epidermal-growth-factor has been researched along with thiazolyl-blue* in 23 studies
23 other study(ies) available for epidermal-growth-factor and thiazolyl-blue
Article | Year |
---|---|
Epidermal growth factor can optimize a serum-free culture system for bone marrow stem cell proliferation in a miniature pig model.
Bone marrow-derived mesenchymal stem cells have become an attractive cell source for periodontal ligament regeneration treatment because of their potential to engraft to several tissue types after injury. Most researchers have focused on the transplantation process, but few have paid attention to cell safety concerns and rapid proliferation before transplantation. Using serum-free medium to culture stem cells may be an effective method to avoid problems associated with exogenous serum and the addition of growth factors to promote cell proliferation. Here, we randomly divided our serum-free cultures and treated them with different levels of epidermal growth factor (EGF). We then evaluated changes in rates of cell adhesion, proliferation, apoptosis, and cell cycle ratio as well as their differentiation potential. The data showed that all of these parameters were significantly different when comparing serum-free cultures with and without 10 nM/L EGF (p < 0.05/0.01); however, cells with 10 nM/L EGF did not respond differently than cells grown in standard serum-containing media without EGF (p > 0.05). In summary, our results demonstrate that 10 nM/L EGF was the optimal dose for serum-free culture, which can replace traditional standard serum medium for in vitro expansion of miniature pig bone marrow-derived mesenchymal stem cells. Topics: Animals; Apoptosis; Cell Adhesion; Cell Culture Techniques; Cell Cycle; Cell Proliferation; Culture Media, Serum-Free; DNA Primers; Dose-Response Relationship, Drug; Epidermal Growth Factor; Mesenchymal Stem Cells; Real-Time Polymerase Chain Reaction; Swine; Swine, Miniature; Tetrazolium Salts; Thiazoles | 2013 |
Random/aligned electrospun PCL/PCL-collagen nanofibrous membranes: comparison of neural differentiation of rat AdMSCs and BMSCs.
In this study, the aligned (A) and randomly oriented (R) polycaprolactone (PCL-A and PCL-R) and PCL/collagen (PCL/Col-A and PCL/Col-R) nanofibers were electrospun onto smooth PCL membranes (PCLMs) prepared by solvent casting. In order to investigate the effects of chemical composition and nanotopography of fibrous surfaces on proliferation and on neural differentiation of mesenchymal stem cells (MSCs), adipose and bone marrow-derived rat MSCs (AdMSCs and BMSCs) were cultivated in suitable media i.e. inducing medium containing basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF), and cell maintenance medium (CMM). BMSCs adhered and proliferated on all nanofibrous membranes more efficiently than AdMSCs. PCL/Col-A was found as the most convenient surface supporting proliferation in both cell types. Immunofluorescence staining indicated that BMSCs and AdMSCs are prone for differentiation to oligodendrocytes more than they differentiate to other neuronal cell types. PCL-A nanofibrous membranes supported differentiation of MSCs to O4(+) (an oligodendrocytes surface antigen) cells in both culture media. The intensity of immunoreactivity of O4(+) cells differentiated from BMSCs on PCL-A was highest when compared with the other groups (p < 0.001). Some BIII-T signed neural cells were investigated on PCL-A nanofibrous membranes, but the intensity of immunoreactivity was lower than that of O4(+) cells. In conclusion, this study can be evaluated to establish the cell therapy strategies in neurodegenerative disorders, which are relevant to oligodendrocyte abstinence using BMSCs or AdMSCs on aligned nanofibrous membranes. Topics: Animals; Bone Marrow Cells; Cell Adhesion; Cell Differentiation; Cell Proliferation; Cell Survival; Collagen; Culture Media; Epidermal Growth Factor; Fibroblast Growth Factor 2; Flow Cytometry; Mesenchymal Stem Cells; Microscopy, Electron, Scanning; Microscopy, Fluorescence; Nanofibers; Neurons; Polyesters; Rats; Tetrazolium Salts; Thiazoles; Tissue Engineering; Tissue Scaffolds | 2012 |
Toxicity of Flow Line, Durafill VS, and Dycal to dental pulp cells: effects of growth factors.
The objective was to determine the effects of growth factor treatment on dental pulp cell sensitivity to toxicity of 2 composite restoration materials, Flow Line and Durafill VS, and a calcium hydroxide pulp capping material, Dycal.. Toxicity of the dental materials to cultures of primary dental pulp cells was determined by the MTT metabolism assay. The ability of 6 different growth factors to influence the toxicity was tested.. A 24-hour exposure to either Flow Line or Durafill VS caused approximately 40% cell death, whereas Dycal exposure caused approximately 80% cell death. The toxicity of Flow Line and Durafill VS was mediated by oxidative stress. Four of the growth factors tested (bone morphogenetic protein [BMP]-2, BMP-7, epidermal growth factor [EGF], and transforming growth factor [TGF]-beta) decreased the basal MTT values while making the cells resistant to Flow Line and Durafill VS toxicity except BMP-2, which made the cells more sensitive to Flow Line. Treatment with fibroblast growth factor-2 caused no change in basal MTT metabolism, prevented the toxicity of Durafill VS, but increased the toxicity of Flow Line. Treatment with insulin-like growth factor-I (IGF-I) increased basal MTT metabolism and made the cells resistant to Flow Line and Durafill VS toxicity. None of the growth factors made the cells resistant to Dycal toxicity.. The results indicated that growth factors can be used to alter the sensitivity of dental pulp cells to commonly used restoration materials. The growth factors BMP-7, EGF, TGF-beta, and IGF-I provided the best profile of effects, making the cells resistant to both Flow Line and Durafill VS toxicity. Topics: Adult; Amino Acid Chloromethyl Ketones; Antioxidants; Bone Morphogenetic Protein 2; Bone Morphogenetic Protein 7; Calcium Hydroxide; Caspase Inhibitors; Cell Death; Cell Survival; Cells, Cultured; Chromans; Coloring Agents; Composite Resins; Dental Materials; Dental Pulp; Drug Tolerance; Epidermal Growth Factor; Fibroblast Growth Factor 2; Humans; Insulin-Like Growth Factor I; Intercellular Signaling Peptides and Proteins; Materials Testing; Minerals; Oxidative Stress; Tetrazolium Salts; Thiazoles; Time Factors; Transforming Growth Factor beta | 2010 |
Inhibitory effects of fucoidan on activation of epidermal growth factor receptor and cell transformation in JB6 Cl41 cells.
