes T, Tbx6 and Fgf8 at the tail bud, leading to spina bifida aperta, caudal axis bending and tail truncation. We demonstrate that Pax3 and Cdx2 are novel downstream targets of Wnt/ß-catenin signaling. Transgenic activation of Pax3 cDNA can rescue the closure defect in the ß-catenin mutants, suggesting that Pax3 is a key downstream effector of ß-catenin signaling in the PNP closure process. Cdx2 is known to be crucial in posterior axis elongation and in neural tube closure. We found that Cdx2 expression is also repressed in the dorsal PNPs of Pax3-null embryos. However, the ectopically activated Pax3 in the ß-catenin mutants cannot restore Cdx2 mRNA in the dorsal PNP, suggesting that the presence of both ß-catenin and Pax3 is required for regional Cdx2 expression. Thus, ß-catenin signaling is required for caudal neural tube closure and elongation, acting through the transcriptional regulation of key target genes in the PNP.


Assuntos
Padronização Corporal/fisiologia , Proteínas de Homeodomínio/metabolismo , Tubo Neural/embriologia , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Animais , Padronização Corporal/genética , Fator de Transcrição CDX2 , Adesão Celular/genética , Polaridade Celular/fisiologia , Fator 8 de Crescimento de Fibroblasto/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Fator de Transcrição MSX1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Neurulação , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição Box Pareados/genética , Disrafismo Espinal/genética , Proteínas com Domínio T , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/genética
7.
Cell Mol Life Sci ; 68(9): 1569-79, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21207098

RESUMO

Fifty years ago, prescription of the sedative thalidomide caused a worldwide epidemic of multiple birth defects. The drug is now used in the treatment of leprosy and multiple myeloma. However, its use is limited due to its potent teratogenic activity. The mechanism by which thalidomide causes limb malformations and other developmental defects is a long-standing question. Multiple hypotheses exist to explain the molecular mechanism of thalidomide action. Among them, theories involving oxidative stress and anti-angiogenesis have been widely supported. Nevertheless, until recently, the direct target of thalidomide remained elusive. We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Our data suggest that thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting its ubiquitin ligase activity. In this review, we summarize the biology of thalidomide, focusing on the molecular mechanisms of its teratogenic effects. In addition, we discuss the questions still to be addressed.


Assuntos
Anormalidades Induzidas por Medicamentos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Estresse Oxidativo , Peptídeo Hidrolases/metabolismo , Teratogênicos/toxicidade , Talidomida/toxicidade , Proteínas Adaptadoras de Transdução de Sinal , Animais , Embrião de Galinha , Fator 8 de Crescimento de Fibroblasto/biossíntese , Humanos , Estresse Oxidativo/efeitos dos fármacos , Coelhos , Especificidade da Espécie , Teratogênicos/química , Teratogênicos/farmacocinética , Talidomida/química , Talidomida/farmacocinética , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Peixe-Zebra
8.
BMC Cancer ; 10: 596, 2010 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-21034500

RESUMO

BACKGROUND: Prostate tumours are commonly poorly oxygenated which is associated with tumour progression and development of resistance to chemotherapeutic drugs and radiotherapy. Fibroblast growth factor 8b (FGF8b) is a mitogenic and angiogenic factor, which is expressed at an increased level in human prostate tumours and is associated with a poor prognosis. We studied the effect of FGF8b on tumour oxygenation and growth parameters in xenografts in comparison with vascular endothelial growth factor (VEGF)-expressing xenografts, representing another fast growing and angiogenic tumour model. METHODS: Subcutaneous tumours of PC-3 cells transfected with FGF8b, VEGF or empty (mock) vectors were produced and studied for vascularity, cell proliferation, glucose metabolism and oxygenation. Tumours were evaluated by immunohistochemistry (IHC), flow cytometry, use of radiolabelled markers of energy metabolism ([18F]FDG) and hypoxia ([18F]EF5), and intratumoral polarographic measurements of pO2. RESULTS: Both FGF8b and VEGF tumours grew rapidly in nude mice and showed highly vascularised morphology. Perfusion studies, pO2 measurements, [18F]EF5 and [18F]FDG uptake as well as IHC staining for glucose transport protein (GLUT1) and hypoxia inducible factor (HIF) 1 showed that VEGF xenografts were well-perfused and oxygenised, as expected, whereas FGF8b tumours were as hypoxic as mock tumours. These results suggest that FGF8b-induced tumour capillaries are defective. Nevertheless, the growth rate of hypoxic FGF8b tumours was highly increased, as that of well-oxygenised VEGF tumours, when compared with hypoxic mock tumour controls. CONCLUSION: FGF8b is able to induce fast growth in strongly hypoxic tumour microenvironment whereas VEGF-stimulated growth advantage is associated with improved perfusion and oxygenation of prostate tumour xenografts.


Assuntos
Fator 8 de Crescimento de Fibroblasto/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/metabolismo , Animais , Humanos , Hipóxia , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oxigênio/química , Oxigênio/metabolismo , Prognóstico , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Dent Res ; 89(9): 909-14, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20530729

RESUMO

beta-Catenin signaling is required for embryonic tooth morphogenesis and promotes continuous tooth development when activated in embryos. To determine whether activation of this pathway in the adult oral cavity could promote tooth development, we induced mutation of epithelial beta-catenin to a stabilized form in adult mice. This caused increased proliferation of the incisor tooth cervical loop, outpouching of incisor epithelium, abnormal morphology of the epithelial-mesenchymal junction, and enhanced expression of genes associated with embryonic tooth development. Ectopic dental-like structures were formed from the incisor region following implantation into immunodeficient mice. Thus, forced activation of beta-catenin signaling can initiate an embryonic-like program of tooth development in adult rodent incisor teeth.


Assuntos
Células-Tronco Adultas/fisiologia , Papila Dentária/citologia , Órgão do Esmalte/citologia , Odontogênese/genética , beta Catenina/fisiologia , Animais , Células Epiteliais/citologia , Feminino , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/genética , Incisivo/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Nus , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Transdução de Sinais , Calcificação de Dente , Regulação para Cima
10.
Cancer Res ; 66(4): 2188-94, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16489020

RESUMO

Two commonly occurring genetic aberrations of human prostate cancer [i.e., overexpression of a mitogenic polypeptide (fibroblast growth factor 8, isoform b or FGF8b) and loss of function of PTEN tumor suppressor] were recapitulated into a new combinatorial mouse model. This model harboring the Fgf8b transgene and haploinsufficiency in Pten, both in a prostate epithelium-specific manner, yielded prostatic adenocarcinoma with readily detectable lymph node metastases, whereas single models with each of the defects were shown earlier to progress generally only up to prostatic intraepithelial neoplasia (PIN). In addition to late age-related development of typical adenocarcinoma, the model also displayed a low incidence of mucinous adenocarcinoma, a rare variant type of human prostatic adenocarcinoma. The cooperation between FGF8b activation and PTEN deficiency must be linked to acquisition of additional genetic alterations for the progression of the lesions to primary adenocarcinoma. Here, we identified loss of heterozygosity at the Pten gene leading to bialleic loss, as a necessary secondary event, indicating that a complete loss of PTEN function is required in the development of invasive cancer in the model. Analyses of expression of downstream mediators phospho-AKT (p-AKT) and p27(KIP1), in various types of lesions, however, revealed a complex picture. Although PIN lesions displayed relatively strong expression of p-AKT and p27(KIP1), there was a notable heterogeneity with variable decrease in their immunostaining in adenocarcinomas. Together, the results further underscore the notion that besides activation of AKT by loss of PTEN function, other PTEN-regulated pathways must be operative for progression of lesions from PIN to adenocarcinoma.


Assuntos
Adenocarcinoma/metabolismo , Fator 8 de Crescimento de Fibroblasto/biossíntese , PTEN Fosfo-Hidrolase/deficiência , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Alelos , Animais , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Inativação Gênica , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Metástase Neoplásica , PTEN Fosfo-Hidrolase/genética , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transgenes
11.
Oncogene ; 25(29): 4122-7, 2006 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-16474841

RESUMO

Fibroblast growth factors (FGF), and in particular FGF8, have been strongly implicated in prostate carcinogenesis. This study investigated the expression of Sef, a key inhibitory regulator of FGF signalling, in prostate cancer. In a panel of cell lines, hSef was detected in both androgen-dependent and independent cells but was significantly reduced in highly metastatic derivative clones. hSef expression was not influenced by androgenic stimulation. Forced downregulation of hSef by siRNA increased FGF8b induced cell migration (P=0.02) and invasion (P=0.007). Reduced hSef levels also enhanced FGF8b stimulated expression of MMP9 (P=0.005). mRNA in situ hybridization revealed hSef expression in 80% (8/10) of benign biopsies but in only 69% (23/33) of Gleason sum 4-7 and 35% (10/28) of Gleason sum 8-10 cancer biopsies (P=0.004). Quantitative PCR of microdissected glands confirmed this trend (P=0.001). hSef was expressed in 69% (27/39) of non-metastatic tumours but in only 18% (2/11) of metastatic tumours (P=0.004, n=50). hSef expression was next correlated with earlier data on FGF8b expression in a subgroup of cancers. In this cohort, 86% (19/22) of high-grade cancers expressed FGF8 but only 31% (7/22) expressed hSef. Positive FGF8 expression but a loss of hSef was observed in 88% (7/8) of metastatic tumours. In contrast, metastasis was evident in only 10% (1/10) of tumours, which co-expressed both FGF8 and hSef (P<0.001). These results suggest evidence that hSef is downregulated in advanced prostate cancer and might facilitate an enhanced tumorigenic response to FGFs. Further research into the role of hSef in cancer cell signalling and the mechanism of its downregulation may contribute to more effective targeting of growth factors in prostate cancer.


Assuntos
Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias da Próstata/genética , Receptores de Interleucina/genética , Transdução de Sinais/genética , Androgênios/metabolismo , Linhagem Celular Tumoral , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/genética , Perfilação da Expressão Gênica , Humanos , Masculino , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Reação em Cadeia da Polimerase , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Interleucina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA