Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Acta Histochem ; 123(5): 151719, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33962151

RESUMO

Aim of our study was to provide insight into the temporal and spatial expression of FGFR1, FGFR2 and CTGF during normal human lung development which may have an important impact on understanding occurrence of developmental lung anomalies. Morphological parameters were analysed using double immunofluorescence on human embryonal (6th and 7th developmental week-dw) and foetal (8th, 9th and 16th developmental week) human lung samples. FGFR1 and FGFR2 was positive during all the dw in both the epithelium and mesenchyme. The highest number of FGFR1 positive cells was observed during the 6th dw (112/mm2) and 9th dw (87/mm2) in the epithelium compared to the 7th, 8th and 16th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001). The highest number of FGFR1 positive cells in the mesenchyme was observed during the 8th dw (19/mm2) and 16th dw (13/mm2) compared to the 6th, 7th, and 9th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001). The number of FGFR1 positive cells in the epithelium was higher for FGFR2 compared to number of positive cells (Mann-Whitney test, p < 0.0001). FGFR2 showed the highest number in the epithelium during the 7th dw (111/mm2) and 9th dw (87/mm2) compared to 6th, 8th and 16th dw (Kruskal-Wallis test, p < 0.001, p < 0.0001, p < 0.01 respectively). The highest number of FGFR2 positive cells in the mesenchyme was observed during the 9th dw (26/mm2), compared to the 6th, 7th,8th and 16th dw (Kruskal-Wallis test, p < 0.0001), while the number of FGFR2 positive cells in the epithelium was significantly higher than in the mesenchyme (Mann-Whitney test, p < 0.0001). CTGF was negative in both epithelium and mesenchyme during all except the 16th dw in the mesenchyme where it co-localized with FGFR2. FGFR1 and FGFR2 might be essential for epithelial-mesenchymal interactions that determine epithelial branching and mesenchymal growth during early lung development. Sudden increase in FGF1 in the epithelium and FGF2 in the mesenchyme in the foetus at 9th dw could be associated with the onset of foetal breathing movements. CTGF first appear during the foetal lung development.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Transição Epitelial-Mesenquimal , Epitélio , Imunofluorescência , Expressão Gênica , Humanos , Mesoderma , Microscopia de Fluorescência
2.
Inflammation ; 44(1): 160-173, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32856219

RESUMO

Acute pancreatitis (AP) is a dysfunctional pancreas disease marked by severe inflammation. Long non-coding RNAs (lncRNAs) involving in the regulation of inflammatory responses have been frequently mentioned. The purpose of this study was to ensure the function and action mode of lncRNA maternally expressed gene 3 (MEG3) in caerulein-induced AP cell model. HPDE cells were treated with caerulein to establish an AP model in vitro. The expression of MEG3, miR-195-5p, and fibroblast growth factor receptor 2 (FGFR2) was measured using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation and apoptosis were detected by 3-(4, 5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry assay, respectively. The expression of CyclinD1, B cell lymphoma/leukemia-2 (Bcl-2), Bcl-2-associated X protein (Bax), FGFR2, P65, phosphorylated P65 (p-P65), alpha inhibitor of nuclear factor kappa beta (NF-κB) (IκB-α), and phosphorylated IκB-α (p-IκB-α) at the protein level was quantified by western blot. The concentrations of tumor necrosis factor α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) were monitored by enzyme-linked immunosorbent assay (ELISA). The targeted relationship between miR-195-5p and MEG3 or FGFR2 was forecasted by the online software starBase v2.0 and verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. As a result, the expression of MEG3 and FGFR2 was decreased in caerulein-induced HPDE cells, while the expression of miR-195-5p was increased. MEG3 overexpression inhibited cell apoptosis and inflammatory responses that were induced by caerulein. Mechanically, miR-195-5p was targeted by MEG3 and abolished the effects of MEG3 overexpression. FGFR2 was a target of miR-195-5p, and MEG3 regulated the expression of FGFR2 by sponging miR-195-5p. FGFR2 overexpression abolished miR-195-5p enrichment-aggravated inflammatory injuries. Moreover, the NF-κB signaling pathway was involved in the MEG3/miR-195-5p/FGFR2 axis. Collectively, MEG3 participates in caerulein-induced inflammatory injuries by targeting the miR-195-5p/FGFR2 regulatory axis via mediating the NF-κB pathway in HPDE cells.


Assuntos
Ceruletídeo/toxicidade , MicroRNAs/biossíntese , NF-kappa B/metabolismo , Pâncreas/metabolismo , RNA Longo não Codificante/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , NF-kappa B/antagonistas & inibidores , Pâncreas/efeitos dos fármacos , Pâncreas/lesões , Pancreatite/induzido quimicamente , Pancreatite/metabolismo
3.
Eur Rev Med Pharmacol Sci ; 24(9): 4652-4664, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32432728

RESUMO

OBJECTIVE: To investigate the expression of LINC00472 in osteoporotic issues of patients, ovariectomized (OVX) mice and mice bone marrow mesenchymal stem cells (BMSCs), its effect on osteogenic differentiation of BMSCs and its mechanism. PATIENTS AND METHODS: The expression of LINC00472 and miR-300 in osteoporosis patients (n=55), ovariectomized (OVX) mice (n=10) and mice BMSCs (n=3) was detected by RT-qPCR and the correlation between the expression of miR-300 and LINC00472 was analyzed. After transferring sh-LINC00472 and overexpression LINC00472 plasmids into mice BMSCs, the expression of ALP, Bglap, OPN, Runx2 was detected by RT-qPCR and Western blot, which were related with osteogenic differentiation. In addition, Luciferase activity was used to detect whether miR-300 combined with LINC00472 and FGFR2 in mice BMSCs directly. Finally, Western blot (WB) was used to detect the change of FGFR2 protein expression by miR-300 inhibitor and sh-LINC00472. RESULTS: We found there was a negative correlation between the expression of miR-300 and LINC00472 in osteoporosis patients, bone tissues of OVX mice and mice BMSCs. The expression of LINC00472 in mice BMSCs was gradually increased with osteogenic differentiation. Transferring overexpression plasmid of LINC00472 into BMSCs, the expression of ALP, Bglap, OPN, Runx2 was increased both in mRNA and protein levels. Transferring sh-LINC00472 to BMSCs, the results were the opposite. Luciferase results showed that miR-300 could directly bind to LINC00472 and FGFR2 in mice BMSCs. What's more, RT-qPCR and WB results showed that transferring sh-LINC00472 could decrease the expression of FGFR2 mRNA and protein, while miR-300 inhibitor could recover this tendency. CONCLUSIONS: According to these results, this study revealed the previously neglected LINC00472/miR-300/FGFR2 regulatory axis for the regulation of osteogenic differentiation in osteoporosis, which may be a potential target for the treatment of osteoporosis.


Assuntos
Diferenciação Celular/fisiologia , MicroRNAs/biossíntese , Osteogênese/fisiologia , Osteoporose/metabolismo , RNA Longo não Codificante/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Animais , Feminino , Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , Osteoporose/genética , Ovariectomia , RNA Longo não Codificante/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Regulação para Cima/fisiologia
4.
PLoS One ; 15(1): e0227440, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31940413

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) recurrence after liver resection depends upon the stage and histological grade of the tumor and the expression of certain biomarkers. However, it remains unclear which of these factors has the highest predictive value regarding HCC recurrence after surgical resection. METHODS: This study investigated the associations among clinicopathological characteristics, expression of biomarkers, and HCC recurrence after liver resection. Fifty-four patients having undergone liver resection for HCC were enrolled prospectively, and their data were analyzed retrospectively. Evaluated variables were clinical data, laboratory findings, modified Union for International Cancer Control (UICC) stage, vascular invasion, histological differentiation, and immunohistochemical staining for fibroblast growth factor receptor 2 (FGFR2), vascular endothelial growth factor, and tumor-necrosis-factor-related apoptosis-inducing ligand receptors 1 and 2. RESULTS: Mean patient age was 58.6 years (range, 30-71), and the mean and SD for follow-up duration were 51.2 ± 34.8 months. Cumulative 1-, 3-, and 5-year recurrence rates were 32.9%, 53.6%, and 68.1%, respectively. In univariate analysis, FGFR2 (p = 0.026) and Edmonson-Steiner grade (E-S grade) (p = 0.030) were associated with recurrence after resection in HCC patients. In multivariate analyses, increased FGFR2 expression (p = 0.017) was the only significant predictor of HCC recurrence. CONCLUSIONS: High FGFR2 expression had marginal association with poor E-S grade (p = 0.056). More intensive surveillance of HCC recurrence is warranted in HCC patients with increased FGFR2 expression.


Assuntos
Carcinoma Hepatocelular , Regulação Neoplásica da Expressão Gênica , Hepatectomia , Neoplasias Hepáticas , Proteínas de Neoplasias/biossíntese , Recidiva Local de Neoplasia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Adulto , Idoso , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/cirurgia , Feminino , Seguimentos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/cirurgia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Estudos Retrospectivos
5.
J Pediatr Urol ; 16(1): 41.e1-41.e10, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31718875

RESUMO

INTRODUCTION: Fibroblast growth factors (FGFs) play a crucial role in early embryogenesis of the genital tubercle and are involved in the development of hypospadias, affecting both endo- and ectodermally derived tissues. It was hypothesized that expression of FGFs could be qualitatively or quantitatively altered in skin of children with hypospadias. OBJECTIVE: The objective of the study was to investigate expression patterns and transcription levels of FGF8, FGF10, and FGF Receptor 2 (FGFR2) in patients with hypospadias compared to normal controls. PATIENTS AND METHODS: Skin samples from the ventro-lateral aspect of the foreskin of 32 patients with hypospadias (17 distal and 15 proximal, mean age 25 months) and 10 normal foreskin samples (mean age 77 months) were analyzed by immunohistochemistry. Staining, localization, and distribution of positive cells in epidermis and dermis were categorized independently by two researchers. Complementary DNA (cDNA) samples prepared from messenger RNA (mRNA) isolates of the same samples were analyzed by quantitative polymerase chain reaction (qPCR), comparing expressions of FGF8, FGF10, and FGFR2 with loading controls. RESULTS: Patients with hypospadias consistently showed aberrant immunohistochemical staining patterns for FGF8/FGF10/FGFR2 in epidermis and dermis compared to patients without penile malformation (p < 0.01 for all markers). qPCR displayed no difference in expression levels on mRNA level (FGFR2 p = 0.44, FGF8 p = 0.77, and FGF10 p = 0.17) comparing normal foreskin with foreskin from patients with hypospadias. Figure. DISCUSSION: The results point at an impact of FGF signaling during embryological development of hypospadias on skin, as an ectodermally derived tissue. Similar to the urethral development, this might be a result of mesothelial-epithelial interactions. The differing expression patterns in immunohistochemistry are not matched by a quantitative difference in marker expression on the mRNA level, putatively caused by post-translational modifications or alterations of the downstream pathway. FGFs, particularly FGF10 and FGFR2, are critically involved in wound healing. CONCLUSIONS: There are significant differences in localization and distribution of FGF8, FGF10, and FGFR2 in comparisons of normal foreskin to foreskin of patients with hypospadias, whereas there is no difference in the quantitative expression of these markers on the mRNA level. This confirms the notion that penile skin is affected as well by the embryological aberrations during the embryogenesis of hypospadias.


Assuntos
Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/genética , Prepúcio do Pênis/metabolismo , Hipospadia/genética , Hipospadia/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transcrição Gênica , Criança , Pré-Escolar , Fator 10 de Crescimento de Fibroblastos/análise , Fator 8 de Crescimento de Fibroblasto/análise , Prepúcio do Pênis/química , Expressão Gênica , Humanos , Hipospadia/patologia , Imuno-Histoquímica , Lactente , Masculino , Estudos Prospectivos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise
6.
Int J Mol Med ; 44(1): 57-66, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115494

RESUMO

Cisplatin is one of the primary compounds used in the treatment of nasopharyngeal carcinoma (NPC), and fibroblast growth factor receptor 2 (FGFR2) has emerged to be a promising target for treatment in various tumors. Therefore, the present study aimed to explore whether the expression levels of FGFR2 in NPC tissues and cell lines were altered, and whether the efficiency of cisplatin was increased following the downregulation of FGFR2. The downregulation of FGFR2 was achieved by transfection with a small interfering RNA against FGFR2. Tissues of patients with NPC were analyzed by immunohistochemistry. Cell viability was examined using a Cell Counting Kit­8 assay. Cell cycle analysis was performed using flow cytometry. mRNA and protein levels were measured by reverse transcription quantitative polymerase chain reaction and western blot analysis, respectively. FGFR2 was observed to be overexpressed in cancer tissues of patients with NPC and in the NPC SUNE1, C666­1, 6­10B and HNE­3 cell lines, and resulted in an unfavorable prognosis. Cisplatin treatment decreased cell viability and increased FGFR2 expression. The silencing of FGFR2 was demonstrated to augment the effects of cisplatin treatment, including decreasing the cell viability and inducing cell cycle arrest, which involved the increase and decrease of the durations of G1 and S phases, respectively, and a decrease in the expression levels of cyclin D1 and CDC25A, and increasing the rate of apoptosis via the intrinsic apoptosis pathway, as demonstrated by the upregulation of cleaved caspase­3 and B­cell lymphoma 2 (Bcl­2)­associated X protein and downregulation of Bcl­2, in SUNE1 and C666­1 cell lines. FGFR2 was overexpressed in the cancer tissues of patients with NPC and in NPC cell lines, resulting in an unfavorable prognosis. The downregulation of FGFR2 decreased cell viability via cell cycle arrest at G1 phase, and increased the efficacy of the cisplatin­based induction of apoptosis through the intrinsic apoptosis pathway.


Assuntos
Cisplatino/farmacologia , Regulação para Baixo/efeitos dos fármacos , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/tratamento farmacológico , Proteínas de Neoplasias/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Feminino , Fase G1/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/tratamento farmacológico , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , Proteínas de Neoplasias/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Fase S/efeitos dos fármacos
7.
Acta Histochem ; 121(5): 531-538, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31047684

RESUMO

AIM: Present study analyses the co-localisation of RIP5 with FGFR1, FGFR2 and HIP2 in the developing kidney, as RIP5 is a major determinant of urinary tract development, downstream of FGF-signaling. METHODS: Paraffin embedded human kidney tissues of 16 conceptuses between the 6th-22th developmental week were analysed using double-immunofluorescence method with RIP5/FGFR1/FGFR2 and HIP2 markers. Quantification of positive cells were performed using Kruskal-Wallis test. RESULTS: In the 6th week of kidney development RIP5 (89.6%) and HIP2 (39.6%) are strongly expressed in the metanephric mesenchyme. FGFR1 shows moderate/strong expression in the developing nephrons (87.3%) and collecting ducts (70.5%) (p < 0.05). RIP5/FGFR1 co-localized at the marginal zone and the ureteric bud with predominant FGFR1 expression. FGFR2 (26.1%) shows similar expression pattern as FGFR1 (70.5%) in the same kidney structures. RIP5/FGFR2 co-localized at the marginal zone and the collecting ducts (predominant expression of FGFR2). HIP2 is strongly expressed in collecting ducts (96.7%), and co-localized with RIP5. In 10th week, RIP5 expression decrease (74.2%), while the pattern of expression of RIP5 and FGFR1 in collecting ducts (33.4% and 91.9%) and developing nephrons (21.9% and 32.4%) (p < 0.05) is similar to that in the 6th developmental week. Ureter is moderately expressing RIP5 while FGFR1 is strongly expressed in the ureteric wall. FGFR2 is strongly expressed in the collecting ducts (84.3%) and ureter. HIP2 have 81.1% positive cells in the collecting duct. RIP5/FGFR1 co-localize in collecting ducts and Henley's loop. CONCLUSIONS: The expression pattern of RIP5, FGFR1, FGFR2 and HIP2 in the human kidney development might indicate their important roles in metanephric development and ureteric muscle layer differentiation through FGF signaling pathways.


Assuntos
Rim/embriologia , Rim/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Proteína Serina-Treonina Quinases de Interação com Receptores/biossíntese , Enzimas de Conjugação de Ubiquitina/biossíntese , Imunofluorescência , Humanos
8.
J Cell Sci ; 131(21)2018 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-30257985

RESUMO

Expression of the tetraspanin CD151 is frequently upregulated in epithelial malignancies and correlates with poor prognosis. Here, we report that CD151 is involved in regulation of the expression of fibroblast growth factor receptor 2 (FGFR2). Depletion of CD151 in breast cancer cells resulted in an increased level of FGFR2. Accordingly, an inverse correlation between CD151 and FGFR2 was observed in breast cancer tissues. CD151-dependent regulation of the FGFR2 expression relies on post-transcriptional mechanisms involving HuR (also known as ELAVL1), a multifunctional RNA-binding protein, and the assembly of processing bodies (P-bodies). Depletion of CD151 correlated with inhibition of PKC, a well-established downstream target of CD151. Accordingly, the levels of dialcylglycerol species were decreased in CD151-negative cells, and inhibition of PKC resulted in the increased expression of FGFR2. Whereas expression of FGFR2 itself did not correlate with any of the clinicopathological data, we found that FGFR2-/CD151+ patients were more likely to have developed lymph node metastasis. Conversely, FGFR2-/CD151- patients demonstrated better overall survival. These results illustrate functional interdependency between CD151 complexes and FGFR2, and suggest a previously unsuspected role of CD151 in breast tumorigenesis.


Assuntos
Neoplasias da Mama/metabolismo , Proteína Quinase C/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Tetraspanina 24/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais , Tetraspanina 24/biossíntese , Tetraspanina 24/genética , Transcrição Gênica
9.
Cell Death Dis ; 9(5): 565, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29752438

RESUMO

The tumor suppressor epithelial isoform of the fibroblast growth factor receptor 2 (FGFR2b) induces human keratinocyte early differentiation. Moreover, protein kinases C (PKCs) are known to regulate the differentiation program in several cellular contexts, including keratinocytes. Therefore, in this paper we propose to clarify if FGFR2b could play a role also in the late steps of keratinocyte differentiation and to assess if this receptor-induced process would sequentially involve PKCδ and PKCα isoforms. Immunofluorescence, biochemical, and molecular approaches, performed on 2D cultures or 3D organotypic rafts of human keratinocytes overexpressing FGFR2b by stable transduction, showed that receptor signaling induced the precocious onset and an accelerated progression of keratinocyte differentiation, indicating that FGFR2b is a crucial regulator of the entire program of keratinocyte differentiation. In addition, the use of specific inhibitors and gene silencing approaches through specific siRNA demonstrated that PKCδ controls the onset of FGFR2b-triggered differentiation, while PKCα plays a role restricted to the terminal stages of the process. Molecular analysis revealed that the two PKC isoforms sequentially act via induction of KLF4 and DLX3, two transcription factors linked by negative loops to p63, suggesting that p63 would represent the hub molecule at the crossroad of an intricate signaling network downstream FGFR2b, involving multiple PKC-induced transcription factors.


Assuntos
Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Queratinócitos/enzimologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Transdução de Sinais , Linhagem Celular , Isoenzimas/genética , Isoenzimas/metabolismo , Queratinócitos/citologia , Fator 4 Semelhante a Kruppel , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
10.
Anticancer Res ; 38(5): 3105-3110, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29715147

RESUMO

BACKGROUND/AIM: The expression of IL-8 and FGFR has been related to prognosis and pathological features in renal cell carcinoma. We investigated the relationship between IL-8 and FGFR and the outcome in metastatic renal cell carcinoma (mRCC) patients. MATERIALS AND METHODS: Clinical data and histological samples of patients affected by mRCC and treated with targeted agents were reviewed. The expression of proteins was assessed using immunohistochemistry. RESULTS: FGFR1, FGFR2, and IL-8 were found to be expressed in 16%, 30%, and 50% of cases, respectively. Significant correlations were found between selected proteins. A lack of expression of FGFR2 and IL8 was found to be correlated with increased progression-free survival (PFS). The survival rate at 24 months was 44%, 38%, and 79% of those expressing both, one, or none of the evaluated proteins, respectively (p=0.047). CONCLUSION: This analysis found a relationship between the expression of IL-8 and FGFR2 in mRCC patients treated with targeted agents.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Idoso , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/mortalidade , Intervalo Livre de Doença , Feminino , Humanos , Interleucina-8/análise , Interleucina-8/biossíntese , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Estudos Retrospectivos
11.
Int J Gynecol Cancer ; 28(1): 152-160, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28953502

RESUMO

OBJECTIVES: AL3818 (anlotinib) is a receptor tyrosine kinase inhibitor targeting vascular endothelial growth factor receptors (VEGFR1, VEGFR2/KDR, and VEGFR3), stem cell factor receptor (C-kit), platelet-derived growth factor (PDGFß), and fibroblast growth factor receptors (FGFR1, FGFR2, and FGFR3). This study evaluates the efficacy of AL3818 studying tumor regression in an orthotopic murine endometrial cancer model. METHODS: We tested the cytotoxicity of AL3818 on a panel of 7 human endometrial cancer cell lines expressing either wild-type or mutant FGFR2 and also assessed the in vivo antitumor efficacy in a murine, orthotopic AN3CA endometrial cancer model. AL3818 was administered daily per os either alone or in combination with carboplatin and paclitaxel, which represent the current standard of adjuvant care for endometrial cancer. RESULTS: AL3818 significantly reduces AN3CA cell number in vitro, characterized by high expression of a mutated FGFR2 protein. Daily oral administration of AL3818 (5 mg/kg) resulted in a complete response in 55% of animals treated and in a reduced tumor volume, as well as decreased tumor weights of AN3CA tumors by 94% and 96%, respectively, following a 29-day treatment cycle. Whereas carboplatin and paclitaxel failed to alter tumor growth, the combination with AL3818 did not seem to exhibit a superior effect when compared with AL3818 treatment alone. CONCLUSIONS: AL3818 shows superior efficacy for the treatment of endometrial cancer irresponsive to conventional carboplatin and paclitaxel combination and warrants further investigation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Indóis/farmacologia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Animais , Carboplatina/administração & dosagem , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Endométrio/enzimologia , Feminino , Humanos , Indóis/administração & dosagem , Camundongos , Camundongos Nus , Paclitaxel/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Quinolinas/administração & dosagem , Distribuição Aleatória , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Tumour Biol ; 39(6): 1010428317707424, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28618942

RESUMO

In the past decades, the oncogenic role of fibroblast growth factor receptor 2 has been demonstrated in a number of cancer types. However, studies have reported contradictory findings concerning the correlation between fibroblast growth factor receptor 2 expression and prognosis in solid tumors. To address this discrepancy, we performed a meta-analysis with 18 published studies (2975 patients) retrieved from PubMed, EMBASE, and Web of science. Data were extracted and computed into odds ratios. The results showed that fibroblast growth factor receptor 2 overexpression was significantly associated with decreased 3-year overall survival (odds ratio = 1.93, 95% confidence interval: 1.30-2.85, p = 0.001) and 5-year overall survival (odds ratio = 1.62, 95% confidence interval: 1.07-2.44, p = 0.02) in patients with solid tumors. Subgroup analysis revealed that high fibroblast growth factor receptor 2 expression was also associated with poor prognosis of gastric cancer, hepatocellular carcinoma, and esophageal cancer, but not correlated with pancreatic cancer. In conclusion, fibroblast growth factor receptor 2 overexpression is correlated with decreased survival in most solid tumors, suggesting that the expression status of fibroblast growth factor receptor 2 is a valuable prognostic biomarker and a novel therapeutic target in human solid tumors.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias/genética , Prognóstico , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Biomarcadores Tumorais/genética , Intervalo Livre de Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , PubMed , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
14.
Invest Ophthalmol Vis Sci ; 58(5): 2638-2646, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28510629

RESUMO

Purpose: Little is known about the signaling mechanisms controlling meibomian gland (MG) homeostasis and the pathogenic processes leading to MG atrophy and dysfunction in dry eye disease (DED). We investigated the role of fibroblast growth factor receptor 2 (FGFR2) in the MG homeostasis of adult mice. Methods: A triple transgenic mouse strain (Krt14-rtTA; tetO-Cre; Fgfr2flox/flox), referred to as Fgfr2CKO mice, was generated in which the Fgfr2 gene is ablated by Cre recombinase in keratin 14 (Krt14)-expressing epithelial cells on doxycycline (Dox) induction. FGFR2 expression in normal human and mouse MGs was evaluated by immunohistochemistry. Pathologic MG changes in transgenic mice with conditional deletion of FGFR2 were examined by lipid staining, histology, and immunostaining. Results: FGFR2 was highly expressed in normal human MGs and adult mouse MGs. Two-month-old Fgfr2CKO mice fed Dox-containing chow for 2 weeks developed severe MG atrophy. MG acinar atrophy in the Fgfr2CKO mice was associated with reduced lipid (meibum) production and the development of clinical findings similar to those in humans with evaporative DED related to MG dysfunction (MGD). Immunohistochemical analyses showed that FGFR2 deletion severely affected proliferation and differentiation of MG acinar cells but affected MG ductal cells to a lesser extent. Conclusions: FGFR2 deletion results in significant MG acinar atrophy and clinical manifestations of MGD in Fgfr2CKO mice, suggesting that MG homeostasis is FGFR2 dependent. The Fgfr2CKO mice with inducible MG atrophy can serve as a valuable animal model for investigating the pathogenesis of MGD and developing novel therapeutic strategies for MGD-related DED.


Assuntos
Síndromes do Olho Seco/metabolismo , Glândulas Tarsais/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Adulto , Animais , Diferenciação Celular , Modelos Animais de Doenças , Síndromes do Olho Seco/diagnóstico , Pálpebras/metabolismo , Pálpebras/patologia , Feminino , Homeostase , Humanos , Imuno-Histoquímica , Glândulas Tarsais/patologia , Camundongos , Camundongos Knockout
15.
Rom J Morphol Embryol ; 58(4): 1333-1338, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29556625

RESUMO

INTRODUCTION: Chronic suppurative otitis media (CSOM) with and without cholesteatoma is regarded as chronic inflammation of the middle ear and mastoid mucosa that can be associated with the presence of granulation tissue and infection, which can lead to ossicular damage and hearing loss, but it is commonly known that cholesteatoma behaves aggressively. Both lesions appear to contain a predominant population of inflammatory cells, among which proinflammatory cytokines secreting keratinocyte growth factor (KGF) and its receptor (KGFR). No clear difference was demonstrated between these entities. The purpose of this study was to investigate the potential influence of KGF and KGFR in increased epithelial-cell proliferation of chronic otitis media (COM) with cholesteatoma in contrast to COM without cholesteatoma (CSOM), particularly in the granulative form, and to compare the rate of proliferation activity of epithelial cells using the Ki-67 epithelial proliferation marker expression. PATIENTS, MATERIALS AND METHODS: We analyzed 105 ears with cholesteatoma vs. 53 ears with CSOM without cholesteatoma using our KGF and KGFR variables, and the ratio of proliferating epithelial cells using Ki-67. The percentage of the specimens expressing KGF and KGFR was compared between the two groups for statistical significance using the Pearson's chi-square test. Immunohistochemical staining was conducted and the proportion of the cells staining positive for the nuclear antigen Ki-67 was evaluated in a quantitative and visual way, using light microscopes. RESULTS: KGF was positive in 88.57% of cholesteatoma and was positive in 41.51% CSOM without cholesteatoma specimens (cholesteatoma vs. CSOM, p=0.001). The positive rate of KGFR in the CSOM group was 33.96% compared to those in cholesteatoma, which was 60.95%. Compared to the cholesteatoma specimens, a significantly smaller number of Ki-67 labeling index was detected in CSOM specimens. CONCLUSIONS: Our results indicated that the abnormal behavior of the cholesteatoma epithelium seems to be induced by the paracrine interaction between KGF and KGFR. Furthermore, we found that cholesteatoma expressing both KGF and KGFR had high Ki-67 index, which correlated with its aggressiveness. These findings suggest that excessive KGF and KGFR synthesis may contribute to the hyperproliferative state in cholesteatoma and could explain the pathological difference between cholesteatoma and CSOM.


Assuntos
Colesteatoma/metabolismo , Fator 7 de Crescimento de Fibroblastos/biossíntese , Otite Média/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Adolescente , Adulto , Idoso , Criança , Colesteatoma/genética , Colesteatoma/patologia , Doença Crônica , Feminino , Fator 7 de Crescimento de Fibroblastos/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Antígeno Ki-67/biossíntese , Masculino , Pessoa de Meia-Idade , Otite Média/genética , Otite Média/patologia , Estudos Prospectivos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Adulto Jovem
16.
Carcinogenesis ; 37(8): 741-750, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27236187

RESUMO

The fibroblast growth factor receptor 2 (FGFR2) locus is consistently the top hit in genome-wide association studies for oestrogen receptor-positive (ER(+)) breast cancer. Yet, its mode of action continues to be controversial. Here, we employ a systems biology approach to demonstrate that signalling via FGFR2 counteracts cell activation by oestrogen. In the presence of oestrogen, the oestrogen receptor (ESR1) regulon (set of ESR1 target genes) is in an active state. However, signalling by FGFR2 is able to reverse the activity of the ESR1 regulon. This effect is seen in multiple distinct FGFR2 signalling model systems, across multiple cells lines and is dependent on the presence of FGFR2. Increased oestrogen exposure has long been associated with an increased risk of breast cancer. We therefore hypothesized that risk variants should reduce FGFR2 expression and subsequent signalling. Indeed, transient transfection experiments assaying the three independent variants of the FGFR2 risk locus (rs2981578, rs35054928 and rs45631563) in their normal chromosomal context show that these single-nucleotide polymorphisms (SNPs) map to transcriptional silencer elements and that, compared with wild type, the risk alleles augment silencer activity. The presence of risk variants results in lower FGFR2 expression and increased oestrogen responsiveness. We thus propose a molecular mechanism by which FGFR2 can confer increased breast cancer risk that is consistent with oestrogen exposure as a major driver of breast cancer risk. Our findings may have implications for the clinical use of FGFR2 inhibitors.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Predisposição Genética para Doença , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Haplótipos , Humanos , Células MCF-7 , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Transdução de Sinais , Biologia de Sistemas
17.
Oncotarget ; 7(15): 19748-61, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26933914

RESUMO

BACKGROUND: Fibroblast growth factor receptor 2 (FGFR2) genetic alterations lead to tumor cell proliferation in various types of cancer. We hypothesized that FGFR2 amplification is associated with FGFR2 expression, resulting in tumor growth and poorer outcome in esophagogastric junction (EGJ) adenocarcinoma. PATIENTS AND METHODS: A total of 176 consecutive chemo-naive patients with EGJ adenocarcinoma were enrolled from two academic institutions. FGFR2 amplification was examined by real-time PCR (N = 140) and FGFR2 expression with immunohistochemical staining (N = 176), and compared against clinicopathological factors and patient outcomes. The effects of FGFR2 inhibition or overexpression on cell proliferation, cell cycle, and apoptosis assays were investigated in EGJ adenocarcinoma cell lines. Downstream FGFR2, AKT and ERK were also examined. RESULTS: Based on the correlation between FGFR2 levels and FGFR2 overexpression in vitro, FGFR2 amplification was defined as copy number > 3.0. In clinical samples, FGFR2 amplification and FGFR2 IHC expression were 15% and 61%, respectively. Although these two statuses were significantly correlated (P < 0.05), only FGFR2 IHC expression was significantly associated with tumor depth (multivariate P < 0.001) and overall survival of patients (univariate P = 0.007). Supporting these findings, FGFR2 overexpression was associated with tumor cell proliferation, cell cycle progression, and anti-apoptosis. Selective inhibition of FGFR2 sufficiently suppressed tumor cell proliferation through de-phosphorylation of AKT and ERK. CONCLUSIONS: FGFR2 amplification was significantly associated with FGFR2 expression. FGFR2 expression (but not FGFR2 amplification) was associated with tumor growth and patient outcomes. Our findings support FGFR2 as a novel therapeutic target for EGJ adenocarcinoma.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Junção Esofagogástrica/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Neoplasias Gástricas/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Idoso , Idoso de 80 Anos ou mais , Apoptose/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Ciclo Celular/genética , Proliferação de Células/genética , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Junção Esofagogástrica/patologia , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética
18.
Oncotarget ; 7(16): 22234-44, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-26993773

RESUMO

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.


Assuntos
Biomarcadores Tumorais/análise , Neurilemoma/patologia , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neurilemoma/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/análise , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/biossíntese , Receptores de Fatores de Crescimento de Fibroblastos/análise , Adulto Jovem
19.
Tumour Biol ; 37(8): 10209-18, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26831663

RESUMO

This study was designed to determine the prognostic impact and clinical significance of FGFR2 in residual disease after preoperative chemoradiotherapy (CRT) in patients with rectal cancer. The surgical specimens of 145 patients with residual rectal cancer after preoperative CRT were analyzed. To evaluate FGFR2 expression, immunohistochemistry was performed on whole section tissues. KRAS exon 2 (codon 12 and 13), BRAF V600E mutational status, and microsatellite instability (MSI) were determined using polymerase chain reactions. Of the eligible 141 patients, FGFR2 over-expression was observed in 75.9 % (n = 107) and was correlated with perineural invasion (P = 0.005) and inferior tumor regression grading (TRG) (P = 0.009). However, FGFR2 expression had no relationship with KRAS and BRAF mutation results or with MSI results. On univariate analysis, FGFR2 over-expression was significantly associated with worse rectal cancer-specific survival (RCSS) (P = 0.005) and disease-free survival (DFS) (P = 0.035). However, multivariate analysis revealed that FGFR2 over-expression was not independently associated with RCSS and DFS (all P > 0.05). Although FGFR2 over-expression did not independently influence patient outcome, FGFR2 over-expression was associated with worse prognosis and inferior TRG. Our data may aid in understanding the therapeutic approaches targeting FGFR2 in patients with residual rectal cancer after preoperative CRT.


Assuntos
Adenocarcinoma/patologia , Biomarcadores Tumorais/análise , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Neoplasias Retais/patologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimiorradioterapia Adjuvante , Análise Mutacional de DNA , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Mutação , Terapia Neoadjuvante , Neoplasia Residual , Reação em Cadeia da Polimerase , Prognóstico , Modelos de Riscos Proporcionais , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias Retais/genética , Neoplasias Retais/metabolismo
20.
Br J Dermatol ; 174(3): 533-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26471375

RESUMO

BACKGROUND: Data indicate that in psoriasis, abnormalities are already present in nonlesional skin. Transforming growth factor-ß and keratinocyte growth factor (KGF), together with fibronectin and α5ß1 integrin, were suggested to play a crucial role in the pathogenesis of psoriasis by influencing inflammation and keratinocyte hyperproliferation. OBJECTIVES: To investigate the expression of KGF, fibroblast growth factor receptor (FGFR)2, fibronectin (FN) and extra domain A (EDA)-positive FN in healthy and nonlesional psoriatic skin, and to study the effect of KGF on the regulation of FN and EDA(+) FN production by fibroblasts. METHODS: Healthy, nonlesional psoriatic skin and lesional psoriatic skin were immunostained for α5 integrin, KGF, FGFR2, EDA(+) FN and signal transducer and activator of transcription (STAT)1. KGF-treated cell cultures were analysed for FN and EDA(+) FN mRNA and protein by real-time reverse-transcriptase polymerase chain reaction and flow cytometry, respectively. The major downstream signalling of KGF was investigated by blocking experiments using inhibitors of mitogen-activated protein kinase (MAPK) kinase (MEK1), AKT1/2, STAT1 and STAT3. RESULTS: The expression of α5 integrin, EDA(+) FN, KGF and its receptor FGFR2 is elevated in psoriatic nonlesional skin compared with healthy skin. KGF mildly induced EDA(+) FN, but not FN expression in healthy fibroblasts through MAPK signalling. Fibroblasts express the FGFR2-IIIc splice variant. STAT1 negatively regulates both FN and EDA(+) FN expression in healthy fibroblasts, and this regulation is compromised in fibroblasts derived from nonlesional psoriatic dermis. We detected active STAT1 in healthy and lesional skin, similarly to a previous report. However, in the nonlesional skin STAT1 activation was absent in tissues far away from lesions. CONCLUSIONS: The production of FN and EDA(+) FN by fibroblasts and the signalling of STAT1 are abnormally regulated in psoriatic nonlesional skin.


Assuntos
Fator 7 de Crescimento de Fibroblastos/fisiologia , Fibroblastos/metabolismo , Fibronectinas/biossíntese , Psoríase/metabolismo , Adolescente , Adulto , Estudos de Casos e Controles , Células Cultivadas , Fibronectinas/metabolismo , Voluntários Saudáveis , Humanos , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Melanócitos/metabolismo , Pessoa de Meia-Idade , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3 , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...