Algal fucoidan is a marine sulfated polysaccharide with a wide variety of biological activities including anti-thrombotic, anti-inflammatory, and anti-tumor activities. In this study, we tested the hypothesis that fucoidan may suppress neoplastic cell transformation by inhibiting the phosphorylation of epidermal growth factor receptor (EGFR) in mouse epidermal JB6 Cl41 cells. Our results provided the first evidence that fucoidan from Laminaria guryanovae exerted a potent inhibitory effect on EGF-induced phosphorylation of EGFR. Consistent with its inhibitory action on phosphorylation of EGFR, fucoidan clearly suppressed the phosphorylation of extracellular signal-regulated kinase or c-jun N-terminal kinases induced by EGF. Moreover, EGF-induced the c-fos and c-jun transcriptional activities were inhibited by fucoidan, resulting to suppressing of activator protein-1 (AP-1) activity and cell transformation induced by EGF. Taken together, these results indicate that fucoidan might exert chemopreventive effects through the inhibition of phosphorylation of the EGFR. Topics: Animals; Antineoplastic Agents; Biotransformation; Blotting, Western; Cell Line, Tumor; Cell Survival; Cell Transformation, Neoplastic; Chromatography, Ion Exchange; Epidermal Growth Factor; ErbB Receptors; Genes, fos; Genes, jun; Genes, Reporter; Indicators and Reagents; Laminaria; Mice; Phosphorylation; Polysaccharides; Signal Transduction; Tetrazolium Salts; Thiazoles; Transcription Factor AP-1 | 2008 |
Evaluation of RGD- or EGF-immobilized chitosan scaffolds for chondrogenic activity.
Chitosan scaffolds were prepared by freeze-drying method and modified with Arg-Gly-Asp (RGD) sequence of fibronectin or epidermal growth factor (EGF) by covalent immobilization. The results obtained from FTIR-ATR, fluorescence visualization and quantitative measurements showed that biosignal molecules, RGD and EGF, were successfully immobilized on chitosan scaffolds. ATDC5 murine chondrogenic cells were seeded on both type of scaffolds, chitosan-RGD and chitosan-EGF, and cultured for 28 days in stationary conditions. According to the results of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazoliumbromide (MTT) test, considerable increase in cell proliferation was only detected on chitosan-EGF scaffolds. Biochemical analysis of the chondrocyte seeded scaffolds showed that glycosaminoglycan (GAG) and deoxyribonucleic acid (DNA) content of the scaffolds increases with time. In conclusion, EGF-modified chitosan scaffolds (containing 1.83 microg EGF/3 mg dry scaffold) have been proposed to promote chondrogenesis and to have potential for reticular cartilage regeneration. Topics: Analysis of Variance; Chitosan; Epidermal Growth Factor; Microscopy, Electron, Scanning; Microscopy, Fluorescence; Oligopeptides; Spectroscopy, Fourier Transform Infrared; Tetrazolium Salts; Thiazoles; Tissue Engineering; Tissue Scaffolds | 2008 |
Mitogenic and apoptotic actions of epidermal growth factor on neuroblastoma cells are concentration-dependent.
In previous studies we have shown that epidermal growth factor (EGF) at concentrations between 50 and 100 ng/mL induced apoptosis in wild-type SK-N-SH neuroblastoma cells. We hypothesize that this apoptotic event separates EGF-induced neuroblastoma cell growth into a biphasic concentration-dependent process, due to activation of different signaling cascades.. Cells were incubated in concentrations of EGF ranging from 5 to 250 ng/mL for 3 days, and cell proliferation was determined by the MTT assay. Cells incubated with EGF 5, 100, or 250 ng/mL for 17 h were also assayed for apoptosis by DNA laddering. Western immunoblots were performed on whole cell lysates prepared from cells incubated with EGF (5-250 ng/mL) for 17 h. Antibodies against cleaved caspase3, p-AKT, p-GSK-3beta, p-BAD, p-RAF, p-ERK, and p-P38 were used as probes.. A triphasic, concentration-dependent response was observed following incubation of cells with EGF. Cell proliferation was increased by EGF 5 ng/mL (P < 0.05), decreased by EGF 100 ng/mL, and increased when incubated with EGF 250 ng/mL (P < 0.05). DNA laddering only occurred after treatment with EGF 100 ng/mL. The expressions of p-ERK, p-RAF, p-BAD, and p-GSK-3beta were increased at EGF concentrations of 5-10 ng/mL. At 50-100 ng/mL EGF, the expression of cleaved caspase3 was increased. Maximal p-P38 expression was at 50 ng/mL EGF. At EGF concentrations of 150-250 ng/mL, the expressions of p-AKT and p-GSK-3beta were elevated.. Neuroblastoma cell growth induced by EGF exhibited a triphasic pattern; cell growth was increased at EGF concentrations 5-20 and 150-250 ng/mL, but decreased at 50-100 mg/mL. Apoptosis was induced at 50-100 ng/mL EGF. Each growth phase activated different signaling molecules. Topics: Apoptosis; Blotting, Western; Cell Line, Tumor; Cell Proliferation; Dose-Response Relationship, Drug; Epidermal Growth Factor; Humans; Signal Transduction; Tetrazolium Salts; Thiazoles | 2006 |
Pulmonary fibroblasts stimulate the proliferation of cell lines from human lung adenocarcinomas.
Human lung cancer cell lines are widely used to test anticancer drugs. These in-vitro tests, however, preclude the detection of responses to paracrine factors from surrounding stroma. We have cocultured pulmonary fibroblasts CCD-19Lu, from a healthy donor, or HLF-A, from a patient with epidermoid carcinoma of the lung, with two human pulmonary adenocarcinoma cell lines to test the hypothesis that the fibroblasts stimulate the growth of the tumor cells. Both fibroblast cell lines significantly increased the proliferation of the pulmonary adenocarcinoma cell lines in 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide assays, with HLF-A fibroblasts yielding the most pronounced responses. The proliferation of the pulmonary adenocarcinoma cell lines in coculture with fibroblasts was blocked by antibodies against the transforming growth factor-alpha and amphiregulin. In addition, reverse transcription-polymerase chain reaction showed expression of mRNA for amphiregulin and transforming growth factor-alpha in all cell lines, whereas mRNA for the epidermal growth factor was detected only in pulmonary adenocarcinoma cell lines. Western blot analysis revealed that medium containing growth factors released by each fibroblast cell line activated extracellular signal-regulated kinase 1/2 in the both tested pulmonary adenocarcinoma cell lines, but activated Akt kinase only in A549 cells. Assessment of protein levels for cyclin D1 and cyclin E by Western blots demonstrated pronounced increases of both proteins in each pulmonary adenocarcinoma cell line, whereas protein levels for cyclin-dependent kinase inhibitor p21 remained unchanged. Immunocytochemical analysis showed positive immunoreactivity for P-extracellular signal-regulated kinase 1/2, cyclin D1 and cyclin E in pulmonary adenocarcinoma cells cocultured with fibroblasts or exposed to fibroblast-conditioned media. Our data suggest that the growth of pulmonary adenocarcinoma is stimulated by amphiregulin and transforming growth factor-alpha released from pulmonary fibroblasts. This may contribute to the disappointing clinical responses to anticancer drugs, which have shown promise in tests with lung cancer cell lines. Topics: Adenocarcinoma; Amphiregulin; Animals; Antibodies, Blocking; Cell Line, Tumor; Cell Proliferation; Coculture Techniques; Culture Media, Conditioned; Cyclin D1; Cyclin E; EGF Family of Proteins; Enzyme Activation; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Fibroblasts; Glycoproteins; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Lung; Lung Neoplasms; Mice; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Tetrazolium Salts; Thiazoles; Transforming Growth Factor alpha | 2006 |
Epidermal growth factor induction of resistance to topoisomerase II toxins in human squamous carcinoma A431 cells.
Alteration of the epidermal growth factor (EGF) signaling pathway occurs frequently in human cancer cells and may subsequently affect the cell survival towards anti-cancer agents. To elucidate the effect of long-term EGF treatment on the chemo-sensitivity of human cancer cells, human squamous carcinoma A431 cells (AP) were incubated continuously with 50 ng/ml EGF for 30 weeks and these cells were designated as the AC cells. The long-term EGF treatment did not alter the EGFR level and the EGF-induced protein tyrosine phosphorylation pattern in the AC cells. By MTT assay, the AC cells were shown to be more resistant than the AP cells to doxorubicin, etoposide and amsacrine but not to cisplatin. Among the drug-resistant proteins, topoisomerase IIalpha (topoII) was downregulated in the AC cells while there was no apparent change in the levels of P-glycoprotein, MRP-1 or glutathione- S-transferase-pi as compared to the AP cells. Furthermore, knockdown of topoII by antisense topoII oligonucleotide transfection decreased the sensitivity to doxorubicin, etoposide and amsacrine in the A431 cells. Results from the present study support an idea that long-term treatment with EGF may induce drug resistance in cells through the downregulation of topoII. Topics: Carcinoma, Squamous Cell; Cell Line, Tumor; Cell Survival; DNA Topoisomerases, Type II; Down-Regulation; Drug Resistance, Neoplasm; Epidermal Growth Factor; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Humans; Oligonucleotides; Phosphorylation; Tetrazolium Salts; Thiazoles; Tyrosine | 2006 |
Epidermal growth factor-induced proliferation requires down-regulation of Pax6 in corneal epithelial cells.
Growth factors play important roles in regulating corneal epithelial cell proliferation/differentiation during wound healing. It is suggested that PAX6 involves corneal epithelium lineage-specific differentiation (Liu, J. J., Kao, W. W., and Wilson, S. E. (1999) Exp. Eye Res. 68, 295-301); however, the regulatory mechanism and function of Pax6 in growth factor-induced corneal epithelial responses is still unknown. In the present study, we found that the mitogenic effect of epidermal growth factor (EGF) in corneal epithelial cells required suppression of PAX6 activity through cellular mechanisms involving Erk-signaling pathway-mediated increase in CTCF expression. EGF-induced CCCTC binding factor (CTCF) activation subsequently inhibited Pax6 expression by interacting with a CTCF-specific region upstream of the pax6 P0 promoter. Suppression of EGF-induced Erk activation by specific inhibitor or by the dominant expression of a silent Erk mutant effectively abolished the effects of EGF stimulation on regulations of CTCF and pax6. Apparently, down-regulation of Pax6 expression induced by EGF is required for corneal epithelial proliferation, because overexpression of pax6 in these cells attenuated EGF-induced proliferation. In contrast, knockdown of mRNA expression with pax6- or CTCF-specific small interfering RNA in corneal epithelial cells significantly promoted or attenuated EGF-induced proliferation, respectively. Thus, our results revealed a new regulatory mechanism that involves cellular signaling events and pax6 transcription regulation in growth factor-mediated proliferation. In corneal epithelial cells, this suggests that inhibition of pax6 expression is a prerequisite for EGF to elicit controls of cell growth and fate. Topics: Animals; beta-Galactosidase; Blotting, Northern; Blotting, Western; Cell Lineage; Cell Proliferation; Coloring Agents; Cornea; Down-Regulation; Epidermal Growth Factor; Epithelial Cells; Eye Proteins; Homeodomain Proteins; Humans; Luciferases; MAP Kinase Signaling System; Paired Box Transcription Factors; PAX6 Transcription Factor; Promoter Regions, Genetic; Protein Binding; Rabbits; Repressor Proteins; RNA, Messenger; RNA, Small Interfering; Signal Transduction; Tetrazolium Salts; Thiazoles; Time Factors; Transfection | 2005 |
Suramin promotes proliferation and scattering of renal epithelial cells.
Primary cultures of renal proximal tubules are known to recapitulate several early events in the process of renal regeneration following injury. In this study, we show that suramin, a polysulfonated naphthylurea, stimulates outgrowth, scattering, and proliferation of primary cultures of renal proximal tubule cells (RPTC). These responses were comparable to those produced by epidermal growth factor (EGF). However, AG-1478 [4-(3'-chloroanilino)-6,7-dimethoxy-quinazoline], a specific inhibitor of the EGF receptor, blocked EGF but not suramin-induced RPTC outgrowth, scattering, and proliferation. Suramin stimulated phosphorylation of Akt, a downstream kinase of phosphoinositide 3-kinase (PI3K), extracellular signaling-regulated kinase 1/2 (ERK1/2), and Src, but not the EGF receptor. Blockade of Src, but not the EGF receptor, inhibited Akt and ERK1/2 phosphorylation. Furthermore, inactivation of PI3K with LY294002 [2-(4morpholinyl)-8-phenyl-4H-1-benzopyran-4-one] blocked suramin-induced RPTC outgrowth, scattering, and proliferation, whereas blockade of ERK1/2 had no effect. These data identify novel effects of suramin in RPTC outgrowth, scattering, and proliferation. Furthermore, suramin-induced outgrowth, scattering, and proliferation of RPTC are through Src-mediated activation of the PI3K pathway but not ERK1/2 or the EGF receptor. Topics: Animals; Blotting, Western; Cell Cycle; Cell Movement; Cell Proliferation; Cells, Cultured; Epidermal Growth Factor; Epithelial Cells; Female; Genes, src; Humans; Immunohistochemistry; Indicators and Reagents; Kidney; Kidney Tubules, Proximal; Phosphatidylinositol 3-Kinases; Rabbits; Signal Transduction; Suramin; Tetrazolium Salts; Thiazoles; Trypanocidal Agents | 2005 |
Epidermal morphogenesis in an in-vitro model using a fibroblasts-embedded collagen scaffold.
A novel culture system included a self-designed bi-layer 3-D collagen scaffold with different pore size on both sides and specific culture media for different culture stages. This skin equivalent culture model provides a new investigating system to study the role of extracellular matrix and growth factors including epidermal growth factor (EGF), keratinocyte growth factor (KGF), transforming growth factor beta 1 (TGF-beta1), in the cell-cell and cell-matrix interactions. Keratinocytes were seeded onto the dermal equivalent and incubated under submerged condition for 5 days then proceeding to air-liquid interface cultured either with or without EGF addition. In this study, EGF has a positive effect on the keratinocyte migration and proliferation in the submerged stage. However, when 10 ng per ml of EGF was continual added in the air-lifted stage, a less organized and thin differentiated keratinocyte layers were found. Continual 10 ng per ml of EGF addition in the air-lifted stage resulted in uneven cell-matrix interface, and disorganization of the suprabasal layers. On the contrary, in the air-lifted stage without excess EGF, the epithelium cells will stratify, differentiate, and form an epidermis completed with basal, spinous, granular, and cornified layers. The results showed that time scale modulation of EGF on keratinocyte cell behavior depend on the expression of paracrine or autocrine growth factors (e.g. KGF and TGF-beta1). Topics: Animals; Cell Communication; Cell Differentiation; Cell Movement; Cell Proliferation; Cells, Cultured; Collagen; Culture Media; Dermis; DNA Primers; Electrophoresis, Polyacrylamide Gel; Epidermal Growth Factor; Epidermis; Extracellular Matrix; Fibroblast Growth Factor 7; Fibroblasts; Immunohistochemistry; Keratinocytes; Microscopy, Electron, Scanning; Models, Biological; Organ Culture Techniques; Phenotype; Polymers; Reverse Transcriptase Polymerase Chain Reaction; Skin; Swine; Tetrazolium Salts; Thiazoles; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
Identification and characterization of a novel peptide ligand of epidermal growth factor receptor for targeted delivery of therapeutics.
Epidermal growth factor receptor (ErbB1, EGFR) is overexpressed in a variety of human cancer cells. It has been considered as a rational target for drug delivery. To identify novel ligands with specific binding capabilities to EGFR, we screened a phage display peptide library and found an enriched phage clone encoding the amino acid sequence YHWYGYTPQNVI (designated as GE11). Competitive binding assay and Scatchard analysis revealed that GE11 peptide bound specifically and efficiently to EGFR with a dissociation constant of approximately 22 nM, but with much lower mitogenic activity than with EGF. We showed that the peptides were internalized preferentially into EGFR highly expressing cells, and they accumulated in EGFR overexpressing tumor xenografts after i.v. delivery in vivo. In gene delivery studies, GE11-conjugated polyethylenimine (PEI) vectors were less mitogenic, but still quite efficient at transfecting genes into EGFR highly expressing cells and tumor xenografts. Taken together, GE11 is a potentially safe and efficient targeting moiety for selective drug delivery systems mediated through EGFR. Topics: Cell Line, Tumor; Deoxyribonucleases; Drug Delivery Systems; Epidermal Growth Factor; ErbB Receptors; Gene Transfer Techniques; Genetic Vectors; Humans; Infusions, Intravenous; Iodides; K562 Cells; Kinetics; Ligands; Neoplasm Transplantation; Oncogene Proteins v-erbB; Peptide Library; Peptides; Polyethyleneimine; Protein Binding; Tetrazolium Salts; Thiazoles; Transfection | 2005 |
3-OH flavone inhibition of epidermal growth factor-induced proliferaton through blocking prostaglandin E2 production.
