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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 291(36): 18632-42, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27432882

RESUMEN

Parathyroid hormone (PTH) and FGF23 are the primary hormones regulating acute phosphate homeostasis. Human renal proximal tubule cells (RPTECs) were used to characterize the mechanism and signaling pathways of PTH and FGF23 on phosphate transport and the role of the PDZ protein NHERF1 in mediating PTH and FGF23 effects. RPTECs express the NPT2A phosphate transporter, αKlotho, FGFR1, FGFR3, FGFR4, and the PTH receptor. FGFR1 isoforms are formed from alternate splicing of exon 3 and of exon 8 or 9 in Ir-like loop 3. Exon 3 was absent, but mRNA containing both exons 8 and 9 is present in cytoplasm. Using an FGFR1c-specific antibody together with mass spectrometry analysis, we show that RPTECs express FGFR-ß1C. The data are consistent with regulated FGFR1 splicing involving a novel cytoplasmic mechanism. PTH and FGF23 inhibited phosphate transport in a concentration-dependent manner. At maximally effective concentrations, PTH and FGF23 equivalently decreased phosphate uptake and were not additive, suggesting a shared mechanism of action. Protein kinase A or C blockade prevented PTH but not FGF23 actions. Conversely, inhibiting SGK1, blocking FGFR dimerization, or knocking down Klotho expression disrupted FGF23 actions but did not interfere with PTH effects. C-terminal FGF23(180-251) competitively and selectively blocked FGF23 action without disrupting PTH effects. However, both PTH and FGF23-sensitive phosphate transport were abolished by NHERF1 shRNA knockdown. Extended treatment with PTH or FGF23 down-regulated NPT2A without affecting NHERF1. We conclude that FGFR1c and PTHR signaling pathways converge on NHERF1 to inhibit PTH- and FGF23-sensitive phosphate transport and down-regulate NPT2A.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Transducción de Señal/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Línea Celular Transformada , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Glucuronidasa/biosíntesis , Glucuronidasa/genética , Humanos , Proteínas Klotho , Hormona Paratiroidea/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética
2.
Tumour Biol ; 39(7): 1010428317712592, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28718374

RESUMEN

Ovarian cancer is the most lethal gynecologic malignancy, due to its high propensity for metastasis. Cancer-associated fibroblasts, as the dominant component of tumor microenvironment, are crucial for tumor progression. However, the mechanisms underlying the regulation of ovarian cancer cells by cancer-associated fibroblasts remain little known. Here, we first isolated cancer-associated fibroblasts from patients' ovarian tissues and found that cancer-associated fibroblasts promoted SKOV3 cells' proliferation, migration, and invasion. Fibroblast growth factor-1 was identified as a highly increased factor in cancer-associated fibroblasts compared with normal fibroblasts by quantitative reverse transcription polymerase chain reaction (~4.6-fold, p < 0.01) and ELISA assays (~4-fold, p < 0.01). High expression of fibroblast growth factor-1 in cancer-associated fibroblasts either naturally or through gene recombination led to phosphorylation of fibroblast growth factor receptor 4 in SKOV3 cells, which is followed by the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway and epithelial-to-mesenchymal transition-associated gene Snail1 and MMP3 expression. Moreover, treatment of SKOV3 cell with fibroblast growth factor receptor inhibitor PD173074 terminated cellular proliferation, migration, and invasion, reduced the phosphorylation level of fibroblast growth factor receptor 4, and suppressed the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway. In addition, the expression level of Snail1 and MMP3 was reduced, while the expression level of E-cadherin increased. These observations suggest a crucial role for cancer-associated fibroblasts and fibroblast growth factor-1/fibroblast growth factor receptor 4 signaling in the progression of ovarian cancer. Therefore, this fibroblast growth factor-1/fibroblast growth factor receptor 4 axis may become a potential target for the treatment of ovarian cancer.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/genética , Metaloproteinasa 3 de la Matriz/genética , Neoplasias Ováricas/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción de la Familia Snail/genética , Adulto , Anciano , Animales , Cadherinas/genética , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Factor 1 de Crecimiento de Fibroblastos/biosíntesis , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Metaloproteinasa 3 de la Matriz/biosíntesis , Persona de Mediana Edad , Invasividad Neoplásica/genética , Neoplasias Ováricas/patología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Transducción de Señal , Factores de Transcripción de la Familia Snail/biosíntesis
3.
Cells Tissues Organs ; 201(1): 26-37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26613250

