Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Asian Pac J Cancer Prev ; 22(12): 3949-3958, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34967576

ABSTRACT

OBJECTIVES: Paeoniflorin (PF), a compound found in Paeonia lactiflora and Paeonia suffruticosa, has anticancer potential, particularly in inhibiting migration and invasion, the resistant cancer cells hallmarks. To date, the mechanism of overcoming tamoxifen resistance in breast cancer is not yet elucidated. This research aims to explore the potential target of PF as a co-treatment for circumventing breast cancer resistance to tamoxifen with a genomic understanding-bioinformatics. METHODS: Microarray data originating from GSE67916 and GSE85871 in the NCBI GEO database was analyzed to obtain differentially expressed genes (DEGs). Further analyses were performed on DEGs using the DAVID v6.8, STRING-DB v11.0, the Cytoscape, and cBioportal. Gene expression analysis validation in breast cancer cells and tamoxifen-resistant breast cancer cells was accomplished using GEPIA and ONCOMINE databases. Survival rate analysis of selected genes was conducted using Kaplan-Meier. RESULTS: We obtained 175 DEGs from the two samples (tamoxifen-resistant and paeoniflorin-treated). DEG involves in 70 biological processes, 26 cellular components, and 18 molecular functions, and three pathways relevant to breast cancer. The PPI network analysis and hub genes selection obtained 10 genes with the highest degree scores. Genetic changes for selected genes, including IFNB1, CDK6, FGFR2, OAS1, BCL2, and STAT2 were found from 0.5% to 7% of the case population per patient case. Additional analysis using cBioportal revealed FGFR signaling pathway through Ras is important for the PF mechanism in circumventing breast cancer resistance to tamoxifen. ONCOMINE and GEPIA analysis emphasized the importance of selected genes in the tamoxifen-resistance mechanism. CONCLUSION: PF has potential to be used as a co-treatment for circumventing breast cancer resistance to tamoxifen by targeting FGFR2 signaling, but further validation is needed.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Glucosides/pharmacology , Monoterpenes/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Breast Neoplasms/drug therapy , Computational Biology , Databases, Genetic , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks , Genomics , Humans , Kaplan-Meier Estimate , Protein Interaction Maps , Signal Transduction , Tamoxifen/pharmacology
2.
Am J Pathol ; 191(4): 631-651, 2021 04.
Article in English | MEDLINE | ID: mdl-33385344

ABSTRACT

Cyclophosphamide may cause hemorrhagic cystitis and eventually bladder urothelial cancer. Genetic determinants for poor outcomes are unknown. We assessed actions of fibroblast growth factor receptor (FGFR) 2 in urothelium after cyclophosphamide exposure. Conditional urothelial deletion of Fgfr2 (Fgfr2KO) did not affect injury severity or proliferation of keratin 14+ (KRT14+) basal progenitors or other urothelial cells 1 day after cyclophosphamide exposure. Three days after cyclophosphamide exposure, Fgfr2KO urothelium had defective regeneration, fewer cells, larger basal cell bodies and nuclei, paradoxical increases in proliferation markers, and excessive replication stress versus controls. Fgfr2KO mice had evidence of pathologic basal cell endoreplication associated with absent phosphorylated ERK staining and decreased p53 expression versus controls. Mice with conditional deletion of Fgfr2 in urothelium enriched for KRT14+ cells reproduced Fgfr2KO abnormalities after cyclophosphamide exposure. Fgfr2KO urothelium had defects up to 6 months after injury versus controls, including larger basal cells and nuclei, more persistent basal and ectopic lumenal KRT14+ cells, and signs of metaplasia (attenuated E-cadherin staining). Mice missing one allele of Fgfr2 also had (less severe) regeneration defects and basal cell endoreplication 3 days after cyclophosphamide exposure versus controls. Thus, reduced FGFR2/ERK signaling apparently leads to abnormal urothelial repair after cyclophosphamide exposure from pathologic basal cell endoreplication. Patients with genetic variants in FGFR2 or its ligands may have increased risks of hemorrhagic cystitis or urothelial cancer from persistent and ectopic KRT14+ cells.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2/genetics , Regeneration/physiology , Urinary Bladder/metabolism , Urothelium/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Cystitis/chemically induced , Cystitis/metabolism , Disease Models, Animal , Mice, Transgenic , Muscle, Smooth/metabolism , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Regeneration/drug effects , Regeneration/genetics , Risk , Urinary Bladder/injuries , Urinary Bladder/pathology , Urothelium/pathology
3.
Osteoarthritis Cartilage ; 29(2): 235-247, 2021 02.
Article in English | MEDLINE | ID: mdl-33248223

ABSTRACT

OBJECTIVE: Fibronectin is a matrix protein that is fragmented during cartilage degradation in osteoarthritis (OA). Treatment of chondrocytes with fibronectin fragments (FN-f) has been used to model OA in vitro, but the system has not been fully characterized. This study sought to define the transcriptional response of chondrocytes to FN-f, and directly compare it to responses traditionally observed in OA. DESIGN: Normal human femoral chondrocytes isolated from tissue donors were treated with either FN-f or PBS (control) for 3, 6, or 18 h. RNA-seq libraries were compared between time-matched FN-f and control samples in order to identify changes in gene expression over time. Differentially expressed genes were compared to a published OA gene set and used for pathway, transcription factor motif, and kinome analysis. RESULTS: FN-f treatment resulted in 3,914 differentially expressed genes over the time course. Genes that are up- or downregulated in OA were significantly up- (P < 0.00001) or downregulated (P < 0.0004) in response to FN-f. Early response genes were involved in proinflammatory pathways, whereas many late response genes were involved in ferroptosis. The promoters of upregulated genes were enriched for NF-κB, AP-1, and IRF motifs. Highly upregulated kinases included CAMK1G, IRAK2, and the uncharacterized kinase DYRK3, while growth factor receptors TGFBR2 and FGFR2 were downregulated. CONCLUSIONS: FN-f treatment of normal human articular chondrocytes recapitulated many key aspects of the OA chondrocyte phenotype. This in vitro model is promising for future OA studies, especially considering its compatibility with genomics and genome-editing techniques.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/drug effects , Fibronectins/pharmacology , Gene Expression/drug effects , Osteoarthritis/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Chondrocytes/metabolism , Femur , Gene Expression/genetics , Humans , In Vitro Techniques , Interferon Regulatory Factors/drug effects , Interferon Regulatory Factors/genetics , Interleukin-1 Receptor-Associated Kinases/drug effects , Interleukin-1 Receptor-Associated Kinases/genetics , NF-kappa B/drug effects , NF-kappa B/genetics , Osteoarthritis/metabolism , Peptide Fragments/pharmacology , Phenotype , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Transforming Growth Factor-beta Type II/drug effects , Receptor, Transforming Growth Factor-beta Type II/genetics , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics
4.
Ann Oncol ; 28(6): 1207-1216, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28327938

ABSTRACT

Gastric cancer is the third leading cause of death from cancer worldwide. Systemic chemotherapy remains the mainstay therapeutic option for this poor prognosis cancer. Trastuzumab, the epidermal growth factor receptor 2 (ERBB2 or HER2)-antibody, is the only biological agent approved for the molecularly selected population of HER2-positive gastric cancer patients. Over the last decade, several groups have been working for deepening into the molecular characterization of gastric cancer, shedding some light into the heterogeneity of this tumour. The published data have broadened the landscape towards a future molecular classification into several subtypes of gastric cancer, enabling a better selection of the optimal therapeutic strategy. The fibroblast growth factor receptor (FGFR) pathway plays a key role in gastric cancer pathogenesis, with 1.2%-9% of gastric cancer patients harbouring FGFR2 amplifications. Several selective FGFR inhibitors have been developed in the last years, with promising efficacy signals. However, there is still scarce evidence of the most reliant molecular determinants of response to these targeted agents. Homogeneous high-level clonal FGFR2-amplification, high FGFR2 mRNA or protein levels, specific FGFR2 C3 isoform expression, FGF ligand co-overexpression or detection of FGFR2 copy number in plasma circulating tumour DNA, are considered some of the potential predictive biomarkers to the FGFR inhibition. The successful development of highly specific FGFR inhibitors will rely on our capacity of establishing new personalized strategies, based on a deeper knowledge of the key alterations that drive oncogenesis in gastric cancer. Further efforts seem mandatory in order to implement accurate predictive biomarkers in the next stages of the FGFR inhibitors development.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2/drug effects , Stomach Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Stomach Neoplasms/metabolism
5.
Am J Physiol Renal Physiol ; 312(4): F607-F618, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28052872

ABSTRACT

While urothelial signals, including sonic hedgehog (Shh), drive bladder mesenchyme differentiation, it is unclear which pathways within the mesenchyme are critical for its development. Studies have shown that fibroblast growth factor receptor 2 (Fgfr2) is necessary for kidney and ureter mesenchymal development. Our objective was to determine the role of Fgfr2 in bladder mesenchyme. We used Tbx18cre mice to delete Fgfr2 in bladder mesenchyme (Fgfr2BM-/-). We performed three-dimensional reconstructions, quantitative real-time PCR, in situ hybridization, immunolabeling, ELISAs, immunoblotting, void stain on paper, ex vivo bladder sheet assays, and in vivo decerebrated cystometry. Compared with controls, embryonic (E) day 16.5 (E16.5) Fgfr2BM-/- bladders have thin muscle layers with reduced α-smooth muscle actin levels and thickened lamina propria with increased collagen expression that intrudes into muscle. From postnatal (P) day 1 (P1) to P30, Fgfr2BM-/- bladders demonstrate progressive muscle loss and increased collagen expression. Postnatal Fgfr2BM-/- bladder sheets exhibit decreased contractility and increased passive stretch tension compared with controls. In vivo cystometry revealed high baseline and threshold pressures and shortened intercontractile intervals in Fgfr2BM-/- bladders compared with controls. Mechanistically, while Shh expression appears normal, mRNA and protein readouts of hedgehog activity are increased in E16.5 Fgfr2BM-/- bladders compared with controls. Moreover, E16.5Fgfr2BM-/- bladders exhibit higher levels of Cdo and Boc, hedgehog coreceptors that enhance sensitivity to Shh, than controls. Fgfr2 is critical for bladder mesenchyme patterning by virtue of its role in modulation of hedgehog signaling.


Subject(s)
Body Patterning , Mesoderm/metabolism , Muscle, Smooth/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Urinary Bladder/metabolism , Actins/genetics , Actins/metabolism , Animals , Apoptosis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Collagen/genetics , Collagen/metabolism , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Male , Mice, Knockout , Muscle Contraction , Muscle, Smooth/embryology , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle , Phenotype , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Urinary Bladder/embryology , Urinary Bladder/physiopathology , Urodynamics
6.
Circulation ; 131(21): 1861-71, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25794851

ABSTRACT

BACKGROUND: Fibroblast growth factor 21 (FGF21) is a metabolic hormone with pleiotropic effects on glucose and lipid metabolism and insulin sensitivity. It acts as a key downstream target of both peroxisome proliferator-activated receptor α and γ, the agonists of which have been used for lipid lowering and insulin sensitization, respectively. However, the role of FGF21 in the cardiovascular system remains elusive. METHODS AND RESULTS: The roles of FGF21 in atherosclerosis were investigated by evaluating the impact of FGF21 deficiency and replenishment with recombinant FGF21 in apolipoprotein E(-/-) mice. FGF21 deficiency causes a marked exacerbation of atherosclerotic plaque formation and premature death in apolipoprotein E(-/-) mice, which is accompanied by hypoadiponectinemia and severe hypercholesterolemia. Replenishment of FGF21 protects against atherosclerosis in apolipoprotein E(-/-)mice via 2 independent mechanisms, inducing the adipocyte production of adiponectin, which in turn acts on the blood vessels to inhibit neointima formation and macrophage inflammation, and suppressing the hepatic expression of the transcription factor sterol regulatory element-binding protein-2, thereby leading to reduced cholesterol synthesis and attenuation of hypercholesterolemia. Chronic treatment with adiponectin partially reverses atherosclerosis without obvious effects on hypercholesterolemia in FGF21-deficient apolipoprotein E(-/-) mice. By contrast, the cholesterol-lowering effects of FGF21 are abrogated by hepatic expression of sterol regulatory element-binding protein-2. CONCLUSIONS: FGF21 protects against atherosclerosis via fine tuning the multiorgan crosstalk among liver, adipose tissue, and blood vessels.


Subject(s)
Adiponectin/physiology , Atherosclerosis/prevention & control , Fibroblast Growth Factors/therapeutic use , Sterol Regulatory Element Binding Protein 2/physiology , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/biosynthesis , Adiponectin/deficiency , Adiponectin/genetics , Animals , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cholesterol/biosynthesis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fibroblast Growth Factors/deficiency , Gene Expression Regulation/drug effects , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Klotho Proteins , Liver/drug effects , Liver/metabolism , Membrane Proteins/deficiency , Membrane Proteins/drug effects , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/physiology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 2/biosynthesis , Sterol Regulatory Element Binding Protein 2/genetics
7.
Inhal Toxicol ; 26(13): 761-71, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25296878

ABSTRACT

CONTEXT: Pulmonary emphysema is resulted due to destruction of the structure of the alveoli. Recently, exogenous recombinant human Keratinocyte growth factor (rHuKGF) has been reported to induce the regeneration of gas exchange structures. However, the molecular mechanisms governing this process are so far unknown. OBJECTIVE: The objective of this study was to investigate the effect of rHuKGF in the lungs of emphysema-challenged mice on Ras-Raf-Erk (Erk, extracellular signal-regulated kinase) mediated signaling pathway that regulates alveolar epithelial cell proliferation. METHODS: Three experimental groups (i.e. emphysema, therapy and control group) were prepared. Lungs of mice were therapeutically treated at three occasions by oropharyngeal instillation of 10 mg rHuKGF per kg body weight after induction of emphysema by porcine pancreatic elastase (PPE). Subsequently, lung tissues from each mouse were collected for histopathology and molecular biology studies. RESULTS AND DISCUSSION: Histopathology photomicrographs and Destructive Index analysis have shown that elastase induced airspace enlargement and loss of alveoli were recovered in therapy group. Moreover, proliferating cell nuclear antigen (PCNA) at mRNA and protein expression level was markedly increased in therapy group than emphysema group. Upon validation at mRNA level, expressions of FGF-7, FGF-R, Ras, c-Raf, Erk-1, Erk-2, c-Myc and were significantly increased, whereas Elk-1 was notably decreased in therapy group when compared with emphysema group and were well comparable with the control group. CONCLUSION: Therapeutic supplementation of rHuKGF rectifies the deregulated Ras-Raf-Erk pathway in emphysema condition, resulting in alveolar epithelium regeneration. Hence, rHuKGF may prove to be a potential drug in the treatment of emphysema.


Subject(s)
Cell Proliferation/physiology , Fibroblast Growth Factor 7/pharmacology , MAP Kinase Signaling System/drug effects , Pulmonary Emphysema/drug therapy , Animals , Blotting, Western , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Lung/pathology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/physiology , Pulmonary Emphysema/pathology , Pulmonary Emphysema/physiopathology , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Recombinant Proteins/pharmacology
8.
Vasc Endovascular Surg ; 48(4): 289-96, 2014 May.
Article in English | MEDLINE | ID: mdl-24472831

ABSTRACT

Our purpose is to investigate the inhibitory effect and mechanisms of heparin-derived oligosaccharide (HDO) on proliferation of vascular smooth muscle cells (VSMCs) induced by basic fibroblast growth factor (bFGF). Proliferation of VSMCs was measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide; cell cycle distribution was analyzed by flow cytometry; bFGF receptor 1 and receptor 2 (bFGFR1 and bFGFR2) messenger RNA (mRNA) expression levels were determined by reverse transcription-polymerase chain reaction; and its protein expression levels were detected by Western blotting and immunocytochemical methods. Results showed that HDO inhibited VSMC proliferation in a dose-dependent manner; HDO inhibited cells in G1 phase entering the S phase; HDO inhibited bFGFR1 and bFGFR2 mRNA expression levels. In addition, bFGFR1 and bFGFR2 protein expression levels were significantly inhibited by HDO dose dependently. These results imply that HDO can inhibit VSMC proliferation. The proliferation of bFGF-induced VSMCs by HDO is associated with the inhibition of bFGFR1 and bFGFR2 expression levels. This altered molecular signature may explain one mechanism of HDO-mediated inhibition of VSMC proliferation.


Subject(s)
Heparin/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oligosaccharides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Animals , Atherosclerosis/drug therapy , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Immunohistochemistry , Rats , Reverse Transcriptase Polymerase Chain Reaction
9.
World J Gastroenterol ; 19(16): 2481-91, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23674849

ABSTRACT

AIM: To investigate the molecular mechanisms underlying the reversal effect of emodin on platinum resistance in hepatocellular carcinoma. METHODS: After the addition of 10 µmol/L emodin to HepG2/oxaliplatin (OXA) cells, the inhibition rate (IR), 50% inhibitory concentration (IC50) and reversal index (IC50 in experimental group/IC50 in control group) were calculated. For HepG2, HepG2/OXA, HepG2/OXA/T, each cell line was divided into a control group, OXA group, OXA + fibroblast growth factor 7 (FGF7) group and OXA + emodin group, and the final concentrations of FGF7, emodin and OXA in each group were 5 ng/mL, 10 µg/mL and 10 µmol/L, respectively. Single-cell gel electrophoresis was conducted to detect DNA damage, and the fibroblast growth factor receptor 2 (FGFR2), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) and excision repair cross-complementing gene 1 (ERCC1) protein expression levels in each group were examined by Western blotting. RESULTS: Compared with the IC50 of 120.78 µmol/L in HepG2/OXA cells, the IC50 decreased to 39.65 µmol/L after treatment with 10 µmol/L emodin; thus, the reversal index was 3.05. Compared with the control group, the tail length and Olive tail length in the OXA group, OXA + FGF7 group and OXA + emodin group were significantly increased, and the differences were statistically significant (P < 0.01). The tail length and Olive tail length were lower in the OXA + FGF7 group than in the OXA group, and this difference was also statistically significant. Compared with the OXA + FGF7 group, the tail extent, the Olive tail moment and the percentage of tail DNA were significantly increased in the OXA + emodin group, and these differences were statistically significant (P < 0.01). In comparison with its parental cell line HepG2, the HepG2/OXA cells demonstrated significantly increased FGFR2, p-ERK1/2 and ERCC1 expression levels, whereas the expression of all three molecules was significantly inhibited in HepG2/OXA/T cells, in which FGFR2 was silenced by FGFR2 shRNA. In the examined HepG2 cells, the FGFR2, p-ERK1/2 and ERCC1 expression levels demonstrated increasing trends in the OXA group and OXA + FGF7 group. Compared with the OXA group and OXA + FGF7 group, the FGFR2, p-ERK1/2, and ERCC1 expression levels were significantly lower in the OXA + emodin group, and these differences were statistically significant. In the HepG2/OXA/T cell line that was transfected with FGFR2 shRNA, the FGFR2, p-ERK1/2 and ERCC1 expression levels were significantly inhibited, but there were no significant differences in these expression levels among the OXA, OXA + FGF7 and OXA + emodin groups. CONCLUSION: Emodin markedly reversed OXA resistance by enhancing OXA DNA damage in HepG2/OXA cells, and the molecular mechanism was related to the inhibitory effect on ERCC1 expression being mediated by the FGFR2/ERK1/2 signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Emodin/pharmacology , Endonucleases/metabolism , Liver Neoplasms/enzymology , Organoplatinum Compounds/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Signal Transduction/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , DNA Damage , Dose-Response Relationship, Drug , Fibroblast Growth Factor 7/metabolism , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxaliplatin , Phosphorylation , RNA Interference , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Transfection
10.
J Periodontal Res ; 48(6): 740-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23528007

ABSTRACT

BACKGROUND AND OBJECTIVE: Keratinocyte growth factor (KGF) and its receptor (KGFR) are involved in hyperplastic diseases. This study explored the effect of intercellular communication on KGF and KGFR in cocultured/monocultured gingival fibroblasts and keratinocytes following treatment with nifedipine. MATERIAL AND METHODS: Human gingival fibroblasts and keratinocytes were monocultured and cocultured, respectively. MTT was used to investigate the effects of nifedipine on the proliferation of gingival fibroblasts and keratinocytes. Monoculture and coculture systems were treated with different concentrations (0, 0.2 or 20 µg/mL) of nifedipine, and the expression of KGF and KGFR mRNAs was examined by RT-PCR, whilst the secretion of KGF and the expression of KGFR on the membrane were analyzed using ELISA and flow cytometry, respectively. RESULTS: Nifedipine (0, 0.2 and 20 µg/mL) had no influence on cell proliferation within 3 d. KGF and KGFR mRNAs were up-regulated, but only in the cocultures. In coculture, the secretion of KGF was significantly increased by nifedipine, while it was only significantly up-regulated by 20 µg/mL of nifedipine in monoculture. Moreover, the level of KGFR protein in the membrane was significantly increased by 20 µg/mL of nifedipine in monocultures, while it was significantly down-regulated by 20 µg/mL of nifedipine in cocultures. CONCLUSION: The expression of KGF and KGFR are influenced by the interplay of gingival keratinocytes and fibroblasts. Epithelial keratinocytes and mesenchymal fibroblasts may interplay to dynamically regulate gene expression, which may have an effect on the gingival condition following treatment with nifedipine.


Subject(s)
Calcium Channel Blockers/pharmacology , Fibroblast Growth Factor 7/drug effects , Fibroblasts/drug effects , Gingiva/drug effects , Keratinocytes/drug effects , Nifedipine/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Cell Communication/drug effects , Cell Culture Techniques , Cell Membrane/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Coloring Agents , Gene Expression Regulation/drug effects , Gingiva/cytology , Humans , Tetrazolium Salts , Thiazoles
11.
Article in English | MEDLINE | ID: mdl-23313229

ABSTRACT

OBJECTIVE: The objective of this study was to find keratinocyte growth factor (KGF) mimic peptides by a phage display library screening and to analyze their effects on proliferation of human oral mucosal epithelial cells (HOMECs). STUDY DESIGN: A phage display library was screened by anti-KGF antibody. ELISA was performed to select monoclonal phages with higher binding activity. The promotion of the phage model peptides on HOMEC proliferation were analyzed by MTT and their cell affinities were confirmed by immunofluorescence assay. Their effect on KGFR, human beta-defensin 3, c-Fos, and c-Jun in HOMEC were analyzed by quantitative real-time PCR. RESULTS: Two model peptides with higher affinity with HOMEC were found to have promotive activity on cell proliferation, similar to that of KGF. These 2 model peptides have no KGF-like promotion effect on the expression of c-Fos and c-Jun. CONCLUSIONS: The 2 phage model peptides can promote the proliferation of HOMEC in vitro without tumorigenic effects, which suggests their possible usages in oral mucosal wound healing.


Subject(s)
Fibroblast Growth Factor 7/pharmacology , Mouth Mucosa/drug effects , Bacteriophages/isolation & purification , Cell Culture Techniques , Cell Line , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Humans , Keratinocytes/drug effects , Molecular Mimicry , Mouth Mucosa/cytology , Peptide Library , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-jun/drug effects , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Sequence Analysis, DNA , beta-Defensins/drug effects
12.
Eur Arch Otorhinolaryngol ; 265(10): 1173-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18246360

ABSTRACT

To investigate the cell-biological effect of topically applied 5-fluorouracil (5-FU) on middle ear cholesteatoma, 12 attic cholesteatomas were treated with topical application of 5-FU cream, two to five times with an interval of 2 weeks (5-FU group). The control group comprised 65 cholesteatoma that were not treated with 5-FU. All lesions were later excised surgically and processed for immunohistochemical analyses of Ki-67, keratinocyte growth factor (KGF) and its receptor (KGFR). 5-FU significantly reduced the expression of KGF, did not change KGFR expression, and significantly reduced the Ki-67 labeling index, relative to the control group. The effect of 5-FU on cholesteatoma seems to be mediated, at least in part, through downregulation of KGF in stromal cells and reduction of the proliferative activity of epithelial cells.


Subject(s)
Cholesteatoma, Middle Ear/drug therapy , Down-Regulation/drug effects , Fibroblast Growth Factors/biosynthesis , Fluorouracil/administration & dosage , Immunosuppressive Agents/administration & dosage , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Cell Proliferation/drug effects , Child , Cholesteatoma, Middle Ear/metabolism , Cholesteatoma, Middle Ear/pathology , Female , Fibroblast Growth Factors/drug effects , Follow-Up Studies , Humans , Immunohistochemistry , Ki-67 Antigen/drug effects , Ki-67 Antigen/metabolism , Male , Middle Aged , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Retrospective Studies , Treatment Outcome , Young Adult
13.
J Cell Physiol ; 210(3): 720-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17133345

ABSTRACT

Gene amplification and protein overexpression of fibroblast growth factor receptor 2 (FGFR2) characterize the SUM-52 breast cancer cell line developed in our laboratory. SUM-52 cells express nine distinct alternatively spliced isoforms of FGFR2. Among these isoforms are two otherwise identical FGFR2 variants that express either the C1 or C3 carboxyl terminus. FGFR2-C3 variants are not normally expressed by human mammary epithelial (HME) cells, and we have shown that overexpression of FGFR2-C3 in HME cells results in potent transformation. In particular, FGFR2-C3 expression leads to robust levels of constitutively tyrosine phosphorylated FRS2 in the absence of ligand stimulation. In contrast, overexpressed FGFR2-C1 requires constant stimulation with exogenous keratinocyte growth factor (KGF) to mimic the signaling capability of FGFR2-C3. However, activation of FRS2 that results from KGF-stimulated FGFR2-C1 signaling is transient and is associated with a mobility shift of FRS2 not observed when this signaling molecule is activated by the C3 isoform of FGFR2. Mutation of the only tyrosine phosphorylated site present in the C1 terminus and absent from C3, Tyr769, did not yield a receptor that rivaled the potent signaling of FGFR2-C3. We therefore conclude that aberrant expression of alternatively spliced isoforms of FGFR2 with the C3 carboxyl terminus in the SUM-52 breast cancer cells results in sustained activation of signal transduction leading to transformation.


Subject(s)
Alternative Splicing/genetics , Epithelial Cells/metabolism , Mammary Glands, Human/cytology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , Epithelial Cells/drug effects , Female , Fibroblast Growth Factor 7/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genetic Variation/genetics , Humans , Mammary Glands, Human/metabolism , Protein Isoforms , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Threonine/metabolism
14.
J Neurosci Res ; 84(3): 483-96, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16770775

ABSTRACT

Prenatal ethanol exposure induces functional abnormalities during brain development affecting neurogenesis and gliogenesis. We have previously reported that alcohol exposure during embryogenesis disrupts radial glia (RG) and gliogenesis. Taking into account the new role of RG as neural progenitors, we have investigated whether ethanol affects RG as a neural stem cell. We found that in utero ethanol exposure impairs cell proliferation and decreases neurons and astrocytes generated in cultured RG and in embryonic cerebral cortex. Telencephalic cultures obtained at E12 from ethanol-treated rats displayed a reduction in the proportion of actively dividing RG progenitors, as demonstrated by 5-bromo-2'-deoxyuridine incorporation, and in the percentage of brain lipid binding protein-positive RG. Consistently, neurosphere formation assay from E12 telencephalon showed a reduced number of multipotent progenitor cells in cultures isolated from ethanol-treated rats in comparison with pair-fed control group. Moreover, levels of activated Notch1 and fibroblast growth factor receptor 2, which regulate the maintenance of the progenitor state of RG, are decreased by prenatal ethanol exposure. These findings demonstrate that ethanol reduces the telencephalic RG progenitor pool and its transformation into neurons and astrocytes, which may contribute to an explanation of the defects in brain function often observed in fetal alcohol syndrome.


Subject(s)
Alcohol-Induced Disorders, Nervous System/physiopathology , Brain/abnormalities , Brain/drug effects , Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Brain/physiopathology , Bromodeoxyuridine , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Central Nervous System Depressants/adverse effects , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Female , Neurons/drug effects , Neurons/physiology , Pregnancy , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Notch1/drug effects , Receptor, Notch1/metabolism , Spheroids, Cellular , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , Telencephalon/abnormalities , Telencephalon/drug effects , Telencephalon/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...