Epidermal growth factor (EGF) has been shown to induce proliferation in cells, however, the role of prostaglandin E(2) (PGE(2)) plays in EGF-induced proliferation in still unclear. EGF and PGE(2) showed proliferation responses in epidermoid carcinoma cell A431 by MTT and [(3)H] thymidine incorporation assay. Activation of the EGF receptor and extracellular signal-regulated protein kinases (ERK1/2), but not p38 and JNK, appeared 10 min after EGF treatment, whereas total amounts of ERK1/2, p38 and JNK remained unchanged in A431 cells, accompanied by induction of COX-2 and PGE(2) production. PD98059, a specific ERK1/2 inhibitor, inhibited EGF-induced proliferation with concomitant decreases in ERK1/2 phosphorylation and COX-2/PGE(2) induction. Non-steroid anti-inflammatory drugs (NSAIDs) such as aspirin and diclofenac, a COX activity inhibitor, inhibited EGF-induced proliferation by blocking PGE(2) production. The addition of PGE(2) reversed the inhibitory effects of PD98059, aspirin, and diclofenac on EGF-induced proliferation. This suggests that COX-2/PGE(2) activation involves in EGF-induced proliferation and locates at the downstream of ERK1/2 activation. Furthermore, the natural product, 3-OH flavone, showed the most-potent inhibitory activity on EGF-induced proliferation among 9 structurally-related compounds, and suppression of EGF receptor phosphorylation, ERK1/2 phosphorylation, and COX-2/PGE(2) production by 3-OH flavone was identified. PGE(2) addition attenuates the inhibitory activity of 3-OH flavone on EGF-induced proliferation by MTT assay and colony formation by soft agar assay. Additionally, 3-OH flavone also showed more-specific inhibition on EGF- than on fetal bovine serum (FBS)-induced proliferation in A431 cells. Results of our present study provide evidence to demonstrate that PGE(2) is an important downstream molecule in EGF-induced proliferation, and 3-OH flavone, which inhibits PGE(2) production by blocking MAPK cascade, might reserve potential for development as an anti-cancer drug. Topics: Anti-Inflammatory Agents, Non-Steroidal; Carcinoma, Squamous Cell; Cell Division; Colony-Forming Units Assay; Cyclooxygenase 2; Cyclooxygenase 2 Inhibitors; Cyclooxygenase Inhibitors; Dinoprostone; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Flavonoids; Gene Expression Regulation, Enzymologic; Humans; Isoenzymes; JNK Mitogen-Activated Protein Kinases; MAP Kinase Kinase 4; Membrane Proteins; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; p38 Mitogen-Activated Protein Kinases; Prostaglandin-Endoperoxide Synthases; Tetrazolium Salts; Thiazoles; Tumor Cells, Cultured | 2004 |
Beta-parvin inhibits integrin-linked kinase signaling and is downregulated in breast cancer.
We analysed breast tumors and breast cancer cell lines for the expression of beta-parvin (ParvB), an adaptor protein that binds to the integrin-linked kinase (ILK). Quantitative RT-PCR indicated that ParvB mRNA was downregulated, by at least 60%, in four of nine breast tumors, relative to patient-matched normal mammary gland tissue. We also found that ParvB protein levels were reduced by > or =90% in five of seven advanced tumors, relative to matched normal breast tissue. Conversely, ILK protein and kinase activity levels were elevated in these tumors, suggesting that downregulation of ParvB stimulates ILK signaling. Western blot analyses indicated very low levels of ParvB protein in MDA-MB-231 and MCF7 breast cancer cells, facilitating functional studies of the effects of ParvB on ILK signaling. Expression of ParvB in MDA-MB-231 and MCF7 cells increased cell adhesion to collagen. ParvB inhibited ILK kinase activity, anchorage-independent cell growth and in vitro matrigel invasion by MDA-MB-231 cells. EGF-induced phosphorylation of two ILK targets, PKB (Ser473) and glycogen synthase kinase 3beta (Ser9), was also inhibited by ParvB. These results indicated that ParvB inhibits ILK signaling downstream of receptor tyrosine kinases. Our results suggest that loss of ParvB expression is a novel mechanism for upregulating ILK activity in tumors. Topics: Actinin; Adenoviridae; Amino Acid Sequence; Antibodies; Blotting, Western; Breast Neoplasms; Cell Adhesion; Cell Cycle; Cell Line, Tumor; Cell Proliferation; Collagen; Coloring Agents; DNA, Complementary; Down-Regulation; Drug Combinations; Epidermal Growth Factor; Genes, Reporter; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Laminin; Models, Genetic; Molecular Sequence Data; Phosphorylation; Plasmids; Protein Serine-Threonine Kinases; Protein Structure, Tertiary; Proteoglycans; Recombinant Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Tetrazolium Salts; Thiazoles; Transfection; Two-Hybrid System Techniques; Up-Regulation | 2004 |
Inhibition of NF-kappaB sensitizes A431 cells to epidermal growth factor-induced apoptosis, whereas its activation by ectopic expression of RelA confers resistance.
Epidermal growth factor (EGF) is a well known mitogen, but it paradoxically induces apoptosis in cells that overexpress its receptor. We demonstrate for the first time that the EGF-induced apoptosis is accelerated if NF-kappaB is inactivated. To inactivate NF-kappaB, human epidermoid carcinoma cells (A431) that overexpress EGF receptor were stably transfected with an IkappaB-alpha double mutant construct. Under the NF-kappaB-inactivated condition, A431 cells were more sensitive to EGF with decreased cell viability and increased externalization of phosphatidylserine on the cell surface, DNA fragmentation, and activation of caspases (3 and 8 but not 9), typical features of apoptosis. These results were further supported by the potentiation of the growth inhibitory effects of EGF by chemical inhibitors of NF-kappaB (curcumin and sodium salicylate) and the protective role of RelA evidenced by the resistance of A431-RelA cells (stably transfected with RelA) to EGF-induced apoptosis. EGF treatment or ectopic expression of RelA in A431 cells induced DNA binding activity of NF-kappaB (p50 and RelA) and the expression of c-IAP1, a downstream target of NF-kappaB. A431-RelA cells exhibited spontaneous phosphorylation of Akt (a downstream target of phosphatidylinositol 3-kinase and regulator of NF-kappaB) and EGF treatment stimulated it further. Blocking this basal Akt phosphorylation with LY294002, an inhibitor of phosphatidylinositol 3-kinase, did not affect their viability but blocking of EGF-induced phosphorylation of Akt sensitized the otherwise resistant A431-RelA cells to EGF-mediated growth inhibition. Our results favor an anti-apoptotic role for NF-kappaB in the regulation of EGF-induced apoptosis. Topics: Apoptosis; Blotting, Western; Caspase 3; Caspase 8; Caspase 9; Caspases; Cell Division; Cell Survival; Chromones; Coloring Agents; Comet Assay; DNA Fragmentation; Dose-Response Relationship, Drug; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; Humans; I-kappa B Proteins; Models, Biological; Morpholines; Mutation; NF-kappa B; NF-KappaB Inhibitor alpha; Phosphatidylserines; Poly(ADP-ribose) Polymerases; Tetrazolium Salts; Thiazoles; Thymidine; Transcription Factor RelA; Transfection; Tumor Cells, Cultured | 2003 |
Mitogenic properties of insulin-like growth factors I and II, insulin-like growth factor binding protein-3 and epidermal growth factor on human breast epithelial cells in primary culture.
Insulin-like growth factor-I (IGF-I) and insulin-like growth factor-II (IGF-II) are growth factors implicated in mammary gland development and are believed to be involved in breast cancer. However, the interactions between components of the IGF system and breast epithelial cells, which give rise to breast cancer, are not well understood. We have investigated the mitogenic properties of IGF-I, IGF-II, IGF binding protein-3 (IGFBP-3) and epidermal growth factor (EGF) on human breast epithelial cells (HBEC) in primary culture. We show that, under serum-free conditions, HBEC are stimulated to grow in response to IGF-I and IGF-II in a dose-dependent manner. IGF-I and EGF, a potent stimulator of HBEC growth in primary culture and also associated with breast cancer, appear to stimulate HBEC in a synergistic manner. IGFBP-3 inhibits the stimulation by IGF-I, IGF-II and IGF-I plus EGE In addition, it appears that IGFBP-3 has an inhibitory effect on HBEC growth that is IGF-independent. This study is the first to address the effects of IGF-I, IGF-II and IGFBP-3 alone and in combination with EGF on HBEC growth in primary culture. Characterizing the role of the IGF system in normal breast biology is significant because the system has been implicated in breast cancer and a number of the anti-estrogens used in treatment are believed to function through the IGF system. Topics: Biomarkers; Blotting, Western; Breast; Cell Division; Culture Media, Serum-Free; Dose-Response Relationship, Drug; Drug Synergism; Epidermal Growth Factor; Epithelial Cells; Female; Humans; Immunoenzyme Techniques; Insulin-Like Growth Factor Binding Protein 3; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Tetrazolium Salts; Thiazoles | 2002 |
The in vitro influence of eight hormones and growth factors on the proliferation of eight sarcoma cell lines.
Little is known about the regulation of sarcoma proliferation by hormones and/or growth factors. We therefore characterised the in vitro proliferative influence on eight sarcoma cell lines of the platelet-derived growth factor, the insulin-like growth factor 1, triiodothyronine, the epidermal growth factor, the luteinising-hormone-releasing hormone, progesterone, gastrin and 17 beta-oestradiol. The influence of the different factors on the proliferation of sarcoma cell lines was measured by the colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test. Two culture media were studied: (1) a nutritionally poor medium containing 2% of fetal calf serum and (2) a nutritionally rich one containing 5% or 10% FCS both with and without the addition of non-essential amino acids. The results were analysed either by conventional statistical analyses or by a classification method based on a decision-tree approach developed in Machine Learning. This latter method was also compared to other classifiers (such as logistic regression and k nearest neighbours) with respect to its accuracy of classification. Monovariate statistical analysis showed that each of the eight cell lines exhibited sensitivity to at least one factor, and each factor significantly modified the proliferation of five or six of the eight cell lines under study. Of these eight lines one of fibrosarcoma origin was the most "factor-sensitive". Decision-tree-related data analysis enabled the specific pattern of factor sensitivity to be characterised for the three histological types of cell line under study. The effects of hormone and growth factors are significantly influenced by the type of culture medium used. The method used appeared to be an accurate classifier for the kind of data analysed. Sarcoma proliferation can be modulated, at least in vitro, by various hormones and growth factors, and the proliferation of each histopathological type exhibited a distinct sensitivity to different hormone and/or growth-factors. Topics: Cell Division; Colorimetry; Culture Media; Epidermal Growth Factor; Estradiol; Fibrosarcoma; Gastrins; Gonadotropin-Releasing Hormone; Growth Substances; Hormones; Humans; Insulin-Like Growth Factor I; Leiomyosarcoma; Platelet-Derived Growth Factor; Progesterone; Reproducibility of Results; Rhabdomyosarcoma; Sensitivity and Specificity; Tetrazolium Salts; Thiazoles; Triiodothyronine; Tumor Cells, Cultured | 1998 |
Effect of growth factors on growth of bovine parathyroid cells in serum-free medium.
Published work has suggested a possible role of the epidermal growth factor receptor (EGFr) in parathyroid disease. Bovine parathyroid cells (BPCs) are frequently used as a tissue model for studying parathyroid disorders. We have studied the effect of the EGFr ligands EGF and transforming growth factor alpha (TGFalpha), alone and with insulin-like growth factors IGF-I and IGF-II on BPC growth. Experiments were run in triplicate and repeated three times. Cell numbers were assessed on day 5 by colorimetric MTT assay as well as by tritiated thymidine uptake. Results show that TGFalpha alone (p < 0.05) and IGF-I and IGF-II alone (p < 0.05) significantly stimulated growth over controls (t-test). Furthermore, the combination of TGFalpha with IGF-I and IGF-II exhibited significant enhancement above that seen with IGF-I and IGF-II alone (p < 0.01). EGF did not stimulate growth over controls. EGFr may be expressed in BPCs, but TGFalpha exhibits a more potent growth stimulus than EGF. Addition of IGF-I or IGF-II to the growth medium further enhances this effect. Topics: Animals; Cattle; Cell Count; Cell Division; Cells, Cultured; Colorimetry; Coloring Agents; Culture Media, Serum-Free; Drug Combinations; Drug Synergism; Epidermal Growth Factor; ErbB Receptors; Growth Substances; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Microscopy, Electron; Parathyroid Glands; Tetrazolium Salts; Thiazoles; Thymidine; Transforming Growth Factor alpha; Tritium | 1996 |
Androgens inhibit the proliferation of a variant of the human prostate cancer cell line LNCaP.
The paradoxical androgen response of R2, a subline of the human prostate cancer cell line LNCaP, is described here. Two androgens (DHT and R1881) decreased, in a dose-dependent manner, R2 cell proliferation and [3H]thymidine incorporation. These ligand and cell specific effects were accompanied by an increase in the metabolism of the vital dye MTT and in cell protein content. Both androgens increased the doubling time and the percentage of G0-G1 cells. No evidence of androgen-induced apoptosis was found. Cloning allowed the selection of two cell populations on the basis of the response to 10 nM of R1881. Long term culture of uncloned R2 cells with R1881 modified reversibly the pattern of androgen response. R2 was compared to the androgen-stimulated LNCaP-FGC subline to investigate the causes of their different androgen responsiveness. The androgen receptor (number, affinity for hormones and antihormones, sedimentation constant and molecular weight) and androgen receptor genes (exon size and exon 8 sequence) were found to be identical in the two sublines. EGF stimulated LNCaP-FGC but not R2. Both cells were slightly stimulated by basic FGF but were insensitive to IGF-I and TGF beta 1.. (1) androgens inhibit the proliferation of R2 cells possibly by introducing a G0-G1 block; (2) this inhibition is incomplete because, at least in part, the R2 cell population is heterogeneous; (3) chronic androgen treatment induces reversible cell adaptation; and (4) there is no evidence that the loss of the classical stimulatory effect of androgen on cell proliferation and the gain of inhibitory effect are due to androgen receptor alteration or to a specific action of one of the four growth factors tested. Topics: Amino Acid Sequence; Androgens; Base Sequence; Cell Division; Coloring Agents; Dihydrotestosterone; DNA, Neoplasm; Epidermal Growth Factor; Exons; Fibroblast Growth Factor 2; Humans; Male; Metribolone; Molecular Sequence Data; Molecular Weight; Neoplasm Proteins; Point Mutation; Prostatic Neoplasms; Receptors, Androgen; Testosterone Congeners; Tetrazolium Salts; Thiazoles; Tumor Cells, Cultured | 1995 |
Characterisation of the influence of anti-gastrin, anti-epidermal growth factor, anti-oestradiol, and anti-luteinising hormone releasing hormone antibodies on the proliferation of 27 cell lines from the gastrointestinal tract.
Numerous data from published reports prove that the proliferation of gastrointestinal tumour cell lines are under the control of many hormones or growth factors, or both. Most of these publications report the influence on a very small number of cell lines of one or two such factors only. This work deals with the in vitro characterisation of the influence of the anti-gastrin, the anti-epidermal growth factor (EGF), the anti-oestradiol (E2), and the anti-luteinising hormone releasing hormone (LHRH) antibodies on the proliferation of a large series of gastrointestinal cell lines. Cell proliferation was assessed by means of the colorimetric MTT assay on a series of 27 gastrointestinal cell lines obtained from the American Type Culture Collection (ATCC). Of the 27 cell lines, the anti-gastrin, the anti-EGF, the anti-E2, and the anti-LHRH neutralising antibodies considerably influenced the proliferation of 13, 25, 12, and 16. No gastrointestinal cell line was unresponsive to the four antibodies simultaneously. The anti-gastrin and anti-EGF antibody induced effects on the 27 gastrointestinal cell line proliferation were significantly correlated, as was also the case for the anti-E2 and anti-LHRH antibody induced effects. Of the anti-gastrin, the anti-EGF, the anti-E2, and the anti-LHRH antibodies, it was the anti-EGF one that had the greatest influence, both quantitatively and qualitatively, on gastrointestinal cell proliferation. The correlation of the effects of definite anti-hormone antibodies is suggestive of a common mechanism of action for the corresponding hormones and casts some doubt on the efficiency of anti-hormone monotherapy. Topics: Animals; Antibodies; Cell Division; Cell Line; Coloring Agents; Digestive System; Dose-Response Relationship, Immunologic; Epidermal Growth Factor; Estradiol; Gastrins; Gonadotropin-Releasing Hormone; Growth Substances; Guinea Pigs; Humans; Mice; Rats; Tetrazolium Salts; Thiazoles | 1995 |
Inhibitory effects of the gastrin receptor antagonist CR2093 on basal, gastrin-stimulated and growth factor-stimulated growth of the rat pancreatic cell line AR42J.
AR42J, a rat pancreatic cell line expressing receptors for both gastrin and epidermal growth factor (EGF), has been used to examine the effect of the gastrin receptor antagonist CR2093 on basal, gastrin-17 (G17), EGF and transforming growth factor (TGF)-alpha stimulated growth in vitro. In serum-free conditions, CR2093 reduced the basal growth of AR42J at a concentration known to displace physiological levels of G17 from gastrin receptors and this effect was reversed by G17 at 1 x 10(-9) M. Alone, G17 had little effect on growth, but EGF and TGF-alpha stimulated growth to 181 and 176% of control values, respectively, and marked synergy was observed when G17 was used in combination with both EGF (212%) and TGF-alpha (259%). When CR2093 was added, the synergistic effects of the G17/EGF and G17/TGF-alpha combinations were reduced to basal levels. In addition, CR2093 inhibited the growth stimulation induced by EGF and TGF-alpha alone. When the ability of CR2093 to bind to EGF receptors was assessed in a ligand binding assay, it was found that the antagonist displaced up to 23% of labeled EGF. Thus CR2093 has potent inhibitory effects on the basal growth of AR42J which can be reversed by G17. It can also inhibit type 1 growth factor-stimulated growth, but although this action may in part be related to the antagonist's ability to inhibit binding to the EGF receptor, other mechanisms may be involved. Topics: Amino Acids; Animals; Cell Division; Coloring Agents; Epidermal Growth Factor; Gastrins; Growth Inhibitors; Growth Substances; Humans; Iodine Radioisotopes; Pancreatic Neoplasms; Rats; Receptors, Cholecystokinin; Tetrazolium Salts; Thiazoles; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1994 |
Influence of different growth factors on a rat choriocarcinoma cell line.
The influence of epidermal growth factor, insulin-like growth factors I and II, insulin, transforming growth factor beta 1 and transferrin on the growth of a postgestational rat choriocarcinoma was examined by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay. The cell line was cultured in RPMI 1640 medium supplemented with fetal calf serum, beta-mercaptoethanol, glucose, sodium pyruvate and antibiotics. The experiments were done in media supplemented with 10% (optimal) or 3% (suboptimal) fetal calf serum. Among the different growth factors tested, only epidermal growth factor and to a certain extent insulin had a growth-promoting effect by themselves. The other growth factors had either an additive effect in the presence of epidermal growth factor or no effect at all. The cytotrophoblast cells expressed both epidermal growth factor and transferrin receptors whereas the more differentiated giant cells expressed only transferrin receptors. Topics: Animals; Cattle; Cell Differentiation; Cell Division; Choriocarcinoma; Colorimetry; Epidermal Growth Factor; ErbB Receptors; Fetal Blood; Growth Substances; Insulin; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Rats; Receptors, Transferrin; Tetrazolium Salts; Thiazoles; Transferrin; Transforming Growth Factor beta; Trophoblasts; Tumor Cells, Cultured | 1993 |
Disruption of cellular energy balance by suramin in intact human prostatic carcinoma cells, a likely antiproliferative mechanism.
The antiparasitic drug, suramin, has antiproliferative effects in human carcinoma cells. It has been suggested that this occurs through blockade of growth factor-receptor interactions. Three types of evidence that suramin rapidly inhibits cellular respiration or disrupts cellular energy balance in intact cells of the human prostate carcinoma cell line, DU145, are presented. Beginning at approximately 10(-4) M, suramin rapidly causes dose-dependent inhibition of tetrazolium conversion by mitochondrial dehydrogenases in intact cells, demonstrating an inhibition of respiration. This effect is reversed by exchange with suramin-free media but not by pretreatment with serum, epidermal growth factor, insulin-like growth factor I, acidic and basic fibroblast growth factors, or calcium. Rhodamine 123 (10 micrograms/ml) uptake by mitochondria in intact DU145 cells is inhibited in the presence of 10(-3) M suramin. Treatment with 10(-4)-10(-3) M suramin causes the loss of rhodamine 123 from cells with mitochondria prestained with rhodamine 123, indicating that suramin is acting as an ionophore or respiratory poison. Also shown by electron microscopy are progressive toxic changes in mitochondria of DU145 cells within 1 h after treatment with 10(-4) M suramin. These data indicate that in intact DU145 cells 10(-4) M suramin rapidly disrupts cellular energy balance or respiration as seen by three studies of mitochondrial state. Disruption of energy balance or respiration represents a likely antiproliferative mechanism, as is thought to be a primary mechanism for the action of suramin in parasitic diseases. This proposed mechanism of action for suramin can explain the most prominent observed clinical toxicities of nephrotoxicity, adrenal toxicity, coagulopathy, and demyelinating neuropathy. Topics: Biological Transport; Calcium; Carcinoma; Cell Division; Energy Metabolism; Epidermal Growth Factor; Humans; In Vitro Techniques; Male; Microscopy, Electron; Mitochondria; Oxidoreductases; Prostatic Neoplasms; Rhodamine 123; Rhodamines; Suramin; Tetrazolium Salts; Thiazoles; Tumor Cells, Cultured | 1991 |