RESUMEN

The Apert syndrome is a rare congenital disorder most often arising from S252W or P253R mutations in fibroblast growth factor receptor (FGFR2). Numerous studies have focused on the regulatory role of Apert FGFR2 signaling in bone formation, whereas its functional role in tooth development is largely unknown. To investigate the role of FGFR signaling in cell proliferation and odontogenic differentiation of human dental cells in vitro, we isolated dental pulp and enamel organ epithelia (EOE) tissues from an Apert patient carrying the S252W FGFR2 mutation. Apert primary pulp and EOE cells were established and shown to exhibit normal morphology and express alkaline phosphatase under differentiation conditions. Similar to control cells, Apert dental pulp and EOE cells expressed all FGFRs, with highest levels of FGFR1 followed by FGFR2 and low levels of FGFR3 and FGFR4. However, Apert cells had increased cell growth compared with control cells. Distinct from previous findings in osteoblast cells, gain-of-function S252W FGFR2 mutation did not upregulate the expression of epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFRα), but elevated extracellular signal-regulated kinase (ERK) signaling in cells after EGF stimulation. Unexpectedly, there was little effect of the S252W mutation on odontogenic gene expression in dental pulp and EOE cells. However, after inhibition of total FGFR signaling or ERK signaling, the expression of odontogenic genes was upregulated in both dental cell types, indicating the negative effect of whole FGFR signaling on odontogenic differentiation. This study provides novel insights on FGFR signaling and a common Apert FGFR2 mutation in the regulation of odontogenic differentiation of dental mesenchymal and epithelial cells.


Asunto(s)
Acrocefalosindactilia/genética , Pulpa Dental/citología , Órgano del Esmalte/citología , Odontogénesis/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Diente/embriología , Fosfatasa Alcalina/biosíntesis , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Receptores ErbB/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Masculino , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal
4.
Growth Factors ; 33(5-6): 393-400, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26768755

RESUMEN

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are increasingly recognized as important regulators of embryo development in mammals. This study investigated the importance of FGF signaling during in vitro development of ovine embryo. The mRNAs of four FGFR subtypes were detected throughout preimplantation development of in vitro fertilized (IVF) embryos, peaked in abundance at the morula stage, and decreased significantly at the blastocyst stage. To gain insight into the role of these mRNAs in embryo development, IVF embryos were cultured in the presence of FGF2 (100 or 500 ng/ml: beginning from days 1 or 4 to 7) or PD173074 (1 µM: beginning from days 1 to 7) as usual treatments for activation or inhibition of FGFRs, respectively. FGF2-supplementation did not affect the percentage of embryos that developed to the blastocyst, blastocyst cell count and the proportion of cells allocated in inner cell mass (ICM) and trophectoderm (TE) compared to control (p > 0.05). Also, increasing the dosage or duration of FGF2 treatment did not significantly alter blastocyst yield or differential cell count (p > 0.05). PD173074-mediated inhibition of FGFRs did not significantly affect blastocyst yield (p > 0.05). Assessment of expression profiles of lineage-associated markers revealed that FGF2 (500 ng/ml) supplementation: (i) significantly increased expression of putative hypoblast marker (GATA4), (ii) significantly decreased expression of putative epiblast (EPI) marker (NANOG) and (iii) did not change TE markers (CDX2 and IFNT) and pluripotency makers (OCT4, SOX2 and REX1). In summary, FGF2-mediated activation of FGFRs may promote a switch in transcriptional profile of ovine ICM from EPI- to hypoblast-associated gene expression.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Pirimidinas/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Animales , Blastocisto/metabolismo , Femenino , Factor de Transcripción GATA4/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/biosíntesis , Masculino , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Ovinos
5.
Tumour Biol ; 36(12): 9677-83, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26152288

