Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Braz. j. infect. dis ; 24(2): 150-159, Mar.-Apr. 2020. tab, graf
Article in English | LILACS, ColecionaSUS | ID: biblio-1132431

ABSTRACT

ABSTRACT Background: Latent HIV-1 is a major hurdle in obtaining HIV-1 sustained virological remission (SVR). Here we explored histone deacetylation inhibition property of nicotinamide (NAM; n = 17) for the first time in comparison to a combination of methyltransferase inhibitors (MTIs; Chaetocin and BIX01294; n = 25) to reactivate latent HIV ex vivo in CD8-depleted PBMCs from antiretroviral treated aviremic individuals. Results: NAM reactivated HIV-1 from 13/17 (76.4%) samples compared to 20/25 (80.0%) using MTIs with mean viral load (VLs) of 4.32 and 3.22 log10 RNA copies/mL, respectively (p = 0.004). Mean purging time after NAM and MTIs stimulation was 5.1 and 6.75 days, respectively (p = 0.73). Viral purging in autologous cultures exhibited blunted HIV recovery with fluctuating VLs followed by a complete viral extinction when expanded in allogenic system. Electron microscopy from five supernatants revealed anomalous viral particles, with lack of complete viral genomes when characterized by ultradeep sequencing through metagenomics approach (n = 4). Conclusion: NAM alone was more potent HIV-1 activator than combination of MTIs, with potential of clinical use.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Young Adult , Quinazolines/pharmacology , Azepines/pharmacology , Virus Activation/drug effects , HIV Infections/virology , HIV-1/drug effects , Niacinamide/pharmacology , Methyltransferases/antagonists & inhibitors , Piperazines/pharmacology , Leukocytes, Mononuclear/virology , CD4-Positive T-Lymphocytes , Gene Expression Regulation, Viral , Virus Latency , Viral Load/drug effects , Viral Tropism/drug effects
2.
Int. braz. j. urol ; 44(3): 600-607, May-June 2018. tab, graf
Article in English | LILACS | ID: biblio-954052

ABSTRACT

ABSTRACT Objectives: Apoptosis effect of oral alpha-blockers is known in the prostate. Apoptosis index of silodosin has not been proved, yet. Aims are to present apoptosis index of silodosin in prostate and to compare this with other currently used alpha-blocker's apoptosis indexes together with their clinical effects. Materials and Methods: Benign prostatic hyperplasia (BPH) patients were enrolled among those admitted to urology outpatient clinic between June 2014 and June 2015. Study groups were created according to randomly prescribed oral alpha-blocker drugs as silodosin 8mg (Group 1; n=24), tamsulosin 0.4mg (Group 2; n=30), alfuzosin 10mg (Group 3; n=25), doxazosin 8mg (Group 4; n=22), terazosin 5mg (Group 5; n=15). Pa- tients who refused to use any alpha-blocker drug were included into Group 6 as control group (n=16). We investigated apoptosis indexes of the drugs in prostatic tissues that were taken from patient's surgery (transurethral resection of prostate) and/or prostate biopsies. Immunochemical dyeing, light microscope, and Image Processing and Analy- sis in Java were used for evaluations. Statistical significant p was p<0.05. Results: There were 132 patients with mean follow-up of 4.2±2.1 months. Pathologist researched randomly selected 10 areas in each microscope set. Group 1 showed statisti- cal significant difference apoptosis index in immunochemical TUNEL dyeing and im- age software (p<0.001). Moreover, we determined superior significant development in parameters as uroflowmetry, quality of life scores, and international prostate symptom score in Group 1. Conclusions: Silodosin has higher apoptosis effect than other alpha-blockers in prostate. Thus, clinic improvement with silodosin was proved by histologic studies. Besides, static factor of BPH may be overcome with creating apoptosis.


Subject(s)
Humans , Male , Aged , Aged, 80 and over , Prostate/drug effects , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/drug therapy , Apoptosis/drug effects , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Quinazolines/pharmacology , Reference Values , Sulfonamides/pharmacology , Time Factors , Biopsy , Prazosin/analogs & derivatives , Prazosin/pharmacology , Immunohistochemistry , Pilot Projects , Retrospective Studies , Treatment Outcome , Prostate-Specific Antigen/blood , Doxazosin/pharmacology , Tamsulosin , Indoles/pharmacology , Middle Aged
3.
Yonsei Medical Journal ; : 9-18, 2017.
Article in English | WPRIM | ID: wpr-222311

ABSTRACT

PURPOSE: Rearrangement of the proto-oncogene rearranged during transfection (RET) has been newly identified potential driver mutation in lung adenocarcinoma. Clinically available tyrosine kinase inhibitors (TKIs) target RET kinase activity, which suggests that patients with RET fusion genes may be treatable with a kinase inhibitor. Nevertheless, the mechanisms of resistance to these agents remain largely unknown. Thus, the present study aimed to determine whether epidermal growth factor (EGF) and hepatocyte growth factor (HGF) trigger RET inhibitor resistance in LC-2/ad cells with CCDC6-RET fusion genes. MATERIALS AND METHODS: The effects of EGF and HGF on the susceptibility of a CCDC6-RET lung cancer cell line to RET inhibitors (sunitinib, E7080, vandetanib, and sorafenib) were examined. RESULTS: CCDC6-RET lung cancer cells were highly sensitive to RET inhibitors. EGF activated epidermal growth factor receptor (EGFR) and triggered resistance to sunitinib, E7080, vandetanib, and sorafenib by transducing bypass survival signaling through ERK and AKT. Reversible EGFR-TKI (gefitinib) resensitized cancer cells to RET inhibitors, even in the presence of EGF. Endothelial cells, which are known to produce EGF, decreased the sensitivity of CCDC6-RET lung cancer cells to RET inhibitors, an effect that was inhibited by EGFR small interfering RNA (siRNA), anti-EGFR antibody (cetuximab), and EGFR-TKI (Iressa). HGF had relatively little effect on the sensitivity to RET inhibitors. CONCLUSION: EGF could trigger resistance to RET inhibition in CCDC6-RET lung cancer cells, and endothelial cells may confer resistance to RET inhibitors by EGF. E7080 and other RET inhibitors may provide therapeutic benefits in the treatment of RET-positive lung cancer patients.


Subject(s)
Humans , Adenocarcinoma/drug therapy , Cell Line, Tumor , Cetuximab/pharmacology , Drug Resistance, Neoplasm/drug effects , Epidermal Growth Factor/metabolism , Gene Rearrangement , Hepatocyte Growth Factor/pharmacology , Indoles/pharmacology , Lung Neoplasms/drug therapy , MAP Kinase Signaling System , Mutation , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Pyrroles/pharmacology , Quinazolines/pharmacology , RNA, Small Interfering/pharmacology , ErbB Receptors/genetics , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/metabolism
4.
The Korean Journal of Parasitology ; : 31-38, 2016.
Article in English | WPRIM | ID: wpr-36486

ABSTRACT

Specific gene expressions of host cells by spontaneous STAT6 phosphorylation are major strategy for the survival of intracellular Toxoplasma gondii against parasiticidal events through STAT1 phosphorylation by infection provoked IFN-γ. We determined the effects of small molecules of tyrosine kinase inhibitors (TKIs) on the growth of T. gondii and on the relationship with STAT1 and STAT6 phosphorylation in ARPE-19 cells. We counted the number of T. gondii RH tachyzoites per parasitophorous vacuolar membrane (PVM) after treatment with TKIs at 12-hr intervals for 72 hr. The change of STAT6 phosphorylation was assessed via western blot and immunofluorescence assay. Among the tested TKIs, Afatinib (pan ErbB/EGFR inhibitor, 5 µM) inhibited 98.0% of the growth of T. gondii, which was comparable to pyrimethamine (5 µM) at 96.9% and followed by Erlotinib (ErbB1/EGFR inhibitor, 20 µM) at 33.8% and Sunitinib (PDGFR or c-Kit inhibitor, 10 µM) at 21.3%. In the early stage of the infection (2, 4, and 8 hr after T. gondii challenge), Afatinib inhibited the phosphorylation of STAT6 in western blot and immunofluorescence assay. Both JAK1 and JAK3, the upper hierarchical kinases of cytokine signaling, were strongly phosphorylated at 2 hr and then disappeared entirely after 4 hr. Some TKIs, especially the EGFR inhibitors, might play an important role in the inhibition of intracellular replication of T. gondii through the inhibition of the direct phosphorylation of STAT6 by T. gondii.


Subject(s)
Humans , Antiparasitic Agents/pharmacology , Blotting, Western , Cell Line , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Phosphorylation/drug effects , Quinazolines/pharmacology , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , Toxoplasma/drug effects , Toxoplasmosis/physiopathology
5.
Clinical and Molecular Hepatology ; : 49-59, 2015.
Article in English | WPRIM | ID: wpr-64643

ABSTRACT

BACKGROUND/AIMS: Silibinin, the main component of silymarin, is used as a hepatoprotectant and exhibits anticancer effects against various cancer cells. This study evaluated the effects of a combination of silibinin with either gefitinib or sorafenib on hepatocellular carcinoma (HCC) cells. METHODS: Several different human HCC cell lines were used to test the growth-inhibiting effects and cell toxicity of silibinin both alone and in combination with either gefitinib or sorafenib. The cell viability and growth inhibition were assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, trypan blue staining, and a colony-forming assay. Furthermore, changes in epidermal growth factor receptor (EGFR)-related signals were evaluated by Western blot analysis. RESULTS: Gefitinib, sorafenib, and silibinin individually exhibited dose-dependent antiproliferative effects on HCC cells. Combined treatment with silibinin enhanced the gefitinib-induced growth-inhibiting effects in some HCC cell lines. The combination effect of gefitinib and silibinin was synergistic in the SNU761 cell line, but was only additive in the Huh-BAT cell line. The combination effect may be attributable to inhibition of EGFR-dependent Akt signaling. Enhanced growth-inhibiting effects were also observed in HCC cells treated with a combination of sorafenib and silibinin. CONCLUSIONS: Combined treatment with silibinin enhanced the growth-inhibiting effects of both gefitinib and sorafenib. Therefore, the combination of silibinin with either sorafenib or gefitinib could be a useful treatment approach for HCC in the future.


Subject(s)
Humans , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , ErbB Receptors/metabolism , Signal Transduction/drug effects , Silymarin/pharmacology
6.
The Korean Journal of Parasitology ; : 439-441, 2014.
Article in English | WPRIM | ID: wpr-70334

ABSTRACT

Toxoplasma gondii is the causative agent of toxoplasmosis with symptoms of congenital neurological and ocular diseases and acquired lymphadenitis, retinochoroiditis, and meningoencephalitis. Small molecules which block the activity of protein kinases were tested in in vitro culture of T. gondii to find new therapeutic drugs of safer and more effective than the combined administration of pyrimethamine and sulfadoxine that sometimes provoke lethal Stevens-Johnson syndrome. Among them, Gefitinib and Crizotinib inhibited intracellular growth of T. gondii in HeLa cells by counting the number of T. gondii per parasitophorous vacuolar membrane whereas Sunitinib did not. Gefitinib inhibited the growth of T. gondii in a dose-dependent manner over 5 microM up to the tolerable concentration of HeLa cells and halted the division of the parasite immediately from the time point of treatment. Gefitinib inhibition suggests that tyrosine kinases of EGFR family or other homologous kinases of the parasite itself may be the target to cause the block of T. gondii growth.


Subject(s)
Humans , Antiprotozoal Agents/pharmacology , Dose-Response Relationship, Drug , Drug Repositioning , HeLa Cells , Parasitic Sensitivity Tests , Quinazolines/pharmacology , Toxoplasma/drug effects
7.
Experimental & Molecular Medicine ; : e23-2013.
Article in English | WPRIM | ID: wpr-159138

ABSTRACT

Recent studies have documented that Janus-activated kinase (JAK)-signal transducer and activator of transcription (STAT) pathway can modulate the apoptotic program in a myocardial ischemia/reperfusion (I/R) model. To date, however, limited studies have examined the role of JAK3 on myocardial I/R injury. Here, we investigated the potential effects of pharmacological JAK3 inhibition with JANEX-1 in a myocardial I/R model. Mice were subjected to 45 min of ischemia followed by varying periods of reperfusion. JANEX-1 was injected 1 h before ischemia by intraperitoneal injection. Treatment with JANEX-1 significantly decreased plasma creatine kinase and lactate dehydrogenase activities, reduced infarct size, reversed I/R-induced functional deterioration of the myocardium and reduced myocardial apoptosis. Histological analysis revealed an increase in neutrophil and macrophage infiltration within the infarcted area, which was markedly reduced by JANEX-1 treatment. In parallel, in in vitro studies where neutrophils and macrophages were treated with JANEX-1 or isolated from JAK3 knockout mice, there was an impairment in the migration potential toward interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1), respectively. Of note, however, JANEX-1 did not affect the expression of IL-8 and MCP-1 in the myocardium. The pharmacological inhibition of JAK3 might represent an effective approach to reduce inflammation-mediated apoptotic damage initiated by myocardial I/R injury.


Subject(s)
Animals , Male , Mice , Apoptosis/drug effects , Cell Movement/drug effects , Chemokines/pharmacology , Heart Function Tests/drug effects , Inflammation/pathology , Janus Kinase 3/antagonists & inhibitors , Macrophages/drug effects , Mice, Inbred C57BL , Myocardial Reperfusion Injury/drug therapy , Myocardium/enzymology , Myocytes, Cardiac/drug effects , Neutrophils/drug effects , Quinazolines/pharmacology
8.
Experimental & Molecular Medicine ; : e64-2013.
Article in English | WPRIM | ID: wpr-152458

ABSTRACT

Lapatinib, a dual inhibitor of epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) tyrosine kinases, has shown promising results as a growth inhibitor of HER2-positive cancer cells in vitro. However, similar to other EGFR-targeting drugs, acquired resistance to lapatinib by HER2-positive cancer cells remains a major clinical challenge. To elucidate resistance mechanisms to EGFR/HER2-targeting agents, we performed a systematic quantitative comparison of the phosphoproteome of lapatinib-resistant (LR) human gastric cancer cells (SNU216-LR) versus parental cells (SNU216) using a titanium dioxide (TiO2) phosphopeptide enrichment method and analysis with a Q-Exactive hybrid quadrupole-Orbitrap mass spectrometer. Biological network analysis of differentially expressed phosphoproteins revealed apparent constitutive activation of the MET-axis phosphatidylinositide 3-kinase (PI3K)/alpha-serine/threonine-protein kinase (AKT) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathways in SNU216-LR. Inhibition of the PI3K/AKT and MAPK/ERK signaling pathways in SNU216-LR also leads to cell cycle arrest, confirming the biological network analysis. Lapatinib sensitivity was restored when cells were treated with several molecular targeting agents in combination with lapatinib. Thus, by integrating phosphoproteomic data, protein networks and effects of signaling pathway modulation on cell proliferation, we found that SNU216-LR maintains constitutive activation of the PI3K/AKT and MAPK/ERK pathways in a MET-dependent manner. These findings suggest that pathway activation is a key compensatory intracellular phospho-signaling event that may govern gastric cancer cell resistance to drug treatment.


Subject(s)
Humans , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology
9.
Pakistan Journal of Pharmaceutical Sciences. 2012; 25 (3): 651-656
in English | IMEMR | ID: emr-144420

ABSTRACT

Vasicine [1] was isolated from the ethanolic extract of Adhatoda vasica Nees [Acanthaceae] and the structure was confirmed using spectroscopic techniques. Acetylcholine esterase, trypsin, DPPH inhibition potential and FRAP assay were carried out using in vitro models. The results showed 38.4 +/- 1.2% and 37.4 +/- 1.1% activity in acetylcholine and trypsin inhibition assays respectively. The compound [1] exhibited significant DPPH inhibition activity [70.4 +/- 1.3%, IC[50] = 212.3 +/- 1.9 microM]. A dose dependant behavior of vasicine [1], was indicated in the FRAP assay. Antibacterial activity was checked according to agar well diffusion assay and results revealed that vasicine [1] showed moderate activity


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Quinazolines/pharmacology , /pharmacology , Alkaloids/chemistry , Microbial Sensitivity Tests , Biphenyl Compounds/antagonists & inhibitors , Picrates/antagonists & inhibitors
10.
Experimental & Molecular Medicine ; : 473-482, 2012.
Article in English | WPRIM | ID: wpr-192555

ABSTRACT

Overexpression of HER2 correlates with more aggressive tumors and increased resistance to cancer chemotherapy. However, a functional comparison between the HER2high/HER3 and the HER2low/HER3 dimers on tumor metastasis has not been conducted. Herein we examined the regulation mechanism of heregulin-beta1 (HRG)-induced MMP-1 and -9 expression in breast cancer cell lines. Our results showed that the basal levels of MMP-1 and -9 mRNA and protein expression were increased by HRG treatment. In addition, HRG-induced MMP-1 and -9 expression was significantly decreased by MEK1/2 inhibitor, U0126 but not by phosphatidylinositol 3-kinase (PI-3K) inhibitor, LY294002. To confirm the role of MEK/ERK pathway on HRG-induced MMP-1 and -9 expression, MCF7 cells were transfected with constitutively active adenoviral-MEK (CA-MEK). The level of MMP-1 and -9 expressions was increased by CA-MEK. MMP-1 and -9 mRNA and protein expressions in response to HRG were higher in HER2 overexpressed cells than in vector alone. The phosphorylation of HER2, HER3, ERK, Akt, and JNK were also significantly increased in HER2 overexpressed MCF7 cells compared with vector alone. HRG-induced MMP-1 and -9 expressions were significantly decreased by lapatinib, which inhibits HER1 and HER2 activity, in both vector alone and HER2 overexpressed MCF7 cells. Finally, HRG-induced MMP-1 and MMP-9 expression was decreased by HER3 siRNA overexpression. Taken together, we suggested that HRG-induced MMP-1 and MMP-9 expression is mediated through HER3 dependent pathway and highly expressed HER2 may be associated with more aggressive metastasis than the low expressed HER2 in breast cancer cells.


Subject(s)
Female , Humans , Breast Neoplasms/enzymology , Butadienes/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Signaling System , MCF-7 Cells , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 9/genetics , Neuregulin-1/pharmacology , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-3/metabolism
12.
West Indian med. j ; 60(2): 107-113, Mar. 2011. ilus
Article in English | LILACS | ID: lil-672734

ABSTRACT

BACKGROUND AND AIMS: Interferon-α (IFN-α) treatment is associated with up-regulation of epidermal growth factor receptor (EGFR) expression and marked growth inhibition of colon cancer cell lines. We aimed to determine the effect of combining IFN-α and gefitinib in the growth of human colon cancer cell lines. METHODS: Two human colon cancer cell lines SW480 and LOVO were treated with IFN-α alone or gefitinib alone or IFN-α plus gefitinib. Proliferation of colon cancer cells was measured by methyl thiazolyl tetrazolium (MTT) assay; the apoptosis rate was analysed by flow cytometry (FCM). The expression of XIAP, XAF1 mRNA was detected by RT-PCR and the expression of XIAP, XAF1 protein was detected by western blotting. RESULTS: Methyl thiazolyl tetrazolium showed that IFN-α, gefitinib and IFN-α plus gefitinib significantly inhibited SW480 and LOVO cells in a dose-dependent manner (p < 0.05). The FCM revealed that IFN α, gefitinib and IFN-α plus gefitinib could markedly upgrade the apoptosis rate (p < 0.05). The expression of XIAP mRNA down-regulated markedly (p < 0.05) while the expression of XAF1 mRNA up-regulated significantly (p < 0.05). The expression of XIAP protein was down-regulated markedly (p < 0.05) while the expression of XAF1 protein was up-regulated significantly (p < 0.05). CONCLUSION: IFN-α promotes the antiproliferaative effect of gefitinib on human colon cancer cell lines and the mechanism may be related to up-regulation expression of EGFR by IFN-α.


ANTECEDENTES Y OBJETIVOS: El tratamiento con interferón α (IFN-α) se halla asociado con la regulación por incremento de la expresión del receptor del factor de crecimiento epidérmico y la acentuada inhibición del crecimiento de las líneas celulares del cáncer colorrectal. El presente trabajo tuvo por objetivo determinar el efecto que se produce al combinar el IFN-α y el gefitinib en el crecimiento de las líneas celulares del cáncer de colon. MÉTODOS: Dos líneas celulares de cáncer del colon en humanos - SW480 y LOVO - fueron tratadas con IFN-α solamente, gefitinib solamente, o IFN-α más gefitinib. La proliferación de las células cancerosas del colon se midió mediante ensayo de metil tiazolil tetrazolio (MTT); la tasa de apoptosis se analizó mediante citometría de flujo (CMF); la expresión de XIAP/XAF1 mRNA fue detectada mediante RT-PCR y la expresión de la proteína XIAP/XAF1 fue detectada mediante inmunoblot (western blot). RESULTADOS: El MTT mostró que el IFN-α, el gefitinib, y el IFN-α más gefitinib inhibían de forma significativa las células SW480 y LOVO en dependencia de la dosis (p < 0.05). La CMF reveló que el IFN-α, el gefitinib, y el IFN-α más gefitinib podían aumentar notablemente la tasa de apoptosis (p < 0.05). La expresión de XIAP mRNA tuvo una marcada regulación por decremento (p < 0.05) mientras que la expresión de XAF1 mRNA tuvo una significativa regulación por incremento (p < 0.05); la expresión de la proteína XIAP fue notablemente regulada por decremento (p < 0.05) mientras que la expresión de la proteína XAF1 fue regulada por incremento de manera significativa (p < 0.05). CONCLUSIÓN: El IFN-α promueve el efecto antiproliferativo del gefitinib sobre las líneas celulares del cáncer colorrectal, y el mecanismo puede hallarse relacionado con la expresión de la regulación por incremento del EGFR mediante el IFN-α.


Subject(s)
Humans , Antineoplastic Agents/pharmacology , Colonic Neoplasms/pathology , Interferon-alpha/pharmacology , Quinazolines/pharmacology , ErbB Receptors/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , ErbB Receptors/metabolism
13.
Journal of Korean Medical Science ; : 214-221, 2011.
Article in English | WPRIM | ID: wpr-123288

ABSTRACT

Hepatitis B virus X (HBx) protein has been known to play an important role in development of hepatocellular carcinoma (HCC). The aim of this study is to find out whether HBx protein expression affects antiproliferative effect of an epidermal growth factor receptor-tyrosine kinase (EGFR-TK) inhibitor and a MEK inhibitor in HepG2 and Huh-7 cell lines. We established HepG2 and Huh-7 cells transfected stably with HBx gene. HBx protein expression increased pERK and pAkt expression as well as beta-catenin activity in both cells. Gefitinib (EGFR-TK inhibitor) inhibited pERK and pAkt expression and beta-catenin activity in both cells. Selumetinib (MEK inhibitor) reduced pERK level and beta-catenin activity but pAkt expression was rather elevated by selumetinib in these cells. Reduction of pERK levels was much stronger with selumetinib than gefitinib in both cells. The antiproliferative efficacy of selumetinib was more potent than that of gefitinib. However, the antiproliferative effect of gefitinib, as well as selumetinib, was not different between cell lines with or without HBx expression. Signal pathway activation by HBx might not be strong enough to attenuate the antiproliferative effect of EGFR-TK inhibitor. Future experiments are needed to understand the role of HBx protein expression in HCC treatment using molecular targeting agent.


Subject(s)
Animals , Humans , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , Quinazolines/pharmacology , ErbB Receptors/antagonists & inhibitors , Signal Transduction/drug effects , Trans-Activators/metabolism , beta Catenin/metabolism
14.
J Biosci ; 2008 Dec; 33(5): 731-42
Article in English | IMSEAR | ID: sea-110807

ABSTRACT

Overexpression of epidermal growth factor receptor (EGFR) is a common phenomenon observed in most cancers. Clinical treatment of such cancer involves the use of chemotherapeutic agents such as ge ? tinib and erlotinib which are inhibitors of tyrosine kinase (TK). These small molecules bind to the ATP-binding sites of the TK domain of EGFR.Our in silico analysis suggests that the TK domains of Drosophila and human EGFR are highly conserved. We therefore employed the Drosophila system to validate the in silico observations made with two important anticancer drugs.Since a large number of mutant flies are available,it was possible to investigate the various components of the EGFR/Ras/Raf/MAPK pathways and the phosphorylation status of diphosphorylated extracellular signal-regulated kinase (dp-ERK1/2). These studies confirm the binding of the anilinoquinazolines to the Drosophila EGFR protein and modulation of its activity. Thus,Drosophila appears to be a robust and simple model system for screening newer anticancer drugs that act as TK inhibitors (TKIs).


Subject(s)
Animals , Antineoplastic Agents/pharmacology , Biological Assay , Catalytic Domain , Computer Simulation , Drosophila melanogaster/growth & development , Eye/anatomy & histology , Gene Expression Regulation/drug effects , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Quinazolines/pharmacology , ErbB Receptors/antagonists & inhibitors , /abnormalities , ras Proteins/antagonists & inhibitors
15.
Biol. Res ; 41(1): 43-50, 2008. ilus, tab
Article in English | LILACS | ID: lil-490631

ABSTRACT

This study describes the effect of novel 6-Arylbenzimidazo [1,2-c] quinazoline derivatives as tumor necrosis factor alpha (TNF-á) production inhibitors. The newly synthesized compounds were tested for their in vitro ability to inhibit the lipolysaccharide (LPS) induced TNF-á secretion in the human promyelocytic cell line HL-60. The compound 6-Phenyl-benzimidazo [1,2-c] quinazoline, coded as Gl, resulted as the most potent inhibitor and with no significant cytotoxic activity. Thus, 6-Arylbenzimidazo [1,2-c] quinazoline derivatives may have a potential as anti-inflammatory agents.


Subject(s)
Humans , Anti-Inflammatory Agents/pharmacology , Quinazolines/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Anti-Inflammatory Agents/chemistry , Lipopolysaccharides/pharmacology , Quinazolines/chemistry , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
16.
Experimental & Molecular Medicine ; : 524-534, 2007.
Article in English | WPRIM | ID: wpr-174050

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a prototypic halogenated aromatic hydrocarbon (HAH), is known as one of the most potent toxicants. At least a part of its toxic effects appears to be derived from its ability to induce TNF-alpha production. However, the signaling pathway of TCDD that leads to TNF-alpha expression has not been elucidated. In this study, we investigated the signaling mechanism of TCDD-induced TNF-alpha expression in PMA-differentiated THP-1 macrophages. TCDD induced both mRNA and protein expression of TNF-alpha in a dose- and time-dependent manner. Alpha-Naphthoflavone (NF), an aryl hydrocarbon receptor (AhR) inhibitor, prevented the TCDD-induced expression of TNF-alpha at both mRNA and protein levels. Genistein, a protein tyrosine kinase (PTK) inhibitor, and PD153035, an EGFR inhibitor, also blocked the increase of TNF-alpha expression by TCDD, indicating the role of EGFR in TCDD-induced TNF-alpha expression. On the other hand, PP2, a c-Src specific inhibitor, did not affect TCDD-induced TNF-alpha expression. EGFR phosphorylation was detected as early as 5 min after TCDD treatment. TCDD-induced EGFR activation was AhR-dependent since co-treatment with alpha-NF prevented it. ERK was found to be a downstream effector of EGFR activation in the signaling pathway leading to TNF-alpha production after TCDD stimulation. Activation of ERK was observed from 30 min after TCDD treatment. PD98059, an inhibitor of the MEK-ERK pathway, completely prevented the TNF-alpha mRNA and protein expression induced by TCDD, whereas inhibitors of JNK and p38 MAPK had no effect. PD153035, an EGFR inhibitor, as well as alpha-NF significantly reduced ERK phosphorylation, suggesting that ERK activation by TCDD was mediated by both EGFR and AhR. These results indicate that TNF-alpha production by TCDD in differentiated THP-1 macrophages is AhR-dependent and involves activation of EGFR and ERK, but not c-Src, JNK, nor p38 MAPK. A signaling pathway is proposed where TCDD induces sequential activation of AhR, EGFR and ERK, leading to the increased expression of TNF-alpha.


Subject(s)
Animals , Humans , Mice , Benzoflavones/pharmacology , Cell Differentiation , Cell Line, Tumor , Enzyme Activation , Genistein/pharmacology , Hazardous Substances/toxicity , MAP Kinase Signaling System/drug effects , Macrophages/metabolism , Phosphorylation , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA, Messenger/metabolism , ErbB Receptors/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Signal Transduction , Polychlorinated Dibenzodioxins/toxicity , Tumor Necrosis Factor-alpha/biosynthesis , src-Family Kinases/antagonists & inhibitors
17.
Indian J Biochem Biophys ; 2005 Dec; 42(6): 358-65
Article in English | IMSEAR | ID: sea-26594

ABSTRACT

Epidermal growth factor receptor (HER1/EGFR)-mediated signal transduction pathways are important in cellular response to ionizing radiation. High HER1/EGFR expression on cancer cells may contribute to radioresistance. In this pre-clinical study, we evaluated the radiosensitizing effect of erlotinib, a small molecule HER1/EGFR inhibitor in three human cancer cell lines with different HER1/EGFR expression--A431 (very high expression), H157 (moderate expression) and H460 (low expression). Our results demonstrated that A431 was the most radioresistant, while H460 was the most radiosensitive. However, A431 cells were the most sensitive to erlotinib (IC50 = 300 nM) and H460 cells the most resistant (IC50 = 8 microM). H157 had intermediate sensitivity to radiation and erlotinib (IC50 = 3 microM). With 300 nM erlotinib, the radiation dose enhancement ratios (DER) were 1.40, 1.17 and 1.04 in A431, H157 and H460, respectively. Treatment with erlotinib for 24 hr at 300 nM increased G1 arrest by 18.6, 2.0 and 4.8% in A431, H157 and H460, respectively. Erlotinib-induced apoptosis was augmented by radiation in A431 cells only. In conclusion, high HER1/EGFR expression may result in a high degree of radiosensitization with erlotinib combined with radiation. The extent of erlotinib-induced radiosensitization was proportional to HER1/EGFR expression, as well as autophosphorylation of the human epidermal growth factor receptor (HER1/EGFR).


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Lung Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Tumor Cells, Cultured
18.
SPJ-Saudi Pharmaceutical Journal. 2005; 8 (4): 198-204
in English | IMEMR | ID: emr-75119

ABSTRACT

The synthesis and biological evaluation of many 3-[4-[[[[cyckohexylamino] carbonyl]amino]sulfonyl]phenyl-4[3H]-quainzolinones [IVa-t] as oral hypoglycemics are described. Derivates [IVf], [IVg], [IVh], [IVj], [IVk], [IVm], [IVq] and [IVr] showed appreciable glucose lowering effects in alloxanized diabetic rats at a dose of 5 mg/kg. They decreased the blood glucose level by 40.7, 36.4, 37.2, 35.4, 43.7, 38.8, 35.3, 42.9 and 45.9 respectively. The hypoglycemic action of the synthesized compounds is comparable to, and in few cases is higher than that exhibited by the well known antidiabetic drug "glyburide" which showed 35.1 percent decrease of the blood glucose level


Subject(s)
Quinazolines/chemistry , Quinazolines/pharmacology , Sulfonylurea Compounds , Hypoglycemic Agents , Glyburide
19.
São Paulo med. j ; 122(3): 128-130, May 2004. ilus, tab
Article in English | LILACS | ID: lil-366405

ABSTRACT

CONTEXTO: Pacientes com carcinoma de pulmão não-pequenas células metastático são considerados incuráveis mas podem se beneficiar de quimioterapia sistêmica paliativa. Recentemente, com o advento de inibidores do receptor do fator de crescimento epitelial (EGFR), uma alternativa eficiente e menos tóxica surgiu para o tratamento destes pacientes. OBJETIVO: Testar em nosso meio a atividade e toxicidade do gefitinib. TIPO DE ESTUDO: Prospectivo, não aleatorizado e não controlado. LOCAL: Clínica de Oncologia e Hematologia (CLIOH), São Paulo, Brasil. PACIENTES E MÉTODOS: De junho de 2002 a abril de 2003, tratamos cinco pacientes com carcinoma de pulmão não-pequenas células metastático e previamente expostos a quimioterapia sistêmica (mediana do número de esquemas quimioterápicos prévios = 2) com gefitinib na dose de 250 mg ao dia, oralmente, no contexto de um protocolo de disponibilização da droga patrocinado pela AstraZeneca. A idade média dos pacientes foi de 65 anos, dois eram de sexo masculino e três tinham um estado funcional de 1, um de 2 e um de 3 na escala do ECOG (Eastern Cooperative Oncology Group). RESULTADOS: Observamos erupção cutânea em dois, diarréia em três e artralgias em dois pacientes. Notamos uma resposta parcial e um paciente teve estabilização de sua doença com durações de 11+ e 4+ meses, respectivamente, ambas acompanhadas de significativa queda em seus marcadores tumorais. Três pacientes progrediram durante o tratamento. DISCUSSAO: Novas opções de agentes quimioterápicos com perfis de toxicidade mais favoráveis são urgentemente necessárias para o tratamento de pacientes com câncer de pulmão não-pequenas células, que normalmente têm baixa expectativa de vida. Na nossa pequena série de cinco pacientes, observamos estabilização da doença em dois e reações de pele e gastrintestinais, freqüentemente descritas na literatura, em todos. Dois tiveram artralgia, não reportada até o momento. CONCLUSÕES: Concluímos que o gefitinib constitui importante adição ao armamentário terapêutico para tratamento de casos avançados de carcinoma de pulmão não-pequenas células.


Subject(s)
Humans , Male , Female , Middle Aged , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local , Quinazolines/pharmacology , ErbB Receptors/drug effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Lung Neoplasms/pathology , Prospective Studies , Quinazolines/adverse effects , Quinazolines/therapeutic use
20.
Indian J Physiol Pharmacol ; 2000 Jan; 44(1): 75-81
Article in English | IMSEAR | ID: sea-108959

ABSTRACT

The present study presents a mode of action profile of RLX (6, 7, 8, 9, 10, 12-hexahydro-azepino-[2, 1-b]-quinazoline-12-one) a bronchodilator obtained by the chemical modification in the molecule of alkaloid vasicine (Ex: Adhatoda vesica). The effect of RLX (p.o.) was observed on: (a) mast cell degranulation, (b) release of histamine and prostaglandin E (PGE), (c) 45Ca uptake and (d) activities of cAMP phosphodiesterase (PDEase) and lipoxygenase enzymes in mesenteries/peritoneal mast cells/lung tissue homogenates in rats under systemic anaphylaxis. RLX (10 and 20 mg/kg) inhibited antigen-induced mast cell degranulation and released of histamine from target tissues. An increased outflow of PGE (lungs) and an inhibited 45Ca uptake (peritoneal mast cells) were noted. Lung PDEase and lipoxygenase activities were decreased. These results suggested that RLX could be acting like disodium cromoglycate and aminophyline with additional attributes its oral efficacy and long duration of action.


Subject(s)
Anaphylaxis/prevention & control , Animals , Bronchodilator Agents/pharmacology , Calcium/metabolism , Cell Degranulation/drug effects , Electrophoresis, Polyacrylamide Gel , Histamine Release/drug effects , Horses/immunology , Hypersensitivity/immunology , Lung/drug effects , Male , Mast Cells/drug effects , Quinazolines/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL