1.Dexmedetomidine Attenuates High Glucose-induced HK-2 Epithelial-mesenchymal Transition by Inhibiting AKT and ERK.
Qi Zheng PAN ; Kai LI ; Zhuo Dong YANG ; Ming GAO ; Jia Hong SHI ; Shu Ping REN ; Guo Qing ZHAO
Biomedical and Environmental Sciences 2020;33(5):323-330
Objective:
To explore the protective effects of dexmedetomidine (Dex) against high glucose-induced epithelial-mesenchymal transition in HK-2 cells and relevant mechanisms.
Methods:
HK-2 cells were exposed to either glucose or glucose+Dex for 6 h. The production of ROS, morphology of HK-2 cells, and cell cycle were detected. Moreover, the expression of AKT, p-AKT, ERK, p-ERK, PI3K, E-Cadherin, Claudin-1, and α-SMA were determined and compared between HK-2 cells exposed to glucose and those exposed to both glucose and Dex with or without PI3K/AKT pathway inhibitor LY294002 and ERK pathway inhibitor U0126.
Results:
Compared with HK-2 cells exposed to high level of glucose, the HK-2 cells exposed to both high level of glucose and Dex showed: (1) lower level of ROS production; (2) cell morphology was complete; (3) more cells in G1 phase; (4) lower expression of p-AKT, p-ERK and α-SMA, higher expression of E-Cadherin and Claudin-1. PI3K/AKT inhibitor LY294002 and ERK inhibitor U0126 decreased the expression of p-AKT, p-ERK and α-SMA, and increased the expression of E-Cadherin and Claudin-1.
Conclusion
Dex can attenuate high glucose-induced HK-2 epithelial-mesenchymal transition by inhibiting AKT and ERK.
Adrenergic alpha-2 Receptor Agonists
;
pharmacology
;
Cell Line
;
Dexmedetomidine
;
pharmacology
;
Epithelial-Mesenchymal Transition
;
drug effects
;
Glucose
;
metabolism
;
Humans
;
MAP Kinase Signaling System
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
antagonists & inhibitors
;
Signal Transduction
;
drug effects
2.RGFP966 inactivation of the YAP pathway attenuates cardiac dysfunction induced by prolonged hypothermic preservation.
Xiao-He ZHENG ; Lin-Lin WANG ; Ming-Zhi ZHENG ; Jin-Jie ZHONG ; Ying-Ying CHEN ; Yue-Liang SHEN
Journal of Zhejiang University. Science. B 2020;21(9):703-715
Oxidative stress and apoptosis are the key factors that limit the hypothermic preservation time of donor hearts to within 4-6 h. The aim of this study was to investigate whether the histone deacetylase 3 (HDAC3) inhibitor RGFP966 could protect against cardiac injury induced by prolonged hypothermic preservation. Rat hearts were hypothermically preserved in Celsior solution with or without RGFP966 for 12 h followed by 60 min of reperfusion. Hemodynamic parameters during reperfusion were evaluated. The expression and phosphorylation levels of mammalian STE20-like kinase-1 (Mst1) and Yes-associated protein (YAP) were determined by western blotting. Cell apoptosis was measured by the terminal deoxynucleotidyl-transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) method. Addition of RGFP966 in Celsior solution significantly inhibited cardiac dysfunction induced by hypothermic preservation. RGFP966 inhibited the hypothermic preservation-induced increase of the phosphorylated (p)-Mst1/Mst1 and p-YAP/YAP ratios, prevented a reduction in total YAP protein expression, and increased the nuclear YAP protein level. Verteporfin (VP), a small molecular inhibitor of YAP-transcriptional enhanced associate domain (TEAD) interaction, partially abolished the protective effect of RGFP966 on cardiac function, and reduced lactate dehydrogenase activity and malondialdehyde content. RGFP966 increased superoxide dismutase, catalase, and glutathione peroxidase gene and protein expression, which was abolished by VP. RGFP966 inhibited hypothermic preservation-induced overexpression of B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax) and cleaved caspase-3, increased Bcl-2 mRNA and protein expression, and reduced cardiomyocyte apoptosis. The antioxidant and anti-apoptotic effects of RGFP966 were cancelled by VP. The results suggest that supplementation of Celsior solution with RGFP966 attenuated prolonged hypothermic preservation-induced cardiac dysfunction. The mechanism may involve inhibition of oxidative stress and apoptosis via inactivation of the YAP pathway.
Acrylamides/pharmacology*
;
Animals
;
Apoptosis/drug effects*
;
Cryopreservation
;
Disaccharides/pharmacology*
;
Electrolytes/pharmacology*
;
Glutamates/pharmacology*
;
Glutathione/pharmacology*
;
Heart/physiology*
;
Heart Transplantation/methods*
;
Hepatocyte Growth Factor/antagonists & inhibitors*
;
Histidine/pharmacology*
;
Histone Deacetylase Inhibitors/pharmacology*
;
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors*
;
Male
;
Mannitol/pharmacology*
;
Oxidative Stress/drug effects*
;
Phenylenediamines/pharmacology*
;
Proto-Oncogene Proteins/antagonists & inhibitors*
;
Rats
;
Rats, Sprague-Dawley
;
Signal Transduction/drug effects*
;
YAP-Signaling Proteins
3.Overexpression of autophagy-related gene 3 promotes autophagy and inhibits salinomycin-induced apoptosis in breast cancer MCF-7 cells.
Fang LI ; Guo HUANG ; Ping PENG ; Yao LIU ; Shuanghui LI ; Luogen LIU ; Yunsheng ZHANG
Journal of Southern Medical University 2019;39(2):162-168
OBJECTIVE:
To study the effects of the overexpression of autophagy-related gene 3 (ATG3) on autophagy and salinomycin-induced apoptosis in breast cancer cells and explore the underlying mechanisms.
METHODS:
We used the lentivirus approach to establish a breast cancer cell line with stable overexpression of ATG3. Western blotting, immunofluorescence staining and transmission electron microscopy were used to analyze the effect of ATG3 overexpression on autophagy in breast cancer MCF-7 cells. Using the AKT/mTOR agonists SC79 and MHY1485, we analyzed the effect of AKT/mTOR signal pathway activation on ATG3 overexpression-induced autophagy. Western blotting and flow cytometry were used to analyze the effect of autophagy on apoptosis of the ATG3-overexpressing cells treated with salinomycin and 3-MA (an autophagy inhibitor).
RESULTS:
In ATG3-overexpressing MCF-7 cells, ATG3 overexpression obviously promoted autophagy, inhibited the AKT/mTOR signaling pathway, significantly weakened salinomycin-induced apoptosis ( < 0.01), caused significant reduction of the levels of the pro-apoptotic proteins cleaved-caspase 3 ( < 0.01) and Bax ( < 0.05), and enhanced the expression of the anti-apoptotic protein Bcl-2 ( < 0.05). The inhibition of autophagy obviously weakened the inhibitory effect of ATG3 overexpression on salinomycin-induced apoptosis.
CONCLUSIONS
ATG3 overexpression promotes autophagy possibly by inhibiting the AKT/mTOR signaling pathway to decrease salinomycin-induced apoptosis in MCF-7 cells, suggesting that autophagy induction might be one of the mechanisms of drug resistance in breast cancer cells.
Acetates
;
pharmacology
;
Apoptosis
;
drug effects
;
genetics
;
Autophagy
;
drug effects
;
Autophagy-Related Proteins
;
metabolism
;
Benzopyrans
;
pharmacology
;
Breast Neoplasms
;
metabolism
;
pathology
;
Cell Line, Tumor
;
Cell Proliferation
;
Drug Resistance, Neoplasm
;
Female
;
Gene Expression Regulation
;
Humans
;
MCF-7 Cells
;
Morpholines
;
pharmacology
;
Proto-Oncogene Proteins c-akt
;
antagonists & inhibitors
;
metabolism
;
Pyrans
;
pharmacology
;
TOR Serine-Threonine Kinases
;
antagonists & inhibitors
;
metabolism
;
Triazines
;
pharmacology
;
Ubiquitin-Conjugating Enzymes
;
metabolism
4.Long noncoding RNA LINC00520 prevents the progression of cutaneous squamous cell carcinoma through the inactivation of the PI3K/Akt signaling pathway by downregulating EGFR.
Chinese Medical Journal 2019;132(4):454-465
BACKGROUND:
Long noncoding RNAs (lncRNAs) play pivotal roles in various malignant tumors. Epidermal growth factor receptor (EGFR) signaling is associated with the pathogenesis of cutaneous squamous cell carcinoma (cSCC). This study aimed to explore the role of LINC00520 in the development of cSCC via EGFR and phosphoinositide 3-kinase-protein kinase B (PI3K/Akt) signaling pathways.
METHODS:
A microarray analysis was applied to screen differentially expressed lncRNAs in cSCC samples. The A431 cSCC cell line was transfected and assigned different groups. The expression patterns of LINC00520, EGFR, and intermediates in the PI3K/Akt pathway were characterized using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis. Cell proliferation, migration, and invasion were detected using the MTT assay, scratch test, and Transwell assay, respectively. Cell-based experiments and a tumorigenicity assay were conducted to assess the effect of LINC00520 on cSCC progression. This study was ended in September 2017. Comparisons between two groups were analyzed with t-test and comparisons among multiple groups were analyzed using one-way analysis of variance. The nonparametric Wilcoxon rank sum test was used to analyze skewed data. The enumerated data were analyzed using the chi-square test or Fisher exact test.
RESULTS:
Data from chip GSE66359 revealed depletion of LINC00520 in cSCC. Cells transfected with LINC00520 vector and LINC00520 vector + si-EGFR showed elevated LINC00520 level but decreased levels of the EGFR, PI3K, AKT, VEGF, MMP-2 and MMP-9 mRNAs and proteins, and inhibition of the growth, migration and adhesion of cSCC cells, while the si-LINC00520 group showed opposite trends (all P < 0.05). Compared with the LINC00520 vector group, the LINC00520 vector + si-EGFR group showed decreased levels of the EGFR, PI3K, AKT, VEGF, MMP-2 and MMP-9 mRNAs and proteins, and inhibition of the growth, migration and adhesion of cSCC cells, while the LINC00520 vector + EGFR vector group showed opposite results (all P < 0.05).
CONCLUSION
Based on our results, LINC00520-targeted EGFR inhibition might result in the inactivation of the PI3K/Akt pathway, thus inhibiting cSCC development.
Animals
;
Carcinoma, Squamous Cell
;
pathology
;
prevention & control
;
Cell Line, Tumor
;
Cell Movement
;
Cell Proliferation
;
Disease Progression
;
ErbB Receptors
;
antagonists & inhibitors
;
Female
;
Humans
;
Lymphatic Metastasis
;
Mice
;
Neoplasm Invasiveness
;
Phosphatidylinositol 3-Kinases
;
physiology
;
Proto-Oncogene Proteins c-akt
;
physiology
;
RNA, Long Noncoding
;
physiology
;
Signal Transduction
;
physiology
;
Skin Neoplasms
;
pathology
;
prevention & control
5.Akt Inhibitor Perifosine Prevents Epileptogenesis in a Rat Model of Temporal Lobe Epilepsy.
Feng ZHU ; Jiejing KAI ; Linglin CHEN ; Meiling WU ; Jingyin DONG ; Qingmei WANG ; Ling-Hui ZENG
Neuroscience Bulletin 2018;34(2):283-290
Accumulating data have revealed that abnormal activity of the mTOR (mammalian target of rapamycin) pathway plays an important role in epileptogenesis triggered by various factors. We previously reported that pretreatment with perifosine, an inhibitor of Akt (also called protein kinase B), abolishes the rapamycin-induced paradoxical increase of S6 phosphorylation in a rat model induced by kainic acid (KA). Since Akt is an upstream target in the mTOR signaling pathway, we set out to determine whether perifosine has a preventive effect on epileptogenesis. Here, we explored the effect of perifosine on the model of temporal epilepsy induced by KA in rats and found that pretreatment with perifosine had no effect on the severity or duration of the KA-induced status epilepticus. However, perifosine almost completely inhibited the activation of p-Akt and p-S6 both acutely and chronically following the KA-induced status epilepticus. Perifosine pretreatment suppressed the KA-induced neuronal death and mossy fiber sprouting. The frequency of spontaneous seizures was markedly decreased in rats pretreated with perifosine. Accordingly, rats pretreated with perifosine showed mild impairment in cognitive functions. Collectively, this study provides novel evidence in a KA seizure model that perifosine may be a potential drug for use in anti-epileptogenic therapy.
Animals
;
Anticonvulsants
;
pharmacology
;
Brain
;
drug effects
;
pathology
;
Convulsants
;
toxicity
;
Disease Models, Animal
;
Epilepsy, Temporal Lobe
;
chemically induced
;
pathology
;
Kainic Acid
;
toxicity
;
Male
;
Neurons
;
drug effects
;
pathology
;
Phosphorylcholine
;
analogs & derivatives
;
pharmacology
;
Protein Kinase Inhibitors
;
pharmacology
;
Proto-Oncogene Proteins c-akt
;
antagonists & inhibitors
;
Rats
;
Rats, Sprague-Dawley
;
Status Epilepticus
;
chemically induced
;
pathology
6.Research progress on mechanism of Nix-mediated mitophagy.
Yanrong ZHENG ; Xiangnan ZHANG ; Zhong CHEN
Journal of Zhejiang University. Medical sciences 2017;46(1):92-96
Autophagy is fundamental to maintain cellular homeostasis. As one kind of the most well-studied selective autophagy, autophagy of mitochondria (mitophagy)is crucial for the clearance of damaged mitochondria. Mitophagy dysfunction has been proved to be closely associated with many human diseases. Nix is a key protein for mitophagy during the maturation of reticulocytes. However, the detailed molecular mechanisms underlying Nix-mediated mitophagy are not fully understood. This article summarizes three possible working models of Nix in mitophagy induction. Firstly, Nix can interplay with Parkin, another important protein for mitophagy, to initiate mitophagy. Secondly, Nix can serve as a receptor for autophagy machinery by interacting with Atg8 family through its LIR motif. Finally, as a BH3-only protein, Nix can compete with Beclin-1 to bind other members of Bcl-2 family resulting in increased free Beclin-1 in cytosol, which further promotes autophagy flux.
Autophagy
;
genetics
;
physiology
;
Autophagy-Related Protein 8 Family
;
physiology
;
Beclin-1
;
physiology
;
Membrane Proteins
;
physiology
;
Mitochondria
;
genetics
;
physiology
;
Mitochondrial Degradation
;
genetics
;
physiology
;
Protein Interaction Domains and Motifs
;
Proto-Oncogene Proteins
;
physiology
;
Proto-Oncogene Proteins c-bcl-2
;
antagonists & inhibitors
;
Tumor Suppressor Proteins
;
physiology
;
Ubiquitin-Protein Ligases
;
physiology
7.Pim-1 Kinase Regulating Dynamics Related Protein 1 Mediates Sevoflurane Postconditioning-induced Cardioprotection.
Jin-Dong LIU ; Hui-Juan CHEN ; Da-Liang WANG ; Hui WANG ; Qian DENG ;
Chinese Medical Journal 2017;130(3):309-317
BACKGROUNDIt is well documented that sevoflurane postconditioning (SP) has a significant myocardial protection effect. However, the mechanisms underlying SP are still unclear. In the present study, we investigated the hypothesis that the Pim-1 kinase played a key role in SP-induced cardioprotection by regulating dynamics-related protein 1 (Drp1).
METHODSA Langendorff model was used in this study. Seventy-two rats were randomly assigned into six groups as follows: CON group, ischemia reperfusion (I/R) group, SP group , SP+proto-oncogene serine/threonine-protein kinase 1 (Pim-1) inhibitor II group, SP+dimethylsufoxide group, and Pim-1 inhibitor II group (n = 12, each). Hemodynamic parameters and infarct size were measured to reflect the extent of myocardial I/R injury. The expressions of Pim-1, B-cell leukemia/lymphoma 2 (Bcl-2) and cytochrome C (Cyt C) in cytoplasm and mitochondria, the Drp1 in mitochondria, and the total Drp1 and p-Drp1ser637 were measured by Western blotting. In addition, transmission electron microscope was used to observe mitochondrial morphology. The experiment began in October 2014 and continued until July 2016.
RESULTSSP improved myocardial I/R injury-induced hemodynamic parametric changes, cardiac function, and preserved mitochondrial phenotype and decreased myocardial infarct size (24.49 ± 1.72% in Sev group compared with 41.98 ± 4.37% in I/R group; P< 0.05). However, Pim-1 inhibitor II significantly (P < 0.05) abolished the protective effect of SP. Western blotting analysis demonstrated that, compared with I/R group, the expression of Pim-1 and Bcl-2 in cytoplasm and mitochondria as well as the total p-Drp1ser637 in Sev group (P < 0.05) were upregulated. Meanwhile, SP inhibited Drp1 compartmentalization to the mitochondria followed by a reduction in the release of Cyt C. Pretreatment with Pim-1 inhibitor II significantly (P < 0.05) abolished SP-induced Pim-1/p-Drp1ser637 signaling activation.
CONCLUSIONSThese findings suggested that SP could attenuate myocardial ischemia-reperfusion injury by increasing the expression of the Pim-1 kinase. Upregulation of Pim-1 might phosphorylate Drp1 and prevent extensive mitochondrial fission through Drp1 cytosolic sequestration.
Animals ; Dynamins ; metabolism ; Hemodynamics ; drug effects ; Ischemic Postconditioning ; methods ; Male ; Methyl Ethers ; therapeutic use ; Mitochondria ; drug effects ; metabolism ; Myocardial Reperfusion Injury ; metabolism ; prevention & control ; Proto-Oncogene Proteins c-pim-1 ; antagonists & inhibitors ; metabolism ; Quinazolinones ; pharmacology ; Rats ; Rats, Sprague-Dawley
8.EGF Induced RET Inhibitor Resistance in CCDC6-RET Lung Cancer Cells.
Hyun CHANG ; Ji Hea SUNG ; Sung Ung MOON ; Han Soo KIM ; Jin Won KIM ; Jong Seok LEE
Yonsei Medical Journal 2017;58(1):9-18
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.
Adenocarcinoma/drug therapy/*genetics
;
Cell Line, Tumor
;
Cetuximab/pharmacology
;
Drug Resistance, Neoplasm/drug effects/*genetics
;
Epidermal Growth Factor/metabolism/*pharmacology
;
*Gene Rearrangement
;
Hepatocyte Growth Factor/*pharmacology
;
Humans
;
Indoles/pharmacology
;
Lung Neoplasms/drug therapy/*genetics
;
MAP Kinase Signaling System
;
*Mutation
;
Niacinamide/analogs & derivatives/pharmacology
;
Phenylurea Compounds/pharmacology
;
Piperidines/pharmacology
;
Protein Kinase Inhibitors/therapeutic use
;
Proto-Oncogene Proteins c-ret/*antagonists & inhibitors/genetics
;
Pyrroles/pharmacology
;
Quinazolines/pharmacology
;
RNA, Small Interfering/pharmacology
;
Receptor, Epidermal Growth Factor/genetics/metabolism
;
Signal Transduction/drug effects
;
fms-Like Tyrosine Kinase 3/metabolism
9.Novel natural product therapeutics targeting both inflammation and cancer.
Jiangjiang QIN ; Wei WANG ; Ruiwen ZHANG
Chinese Journal of Natural Medicines (English Ed.) 2017;15(6):401-416
Inflammation is recently recognized as one of the hallmarks of human cancer. Chronic inflammatory response plays a critical role in cancer development, progression, metastasis, and resistance to chemotherapy. Conversely, the oncogenic aberrations also generate an inflammatory microenvironment, enabling the development and progression of cancer. The molecular mechanisms of action that are responsible for inflammatory cancer and cancer-associated inflammation are not fully understood due to the complex crosstalk between oncogenic and pro-inflammatory genes. However, molecular mediators that regulate both inflammation and cancer, such as NF-κB and STAT have been considered as promising targets for preventing and treating these diseases. Recent works have further demonstrated an important role of oncogenes (e.g., NFAT1, MDM2) and tumor suppressor genes (e.g., p53) in cancer-related inflammation. Natural products that target these molecular mediators have shown anticancer and anti-inflammatory activities in preclinical and clinical studies. Sesquiterpenoids (STs), a class of novel plant-derived secondary metabolites have attracted great interest in recent years because of their diversity in chemical structures and pharmacological activities. At present, we and other investigators have found that dimeric sesquiterpenoids (DSTs) may exert enhanced activity and binding affinity to molecular targets due to the increased number of alkylating centers and improved conformational flexibility and lipophilicity. Here, we focus our discussion on the activities and mechanisms of action of STs and DSTs in treating inflammation and cancer as well as their structure-activity relationships.
Animals
;
Anti-Inflammatory Agents
;
pharmacology
;
Antineoplastic Agents, Phytogenic
;
pharmacology
;
Humans
;
Inflammation
;
drug therapy
;
etiology
;
NF-kappa B
;
antagonists & inhibitors
;
NFATC Transcription Factors
;
antagonists & inhibitors
;
Neoplasms
;
drug therapy
;
etiology
;
Proto-Oncogene Proteins c-mdm2
;
antagonists & inhibitors
;
physiology
;
Sesquiterpenes
;
chemistry
;
pharmacology
;
Structure-Activity Relationship
10.Activation of NF-κB and AP-1 Mediates Hyperproliferation by Inducing β-Catenin and c-Myc in Helicobacter pylori-Infected Gastric Epithelial Cells.
Eunyoung BYUN ; Bohye PARK ; Joo Weon LIM ; Hyeyoung KIM
Yonsei Medical Journal 2016;57(3):647-651
PURPOSE: In the gastric mucosa of Helicobacter pylori (H. pylori)-infected patients with gastritis or adenocarcinoma, proliferation of gastric epithelial cells is increased. Hyperproliferation is related to induction of oncogenes, such as β-catenin and c-myc. Even though transcription factors NF-κB and AP-1 are activated in H. pylori-infected cells, whether NF-κB or AP-1 regulates the expression of β-catenein or c-myc in H. pylori-infected cells has not been clarified. The present study was undertaken to investigate whether H. pylori-induced activation of NF-κB and AP-1 mediates the expression of oncogenes and hyperproliferation of gastric epithelial cells. MATERIALS AND METHODS: Gastric epithelial AGS cells were transiently transfected with mutant genes for IκBα (MAD3) and c-Jun (TAM67) or treated with a specific NF-κB inhibitor caffeic acid phenethyl ester (CAPE) or a selective AP-1 inhibitor SR-11302 to suppress activation of NF-κB or AP-1, respecively. As reference cells, the control vector pcDNA was transfected to the cells. Wild-type cells or transfected cells were cultured with or without H. pylori. RESULTS: H. pylori induced activation of NF-κB and AP-1, cell proliferation, and expression of oncogenes (β-catenein, c-myc) in AGS cells, which was inhibited by transfection of MAD3 and TAM67. Wild-type cells and the cells transfected with pcDNA showed similar activities of NF-κB and AP-1, proliferation, and oncogene expression regardless of treatment with H. pylori. Both CAPE and SR-11302 inhibited cell proliferation and expression of oncogenes in H. pylori-infected cells. CONCLUSION: H. pylori-induced activation of NF-κB and AP-1 regulates transcription of oncogenes and mediates hyperproliferation in gastric epithelial cells.
Blotting, Western
;
Caffeic Acids
;
Cell Line, Tumor
;
Cell Proliferation
;
DNA, Bacterial/analysis/genetics
;
DNA-Binding Proteins/*metabolism
;
Epithelial Cells/*metabolism
;
Gastric Mucosa/*metabolism/pathology
;
Gastritis/pathology
;
Gene Expression Regulation, Bacterial
;
Helicobacter Infections/metabolism/pathology/physiopathology
;
Helicobacter pylori/pathogenicity/physiology
;
Humans
;
NF-kappa B/antagonists & inhibitors/*biosynthesis/metabolism
;
Peptide Fragments
;
Phenylethyl Alcohol/analogs & derivatives
;
Proto-Oncogene Proteins c-jun
;
Repressor Proteins
;
Transcription Factor AP-1/*biosynthesis
;
Transcription Factors/*metabolism
;
beta Catenin/*metabolism

Result Analysis
Print
Save
E-mail