RESUMEN

Invasive ductal carcinoma of the breast is the most common cancer affecting women worldwide. The marked heterogeneity of breast cancer is matched only with the heterogeneity in its associated or causative factors. Breast cancer in Saudi Arabia is apparently an early onset with many of the affected females diagnosed before they reach the age of 50 years. One possible rationale underlying this observation is that consanguinity, which is widely spread in the Saudi community, is causing the accumulation of yet undetermined cancer susceptibility mutations. Another factor could be the accumulation of epigenetic aberrations caused by the shift toward a Western-like lifestyle in the past two decades. In order to shed some light into the molecular mechanisms underlying breast cancer in the Saudi community, we identified KLOTHO (KL) as a tumor-specific methylated gene using genome-wide methylation analysis of primary breast tumors utilizing the MBD-seq approach. KL methylation was frequent as it was detected in 55.3 % of breast cancer cases from Saudi Arabia (n = 179) using MethyLight assay. Furthermore, KL is downregulated in breast tumors with its expression induced following treatment with 5-azacytidine. The involvement of KL in breast cancer led us to investigate its relationship in the context of breast cancer, with one of the protagonists of its function, fibroblast growth factor receptor 4 (FGFR4). Overexpression of FGFR4 in breast cancer is frequent in our cohort and this overexpression is associated with poor overall survival. Interestingly, FGFR4 expression is higher in the absence of KL methylation and lower when KL is methylated and presumably silenced, which is suggestive of an intricate relationship between the two factors. In conclusion, our findings further implicate "metabolic" genes or pathways in breast cancer that are disrupted by epigenetic mechanisms and could provide new avenues for understanding this disease in a new context.


Asunto(s)
Carcinoma Ductal de Mama/genética , Factores de Crecimiento de Fibroblastos/biosíntesis , Glucuronidasa/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Islas de CpG , Metilación de ADN/genética , Epigénesis Genética , Femenino , Factores de Crecimiento de Fibroblastos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Klotho , Persona de Mediana Edad , Regiones Promotoras Genéticas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética
6.
Gastroenterology ; 139(5): 1549-58, 1558.e1, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20691689

RESUMEN

BACKGROUND & AIMS: Sodium chenodeoxycholate (CDC) accelerates colonic transit in health. Our aim was to examine pharmacodynamics (colonic transit, bowel function) and pharmacogenetics of CDC in constipation-predominant irritable bowel syndrome (IBS-C). METHODS: In a double-blind placebo-controlled study, 36 female patients with IBS-C were randomized to treatment with delayed-release oral formulations of placebo, 500 mg CDC, or 1000 mg CDC for 4 days. We assessed gastrointestinal and colonic transit, stool characteristics, and associations of transit with fasting serum 7αC4 (surrogate of bile acid synthesis) and FGF19 (negative regulator of bile acid synthesis) levels. Candidate genetic polymorphisms involved in regulation of bile acid synthesis were analyzed in the 36 patients with IBS-C and 57 healthy volunteers to assess genetic influence on effects of CDC on transit. RESULTS: Overall colonic transit and ascending colon emptying (AC t(½)) were significantly accelerated in the CDC group compared with placebo (P = .005 and P = .028, respectively). Looser stool consistency (P = .003), increased stool frequency (P = .018), and greater ease of passage (P = .024) were noted with CDC compared with placebo. The most common side effect was lower abdominal cramping/pain (P = .01). Fasting serum 7αC4 (but not FGF19) was positively associated with colonic transit (r(s) = 0.749, P = .003, placebo group). Genetic variation in FGFR4 was associated with AC t(½) in response to CDC (uncorrected P = .015); αKlothoß variant showed a gene-by-treatment interaction based on patient subgroup (uncorrected P = .0088). CONCLUSIONS: CDC accelerates colonic transit and improves bowel function in female patients with IBS-C. The rate of bile acid synthesis influences colonic transit. Genetic variation in negative feedback inhibition of bile acid synthesis may affect CDC-mediated acceleration of colonic transit.


Asunto(s)
Ácido Quenodesoxicólico/farmacocinética , Fármacos Gastrointestinales/farmacocinética , Tránsito Gastrointestinal/fisiología , Síndrome del Colon Irritable/tratamiento farmacológico , Administración Oral , Adulto , Ácidos y Sales Biliares/biosíntesis , Ácido Quenodesoxicólico/administración & dosificación , Colestenonas/metabolismo , Cromatografía Líquida de Alta Presión , ADN/genética , Preparaciones de Acción Retardada , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Ensayo de Inmunoadsorción Enzimática , Femenino , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Estudios de Seguimiento , Fármacos Gastrointestinales/administración & dosificación , Tránsito Gastrointestinal/efectos de los fármacos , Frecuencia de los Genes , Variación Genética , Genotipo , Humanos , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/metabolismo , Espectrometría de Masas , Persona de Mediana Edad , Polimorfismo Genético , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Estudios Retrospectivos , Resultado del Tratamiento
7.
Front Endocrinol (Lausanne) ; 12: 795116, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956100

RESUMEN

Adrenocortical carcinoma (ACC) is a rare endocrine malignancy and treatment of advanced disease is challenging. Clinical trials with multi-tyrosine kinase inhibitors in the past have yielded disappointing results. Here, we investigated fibroblast growth factor (FGF) receptors and their pathways in adrenocortical tumors as potential treatment targets. We performed real-time RT-PCR of 93 FGF pathway related genes in a cohort of 39 fresh frozen benign and malignant adrenocortical, 9 non-adrenal tissues and 4 cell lines. The expression of FGF receptors was validated in 166 formalin-fixed paraffin embedded (FFPE) tissues using RNA in situ hybridization (RNAscope) and correlated with clinical data. In malignant compared to benign adrenal tumors, we found significant differences in the expression of 16/94 FGF receptor pathway related genes. Genes involved in tissue differentiation and metastatic spread through epithelial to mesechymal transition were most strongly altered. The therapeutically targetable FGF receptors 1 and 4 were upregulated 4.6- and 6-fold, respectively, in malignant compared to benign adrenocortical tumors, which was confirmed by RNAscope in FFPE samples. High expression of FGFR1 and 4 was significantly associated with worse patient prognosis in univariate analysis. After multivariate adjustment for the known prognostic factors Ki-67 and ENSAT tumor stage, FGFR1 remained significantly associated with recurrence-free survival (HR=6.10, 95%CI: 1.78 - 20.86, p=0.004) and FGFR4 with overall survival (HR=3.23, 95%CI: 1.52 - 6.88, p=0.002). Collectively, our study supports a role of FGF pathways in malignant adrenocortical tumors. Quantification of FGF receptors may enable a stratification of ACC for the use of FGFR inhibitors in future clinical trials.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Carcinoma Corticosuprarrenal/metabolismo , Biomarcadores de Tumor/biosíntesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/mortalidad , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/mortalidad , Adulto , Biomarcadores de Tumor/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Tasa de Supervivencia/tendencias
8.
Cells ; 8(9)2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31527546

RESUMEN

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.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adenocarcinoma/diagnóstico , Adenocarcinoma/patología , Anciano , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/patología , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Tasa de Supervivencia
9.
Oncogene ; 37(41): 5477-5491, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29884889

RESUMEN

Metastatic colorectal cancer (CRC) is one of the most common causes of cancer death worldwide; however, the molecular mechanism underlying CRC metastasis remains unknown. Using an integrated approach, we identified forkhead box C1 (FOXC1) as a novel regulator of CRC metastasis. Elevated expression of FOXC1 is significantly correlated with metastasis, recurrence and reduced survival. FOXC1 overexpression promotes CRC invasion and lung metastasis, whereas FOXC1 knockdown has the opposite effect. In addition, FOXC1 directly binds its target genes integrin α7 (ITGA7) and fibroblast growth factor receptor 4 (FGFR4) and activates their expression. Genetic epistasis analysis confirmed that ITGA7 and FGFR4 act downstream of FOXC1. Furthermore, pharmaceutical inhibition of FGFR4 can reverse CRC metastasis mediated by FOXC1 overexpression. These results suggest that FOXC1 is a prognostic biomarker in CRC patients and targeting the FGFR4 signaling pathway may provide a promising strategy for the treatment of FOXC1-driven CRC metastasis.


Asunto(s)
Antígenos CD/biosíntesis , Neoplasias Colorrectales/patología , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Cadenas alfa de Integrinas/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Anciano , Animales , Antígenos CD/genética , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Femenino , Xenoinjertos , Humanos , Cadenas alfa de Integrinas/genética , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Activación Transcripcional
10.
Oncotarget ; 8(24): 38592-38601, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28445152

RESUMEN

BACKGROUND: The fibroblast growth factor receptor 4 (FGFR4) pathway is an essential regulatory component of bile acid synthesis, and its relationship with hepatocellular carcinoma (HCC) has been reported. We investigated the gene expression and clinical significance of FGFR4 and related pathways in intrahepatic cholangiocarcinoma (iCCA). RESULTS: The median age was 56 years (range 30-78) and 34 patients (74%) were male. Six patients (13%) had hepatitis B virus infection, with or without liver cirrhosis. Overall survival was significantly associated with FGFR4 (p = 0.004), FGF19 (p = 0.047), FGF21 (p = 0.04), and KLB (p = 0.03) expression. In the multivariate analysis with potential prognostic factors, high expression of FGF19, FGF21, and FGFR4 was significantly associated with better survival. In the analysis using the TCGA iCCA dataset, mRNA overexpression of at least 1 of the FGFR4-related genes was significantly associated with better disease-free survival (p = 0.02). MATERIALS AND METHODS: We assessed the expression of 98 genes in formalin-fixed paraffin embedded tumor tissue specimens from 46 patients with surgically resected iCCA using a NanoString platform. This included 10 FGF pathway genes (e.g. FGFR1-4, KLB, FGF3, 4, 19, 21, and 23), 19 distal marker genes (e.g. CYP7A1 and CYP17A1), 31 genes relevant to HCC and iCCA (e.g. AFP, TS), 18 copy number variation matched genes, and 20 control genes. Log-transformation of gene expression was performed for normalization and statistical analysis. Overall survival was correlated with gene expression (< median vs. ≥ median) using a log-rank test. The prognostic impact of FGFR4-related genes was validated using the public TCGA dataset for iCCA. CONCLUSIONS: Our results indicate that mRNA expression of FGFR4-related genes may be a biomarker to define the distinctive molecular phenotype of iCCA. Future preclinical and clinical validation is required to define the role of the FGFR4 pathway in iCCA.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Biomarcadores de Tumor/análisis , Colangiocarcinoma/patología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adulto , Anciano , Neoplasias de los Conductos Biliares/mortalidad , Colangiocarcinoma/mortalidad , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/análisis , Transcriptoma
11.
J Oral Sci ; 48(1): 9-14, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16617195

RESUMEN

A number of fibroblast growth factors (FGFs) are involved in regulatory mechanisms of the salivary gland development. However, the role of FGF-6 unique in myogenic cells has not been elucidated in the developing sublingual gland. In the present study, temporo-spatial expression of FGF-6 and its receptor (FGFR)-4, in conjunction with some related histo-chemical properties, were investigated in the sublingual gland of the prenatal and early postnatal mice. The earliest expression of both FGF-6 and FGFR-4 was detected in immature acinar cells at gestational day 17 (GD17). The staining intensity increased gradually and some acinar cells showed a distinct staining at postnatal day 0 (PD0). The immunopositive cells had a relatively round profile and were assumed to be acinar cells. The positive staining decreased thereafter and disappeared completely by PD11. To confirm the identity of cells positive for FGF-6, double immunolabeling with anti-alphasmooth muscle actin (alphaSMA) and anti-FGF-6 antibodies was performed. The positive staining of alphaSMA, a marker of myoepithelial cells, was detected in the flattened cells surrounding the acini but not in the cells positive for FGF-6. The staining properties of secretory granules in acinar cells were also examined with periodic acid-Shiff (PAS) and alcian blue (AB). PAS-positive granules abundant in the late gestational stages (GD17 to PD0) began to be replaced with AB-positive mucous granules at early neonatal days (PD0-3), when the FGF-6/FGFR-4 expression was the strongest. These findings suggest that FGF-6/FGFR-4 might be involved in the changes of secretory granule content of acinar cells in the sublingual gland during the late gestational and early neonatal stages.


Asunto(s)
Factor 6 de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Glándula Sublingual/química , Glándula Sublingual/embriología , Animales , Animales Lactantes/metabolismo , Gránulos Citoplasmáticos/química , Factor 6 de Crecimiento de Fibroblastos/análisis , Regulación del Desarrollo de la Expresión Génica , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/análisis , Glándula Sublingual/citología , Glándula Sublingual/crecimiento & desarrollo
12.
Oncotarget ; 7(16): 22234-44, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-26993773

RESUMEN

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNST) are rare, highly malignant, and poorly understood sarcomas. The often poor outcome of MPNST highlights the necessity of identifying prognostic predictors for this aggressive sarcoma. Here, we investigate the role of fibroblast growth factor receptor (FGFR) family members in human MPNSTs. RESULTS: aCGH and bioinformatics analysis identified frequent amplification of the FGFR1 gene. FISH analysis revealed that 26.9% MPNST samples had amplification of FGFR1, with both focal and polysomy patterns observed. IHC identified that FGFR1 protein expression was positively correlated with FGFR1 gene amplification. High expression of FGFR1 protein was associated with better overall survival (OS) and was an independent prognostic predictor for OS of MPNST patients. Additionally, combined expression of FGFR1 and FGFR2 protein characterized a subtype of MPNST with better OS. FGFR4 protein was expressed 82.3% of MPNST samples, and was associated with poor disease-free survival. MATERIALS AND METHODS: We performed microarray-based comparative genomic hybridization (aCGH) profiling of two cohorts of primary MPNST tissue samples including 25 patients treated at The University of Texas MD Anderson Cancer Center and 26 patients from Tianjin Medical University Cancer Institute and Hospital. Fluorescence in situ hybridization (FISH) was used to validate the gene amplification detected by aCGH analysis. Another cohort of 63 formalin-fixed paraffin-embedded MPNST samples (including 52 samples for FISH assay) was obtained to explore FGFR1, 2, 3, and 4 protein expression by immunohistochemical (IHC) analysis. CONCLUSIONS: Our integrated genomic and molecular studies provide evidence that FGFRs play different prognostic roles in MPNST.


Asunto(s)
Biomarcadores de Tumor/análisis , Neurilemoma/patología , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neurilemoma/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Adulto Joven
13.
Cancer Discov ; 3(3): 280-93, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23384775

RESUMEN

Protein kinases represent the most effective class of therapeutic targets in cancer; therefore, determination of kinase aberrations is a major focus of cancer genomic studies. Here, we analyzed transcriptome sequencing data from a compendium of 482 cancer and benign samples from 25 different tissue types, and defined distinct "outlier kinases" in individual breast and pancreatic cancer samples, based on highest levels of absolute and differential expression. Frequent outlier kinases in breast cancer included therapeutic targets like ERBB2 and FGFR4, distinct from MET, AKT2, and PLK2 in pancreatic cancer. Outlier kinases imparted sample-specific dependencies in various cell lines, as tested by siRNA knockdown and/or pharmacologic inhibition. Outlier expression of polo-like kinases was observed in a subset of KRAS-dependent pancreatic cancer cell lines, and conferred increased sensitivity to the pan-PLK inhibitor BI-6727. Our results suggest that outlier kinases represent effective precision therapeutic targets that are readily identifiable through RNA sequencing of tumors.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/biosíntesis , Análisis de Secuencia de ARN/métodos , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Interferencia de ARN , Distribución Aleatoria , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/biosíntesis , Proteínas ras/genética
14.
Oncol Rep ; 30(6): 2777-84, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24126887

RESUMEN

Our previous findings revealed that FGFR4 may be a novel therapeutic target for gastric cancer. The aim of the present study was to explore the effects of a combination of PD173074 (PD) and 5-fluorouracil (5-Fu) on the biological behavior of gastric cancer cell lines and the relevant mechanisms involved. MKN45, a gastric cancer cell line, was treated with each single agent alone or a combination of FGF19, PD and 5-Fu. Then, a series of functional assays were performed using CCK-8 assay and flow cytometry. Western blot analysis was used to determine the expression of signaling pathway and downstream-related molecules in the MKN45 cells following the different treatments. As the concentration of PD and 5-Fu increased, the cell viability gradually decreased; the viability of the combination group was less than the viability following single administration. Western blot analysis showed that FGFR4 expression was weak in the 5-Fu-treated groups when compared with the control. PD markedly increased the apoptosis rate of MKN45 cells when compared to the control; the apoptosis rate in the cells treated with the combination of PD and 5-Fu was higher than that in the cells following single treatment. Furthermore, PD reduced the expression of p-ERK and Bcl-xl and increased caspase-3 expression. Inhibition of the activity of FGFR4 may be the main mechanisms of PD effect while 5-Fu reduced FGFR4 expression. Furthermore, the effects of the combination of 5-Fu and PD in inhibiting proliferation, increasing apoptosis and arresting cell cycle were superior to these effects following the single agent treatments, suggesting that the two drugs applied in combination may contribute to the effective treatment of gastric cancer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fluorouracilo/farmacología , Pirimidinas/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína bcl-X/biosíntesis
15.
Cancer Res ; 72(22): 5767-77, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22971346

RESUMEN

A gly(388)arg polymorphism (rs351855) in the transmembrane domain of the fibroblast growth factor receptor (FGFR4) is associated with increased risk, staging, and metastasis in several different types of cancer. To specifically assess the impact of the polymorphic FGFR4 in colorectal cancer (CRC), we engineered CRC cell lines with distinct endogenous expression patterns to overexpress either the FGFR4(gly) or FGFR4(arg) alleles. The biologic analyses revealed an oncogenic importance for both polymorphic alleles, but FGFR4(gly) was the stronger inducer of tumor growth, whereas FGFR4(arg) was the stronger inducer of migration. An evaluation of clinical specimens revealed that FGFR4 was upregulated in 20/71 patients independent of gly(388)arg status. There was no correlation between the presence of an FGFR4(arg) allele and CRC or polyp risk in 3,471 participants of the CORSA study. However, among 182 patients with CRC, FGFR4(arg)-carriers had a fivefold higher risk of tumors that were stage II or greater. Together, our results established that both allelic forms of FGFR4 exert an oncogenic impact and may serve equally well as therapeutic targets in CRC. One important implication of our findings is that FGFR4(arg)-carriers are at a higher risk for more aggressive tumors and therefore may profit from early detection measures.


Asunto(s)
Alelos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Anciano , Anciano de 80 o más Años , Células CACO-2 , Procesos de Crecimiento Celular/genética , Neoplasias Colorrectales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Genotipo , Células HCT116 , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Polimorfismo de Nucleótido Simple , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis
16.
Int J Oncol ; 38(1): 133-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21109934

RESUMEN

The overexpression of fibroblast growth factor receptor (FGFR) 4 has been reported in various human cancers, but it has not been studied in pancreatic ductal adenocarcinoma (PDAC) or its precursor lesion, pancreatic intraepithelial neoplasia (PanIN). Moreover, there is controversy as to whether FGFR4 has a mitogenic role in carcinogenesis or other functions. Therefore, the expression and roles of FGFR4 in pancreatic cancer were investigated. Immunohistochemical staining was performed using an anti-FGFR4 antibody in PDAC and PanIN cases. The expression levels of FGFR4 mRNA and protein were investigated in PDAC cell lines by qRT-PCR and Western blot, respectively. Changes were analyzed in cell morphology, proliferation, migration, invasion and attachment in PDAC cell lines with or without the stimulation of FGFR4 by FGF19, as a known specific ligand. The changes in mRNA levels associated with transformation and tumorigenesis as a result of FGF19 administration were also evaluated. FGFR4 was expressed in 39 of 53 PDAC cases (73.6%) and its expression tended to be related to longer overall survival (P=0.068). Moreover, it was frequently expressed in high-grade PanIN lesions [10 of 11 lesions (90.9%)], whereas it was hardly expressed in low-grade PanIN lesions [1 of 10 lesions (10.0%)] (P=0.0003). FGFR4 stimulation of PDAC cells resulted in significantly increased cell adhesion to laminin and fibronectin (P<0.05) and decreased cell migration (P<0.05). The results of PCR array analysis indicated that this was a result of up-regulation of the integrin α4 family. In contrast, cell morphology or proliferation in PDAC cells was not affected. We showed that FGFR4 expression is markedly increased in high-grade PanIN and PDAC compared with that in normal and low-grade PanIN, and that FGFR4 stimulation by FGF19 of PDAC cells contributes to tumor suppression by increasing cell adhesion to extracellular matrix.


Asunto(s)
Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Anciano , Anciano de 80 o más Años , Carcinoma in Situ/genética , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal , Tasa de Supervivencia , Regulación hacia Arriba
17.
Oncogene ; 27(1): 85-97, 2008 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17599042

RESUMEN

Although fibroblast growth factor 19 (FGF19) can promote liver carcinogenesis in mice its involvement in human cancer is not well characterized. Here we report that FGF19 and its cognate receptor FGF receptor 4 (FGFR4) are coexpressed in primary human liver, lung and colon tumors and in a subset of human colon cancer cell lines. To test the importance of FGF19 for tumor growth, we developed an anti-FGF19 monoclonal antibody that selectively blocks the interaction of FGF19 with FGFR4. This antibody abolished FGF19-mediated activity in vitro and inhibited growth of colon tumor xenografts in vivo and effectively prevented hepatocellular carcinomas in FGF19 transgenic mice. The efficacy of the antibody in these models was linked to inhibition of FGF19-dependent activation of FGFR4, FRS2, ERK and beta-catenin. These findings suggest that the inactivation of FGF19 could be beneficial for the treatment of colon cancer, liver cancer and other malignancies involving interaction of FGF19 and FGFR4.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Marcación de Gen/métodos , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Anticuerpos Monoclonales/uso terapéutico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/inmunología , Células HCT116 , Células HT29 , Humanos , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Trasplante Heterólogo
18.
Dev Biol ; 302(2): 536-52, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17123506

RESUMEN

During early vertebrate development Fibroblast Growth Factor (FGF) signalling is required for multiple activities including specification of mesodermal, neural and heart tissue, as well as gastrulation movements and regulation of differentiation and pattern onset in the extending body axis. A current challenge is to understand how FGF signalling generates such diverse outcomes. A key FGF downstream pathway is the Ras-MAPK/Erk1/2 cascade, which culminates in the phosphorylation of target proteins, such as the Ets family of transcription factors. To begin to assess specificity downstream of FGF in the chick embryo we have characterised the patterns of Fgfr1-4 expression and Erk1/2 activation, as well as expression of the Erk1/2 specific phosphatase, Mkp3 and of three Ets factor genes (Erm, Pea3 and Er81) from early blastula to the 10 somite stage. We identify new sites of Fgfr expression and show that nearly all regions of Erk1/2 activity are within Fgfr expression domains and require FGF signalling. Differences in intensity, duration, distribution and sub-cellular localisation of activated Erk1/2 are observed in distinct cell populations within the embryo and during wound healing. With few exceptions, a tight correspondence between Erk1/2 activation and Mkp3 expression is found, while specific combinations of Ets factors are associated with distinct regions of Erk1/2 activation. These findings provide a comprehensive spatial and temporal map of FGF/Erk1/2 activity during early chick development and identify region and tissue specific differences in expression of Fgfrs as well as Erk1/2 phosphorylation and transcriptional targets which help to define response specificity.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Animales , Embrión de Pollo , Fosfatasa 6 de Especificidad Dual , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/biosíntesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Transducción de Señal , Factores de Transcripción/biosíntesis
19.
Endocrine ; 28(1): 43-7, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16311409

RESUMEN

Genetics and proteomics determine structure and function of normal tissues, and the molecular alterations that underlie tumorigenesis result in changes in these aspects of tissue biology in neoplasms. We review the known genetic alterations in pituitary tumors. These include the oncogenic Gsalpha protein (GSP)-activating mutations, and pituitary tumor-derived fibroblast growth factor receptor-4 (ptd-FGFR4), as well as tumor suppressor gene mutations associated with multiple endocrine neoplasia type 1 (MEN1). Other candidates identified from expression profiling include pituitary tumor-transforming gene (PTTG), GADD45, and bone morphogenic protein (BMP)4. Proteomic changes in pituitary tumors include classical alterations identified by immunohistochemistry as well as epigenetic reductions in p27. The underlying mechanisms for dysregulated cell adhesive molecules including cadherins and FGFRs are reviewed. The combined use of genetic and proteomic approaches will enhance novel drug therapeutic development.


Asunto(s)
Adenoma/genética , Adenoma/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP/genética , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Hormona Liberadora de Hormona del Crecimiento/biosíntesis , Hormona Liberadora de Hormona del Crecimiento/genética , Humanos , Proteómica , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética
20.
Fertil Steril ; 84 Suppl 2: 1257-68, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16210019

RESUMEN

OBJECTIVE: To investigate the immunocytochemical expression and presence of mRNA transcripts for basic fibroblast growth factor (bFGF) and its four receptors (FGFR-1, -2, -3, and -4) in ovaries from human adults and fetuses. DESIGN: Immunocytochemical and reverse transcription polymerase chain reaction (RT-PCR) study. SETTING: Major tertiary care and referral academic centers. PATIENT(S): Nine women and girls undergoing laparoscopic ovarian biopsy and 26 women undergoing second- and third-trimester pregnancy terminations. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Microscopic morphometric analysis, immunocytochemistry for bFGF and its receptors, and RT-PCR analyses. RESULT(S): The proteins for bFGF, FGFR-2, FGFR-3, and FGFR-4 were identified in oocytes of all follicular classes. Immunocytochemical expression of bFGF and its receptors was detected in granulosa cells of follicles from adolescents/women but not from fetuses. There was no immunocytochemical expression of FGFR-1. Transcripts of bFGF and its four receptors were identified by RT-PCR in all samples. CONCLUSION(S): The expression of bFGF and its receptors in human ovaries suggests that bFGF might have a role in early folliculogenesis.


Asunto(s)
Feto/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Folículo Ovárico/embriología , Folículo Ovárico/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adulto , Distribución de Chi-Cuadrado , Femenino , Feto/citología , Feto/fisiología , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Folículo Ovárico/citología , Embarazo , Segundo Trimestre del Embarazo/fisiología , Tercer Trimestre del Embarazo/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA