1
|
Abstract
Transcription factors (TFs) are proteins that control the transcription of genetic information from DNA to mRNA by binding to specific DNA sequences either on their own or with other proteins as a complex. TFs thus support or suppress the recruitment of the corresponding RNA polymerase. In general, TFs are classified by structure or function. The TF, Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), is expressed in all cell types and tissues. NF-κB signaling and crosstalk are involved in several steps of carcinogenesis including in sequences involving pathogenic stimulus, chronic inflammation, fibrosis, establishment of its remodeling to the precancerous niche (PCN) and transition of a normal cell to a cancer cell. Triggered by various inflammatory cytokines, NF-κB is activated along with other TFs with subsequent stimulation of cell proliferation and inhibition of apoptosis. The involvement of NF-κB in carcinogenesis provides an opportunity to develop anti-NF-κB therapies. The complexity of these interactions requires that we elucidate those aspects of NF-κB interactions that play a role in carcinogenesis, the sequence of events leading to cancer.
Collapse
|
2
|
Xiao Y, Wang J, Qin Y, Xuan Y, Jia Y, Hu W, Yu W, Dai M, Li Z, Yi C, Zhao S, Li M, Du S, Cheng W, Xiao X, Chen Y, Wu T, Meng S, Yuan Y, Liu Q, Huang W, Guo W, Wang S, Deng W. Ku80 cooperates with CBP to promote COX-2 expression and tumor growth. Oncotarget 2016; 6:8046-61. [PMID: 25797267 PMCID: PMC4480734 DOI: 10.18632/oncotarget.3508] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/04/2015] [Indexed: 01/06/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) plays an important role in lung cancer development and progression. Using streptavidin-agarose pulldown and proteomics assay, we identified and validated Ku80, a dimer of Ku participating in the repair of broken DNA double strands, as a new binding protein of the COX-2 gene promoter. Overexpression of Ku80 up-regulated COX-2 promoter activation and COX-2 expression in lung cancer cells. Silencing of Ku80 by siRNA down-regulated COX-2 expression and inhibited tumor cell growth in vitro and in a xenograft mouse model. Ku80 knockdown suppressed phosphorylation of ERK, resulting in an inactivation of the MAPK pathway. Moreover, CBP, a transcription co-activator, interacted with and acetylated Ku80 to co-regulate the activation of COX-2 promoter. Overexpression of CBP increased Ku80 acetylation, thereby promoting COX-2 expression and cell growth. Suppression of CBP by a CBP-specific inhibitor or siRNA inhibited COX-2 expression as well as tumor cell growth. Tissue microarray immunohistochemical analysis of lung adenocarcinomas revealed a strong positive correlation between levels of Ku80 and COX-2 and clinicopathologic variables. Overexpression of Ku80 was associated with poor prognosis in patients with lung cancers. We conclude that Ku80 promotes COX-2 expression and tumor growth and is a potential therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Yao Xiao
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jingshu Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yu Qin
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Xuan
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yunlu Jia
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wenxian Hu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wendan Yu
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Meng Dai
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhenglin Li
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Canhui Yi
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shilei Zhao
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Mei Li
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Sha Du
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wei Cheng
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yiming Chen
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Taihua Wu
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Songshu Meng
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuhui Yuan
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Quentin Liu
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Wei Guo
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wuguo Deng
- Institute of Cancer Stem Cell & First Affiliated Hospital, Dalian Medical University, Dalian, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| |
Collapse
|
3
|
HDAC and NF-κB mediated cytotoxicity induced by novel N-Chloro β-lactams and benzisoxazole derivatives. Chem Biol Interact 2016; 246:69-76. [PMID: 26776669 DOI: 10.1016/j.cbi.2016.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 12/17/2015] [Accepted: 01/12/2016] [Indexed: 11/22/2022]
Abstract
Novel N-chloro â-Lactam and benzisoxazole derivatives were successfully synthesized with excellent yields (92-96%) under simple and mild reaction conditions. The β-lactams as a class acquired importance since the discovery of penicillin which contains β-lactam unit as an essential structural feature of its molecule, this interest continued unabated because of the therapeutic importance of β-lactam antibiotics. In silico studies of the compounds with cancer drug target enzymes showed the inhibition of HDAC (Histone Deacetylase) and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) significantly. The compounds were then investigated for the inhibitory potential against the same enzymes in vitro. NF-κB inhibition was investigated by trans activation assay using HEK293/NF-κB-luc cells. Overall, the synthesized compounds induce the cancer cell toxicity by restraining the NF-κB transcription factor mediated by HDAC inhibition and thus the compounds act as dual inhibitors.
Collapse
|
4
|
Walther W, Kobelt D, Bauer L, Aumann J, Stein U. Chemosensitization by diverging modulation by short-term and long-term TNF-α action on ABCB1 expression and NF-κB signaling in colon cancer. Int J Oncol 2015; 47:2276-85. [PMID: 26463665 DOI: 10.3892/ijo.2015.3189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/19/2015] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) is a major cause for cancer chemotherapy failure. Among the numerous strategies to overcome persistent action of proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) permits downregulation of MDR-associated genes, including ATP-binding cassette, subfamily B 1 gene (ABCB1). A key regulator of ABCB1 expression is the transcription factor nuclear factor κ light chain enhancer (NF-κB)/p65. We analyzed diverging short- and long-term effects of TNF-α regarding modulation of NF-κB/p65 signaling and ABCB1 expression in colon cancer cells. Highly resistant ABCB1 overexpressing human HCT15 colorectal carcinoma cells were subjected to short- (30-120 min) or long-term (24-96 h) TNF-α treatment. TNF-α mediated modulation of ABCB1 expression was analyzed by real-time RT-PCR and western blot analysis. The TNF-mediated chemosensitization was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay. The involvement of TNF receptors and of NF-κB/p65 signaling was analyzed by western blot analysis, ABCB1 promoter analysis and electrophoretic mobility shift assay (EMSA). The study revealed, that long-term, but not short-term TNF-α treatment leads to TNF-receptor 1 (TNFR1) mediated downregulation of ABCB1 resulting in sensitization towards drug treatment. It dampens NF-κB/p65 activation and nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor α (IκBα) resynthesis, associated with reduced nuclear accumulation of NF-κB/p65 and reduced binding to its consensus sequence in the ABCB1 promoter. The study reveals the diverging effects of short- or long-term TNF-α action and provides novel insights on downregulation of ABCB1 expression by TNF-mediated repression of NF-κB signaling.
Collapse
Affiliation(s)
- Wolfgang Walther
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Dennis Kobelt
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Lisa Bauer
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Jutta Aumann
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| |
Collapse
|
5
|
Wang Z, Zhang L, Ni Z, Sun J, Gao H, Cheng Z, Xu J, Yin P. Resveratrol induces AMPK-dependent MDR1 inhibition in colorectal cancer HCT116/L-OHP cells by preventing activation of NF-κB signaling and suppressing cAMP-responsive element transcriptional activity. Tumour Biol 2015; 36:9499-510. [PMID: 26124005 DOI: 10.1007/s13277-015-3636-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
Resveratrol, a natural polyphenolic compound found in foods and beverages, has attracted increasing attention in recent years because of its potent chemopreventive and anti-tumor effects. In this study, the effects of resveratrol on the expression of P-glycoprotein/multi-drug resistance protein 1 (P-gp/MDR1), and the underlying molecular mechanisms, were investigated in oxaliplatin (L-OHP)-resistant colorectal cancer cells (HCT116/L-OHP). Resveratrol downregulated MDR1 protein and mRNA expression levels and reduced MDR1 promoter activity. It also enhanced the intracellular accumulation of rhodamine 123, suggesting that resveratrol can reverse multi-drug resistance by downregulating MDR1 expression and reducing drug efflux. Resveratrol treatment also reduced nuclear factor-κB (NF-κB) activity, reduced phosphorylation levels of IκBα, and reduced nuclear translocation of the NF-κB subunit p65. Moreover, downregulation of MDR1 expression and promoter activity was mediated by resveratrol-induced AMP-activated protein kinase (AMPK) phosphorylation. The inhibitory effects of resveratrol on MDR1 expression and cAMP-responsive element-binding protein (CREB) phosphorylation were reversed by AMPKα siRNA transfection. We found that the transcriptional activity of cAMP-responsive element (CRE) was inhibited by resveratrol. These results demonstrated that the inhibitory effects of resveratrol on MDR1 expression in HCT116/L-OHP cells were closely associated with the inhibition of NF-κB signaling and CREB activation in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Ziyuan Wang
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Long Zhang
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Zhenhua Ni
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Jian Sun
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Hong Gao
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Zhuoan Cheng
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China
| | - Jianhua Xu
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China. .,Department of Clinical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China.
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062, China.
| |
Collapse
|
6
|
Nabekura T, Hiroi T, Kawasaki T, Uwai Y. Effects of natural nuclear factor-kappa B inhibitors on anticancer drug efflux transporter human P-glycoprotein. Biomed Pharmacother 2015; 70:140-5. [DOI: 10.1016/j.biopha.2015.01.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/04/2015] [Indexed: 02/02/2023] Open
|
7
|
Silva R, Vilas-Boas V, Carmo H, Dinis-Oliveira RJ, Carvalho F, de Lourdes Bastos M, Remião F. Modulation of P-glycoprotein efflux pump: induction and activation as a therapeutic strategy. Pharmacol Ther 2014; 149:1-123. [PMID: 25435018 DOI: 10.1016/j.pharmthera.2014.11.013] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/03/2023]
Abstract
P-glycoprotein (P-gp) is an ATP-dependent efflux pump encoded by the MDR1 gene in humans, known to mediate multidrug resistance of neoplastic cells to cancer therapy. For several decades, P-gp inhibition has drawn many significant research efforts in an attempt to overcome this phenomenon. However, P-gp is also constitutively expressed in normal human epithelial tissues and, due to its broad substrate specificity, to its cellular polarized expression in many excretory and barrier tissues, and to its great efflux capacity, it can play a crucial role in limiting the absorption and distribution of harmful xenobiotics, by decreasing their intracellular accumulation. Such a defense mechanism can be of particular relevance at the intestinal level, by significantly reducing the intestinal absorption of the xenobiotic and, consequently, avoiding its access to the target organs. In this review, the current knowledge on this important efflux pump is summarized, and a new focus is brought on the therapeutic interest of inducing and/or activating P-gp for limiting the toxicity caused by its substrates. Several in vivo and in vitro studies validating the use of such a therapeutic strategy are discussed. An extensive literature search for reported P-gp inducers/activators and for the experimental models used in their characterization was conducted. Those studies demonstrate that effective antidotal pathways can be achieved by efficiently promoting the P-gp-mediated efflux of deleterious xenobiotics, resulting in a significant reduction in their intracellular levels and, consequently, in a significant reduction of their toxicity.
Collapse
Affiliation(s)
- Renata Silva
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Vânia Vilas-Boas
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Helena Carmo
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Ricardo Jorge Dinis-Oliveira
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; INFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, Advanced Institute of Health Sciences - North (ISCS-N), CESPU, CRL, Gandra, Portugal; Department of Legal Medicine and Forensic Sciences, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Félix Carvalho
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
8
|
Novel thalidomide analogs: Anti-angiogenic and apoptotic effects on Hep-G2 and MCF-7 cancer cell lines. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.biomag.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
9
|
Wang Z, Xia Q, Cui J, Diao Y, Li J. Reversion of P-glycoprotein-mediated multidrug resistance by diallyl trisulfide in a human osteosarcoma cell line. Oncol Rep 2014; 31:2720-6. [PMID: 24788927 DOI: 10.3892/or.2014.3154] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/11/2014] [Indexed: 11/06/2022] Open
Abstract
Diallyl trisulfide (DATS), the main sulfuric compound in garlic, has been shown to have antitumor effects. The present study aimed to ascertain whether DATS reverses the drug resistance of human osteosarcoma cells in vitro and to investigate its potential mechanisms. Human osteosarcoma U2-OS cells were treated with different concentrations of DATS. Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, while P-glycoprotein (P-gp) expression and the proportion of apoptotic cells were measured by flow cytometry. Morphological changes were observed under an optical microscope. Νuclear factor-κB (NF-κB) and inhibitor of NF-κB (IκB) activities were measured by PCR and western blot analysis. Results showed that the proliferation of U2-OS cells treated with different concentrations of DATS was significantly decreased in a concentration- and time-dependent manner. DATS increased the toxic effect of adriamycin on U2-OS cells. Moreover, P-gp expression was decreased and the apoptosis rate was increased in a concentration-dependent manner following treatment of DATS. Additionally, NF-κB activity was inhibited by DATS while expression of IκB was increased. Our data clearly suggest that DATS has significant anticancer effects on human osteosarcoma cells. The potential mechanisms include reducing the multidrug resistance and inducing apoptosis. NF-κB suppression may be involved in DATS-induced inhibition of cell proliferation.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Emergency Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing Xia
- Department of Urinary Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jia Cui
- Shouguang Centre for Disease Control and Prevention, Shouguang, Shandong 262700, P.R. China
| | - Yutao Diao
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Jianmin Li
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
10
|
Rahman M, Salajegheh A, Smith R, Lam AY. B-Raf mutation: A key player in molecular biology of cancer. Exp Mol Pathol 2013; 95:336-42. [DOI: 10.1016/j.yexmp.2013.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 12/21/2022]
|
11
|
Kovalchuk I, Walz P, Thomas J, Kovalchuk O. Genomic instability in liver cells caused by an LPS-induced bystander-like effect. PLoS One 2013; 8:e67342. [PMID: 23874414 PMCID: PMC3706549 DOI: 10.1371/journal.pone.0067342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Bacterial infection has been linked to carcinogenesis, however, there is lack of knowledge of molecular mechanisms that associate infection with the development of cancer. We analyzed possible effects of the consumption of heat-killed E. coli O157:H7 cells or its cellular components, DNA, RNA, protein or lipopolysaccharides (LPS) on gene expression in naïve liver cells. Four week old mice were provided water supplemented with whole heat-killed bacteria or bacterial components for a two week period. One group of animals was sacrificed immediately, whereas another group was allowed to consume uncontaminated tap water for an additional two weeks, and liver samples were collected, post mortem. Liver cells responded to exposure of whole heat-killed bacteria and LPS with alteration in γH2AX levels and levels of proteins involved in proliferation, DNA methylation (MeCP2, DNMT1, DNMT3A and 3B) or DNA repair (APE1 and KU70) as well as with changes in the expression of genes involved in stress response, cell cycle control and bile acid biosynthesis. Other bacterial components analysed in this study did not lead to any significant changes in the tested molecular parameters. This study suggests that lipopolysaccharides are a major component of Gram-negative bacteria that induce molecular changes within naïve cells of the host.
Collapse
Affiliation(s)
- Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada.
| | | | | | | |
Collapse
|
12
|
Xia Q, Wang ZY, Li HQ, Diao YT, Li XL, Cui J, Chen XL, Li H. Reversion of p-glycoprotein-mediated multidrug resistance in human leukemic cell line by diallyl trisulfide. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2012; 2012:719805. [PMID: 22919419 PMCID: PMC3418761 DOI: 10.1155/2012/719805] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/16/2012] [Indexed: 12/15/2022]
Abstract
Multidrug resistance (MDR) is the major obstacle in chemotherapy, which involves multiple signaling pathways. Diallyl trisulfide (DATS) is the main sulfuric compound in garlic. In the present study, we aimed to explore whether DATS could overcome P-glycoprotein-(P-gp-)mediated MDR in K562/A02 cells, and to investigate whether NF-κB suppression is involved in DATS-induced reversal of MDR. MTT assay revealed that cotreatment with DATS increased the response of K562/A02 cells to adriamycin (the resistance reversal fold was 3.79) without toxic side effects. DATS could enhance the intracellular concentration of adriamycin by inhibiting the function and expression of P-gp, as shown by flow cytometry, RT-PCR, and western blot. In addition, DATS resulted in more K562/A02 cell apoptosis, accompanied by increased expression of caspase-3. The expression of NF-κB/p65 (downregulation) was significantly linked to the drug-resistance mechanism of DATS, whereas the expression of IκBα was not affected by DATS. Our findings demonstrated that DATS can serve as a novel, nontoxic modulator of MDR, and can reverse the MDR of K562/A02 cells in vitro by increasing intracellular adriamycin concentration and inducing apoptosis. More importantly, we proved for the first time that the suppression of NF-κB possibly involves the molecular mechanism in the course of reversion by DATS.
Collapse
Affiliation(s)
- Qing Xia
- Department of Hematology, Qilu Hospital, Shandong University, Shandong, Jinan 250012, China
| | - Zhi-Yong Wang
- Department of Emergency, Qilu Hospital, Shandong University, Shandong, Jinan 250012, China
| | - Hui-Qing Li
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Shandong, Jinan 250062, China
| | - Yu-Tao Diao
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Shandong, Jinan 250062, China
| | - Xiao-Li Li
- Soochow University and Department of Hematology, Branch Guangci of First Affiliated Hospital, Jiangsu, Suzhou 215128, China
| | - Jia Cui
- Shouguang Centre for Disease Control and Prevention, Shouguang 262700, China
| | - Xue-Liang Chen
- Department of Hematology, Qilu Hospital, Shandong University, Shandong, Jinan 250012, China
| | - Hao Li
- Department of Hematology, Qilu Hospital, Shandong University, Shandong, Jinan 250012, China
| |
Collapse
|
13
|
Lee H, Jeon J, Ryu YS, Jeong JE, Shin S, Zhang T, Kang SW, Hong JH, Hur GM. Disruption of microtubules sensitizes the DNA damage-induced apoptosis through inhibiting nuclear factor κB (NF-κB) DNA-binding activity. J Korean Med Sci 2010; 25:1574-81. [PMID: 21060745 PMCID: PMC2966993 DOI: 10.3346/jkms.2010.25.11.1574] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 07/22/2010] [Indexed: 02/02/2023] Open
Abstract
The massive reorganization of microtubule network involves in transcriptional regulation of several genes by controlling transcriptional factor, nuclear factor-kappa B (NF-κB) activity. The exact molecular mechanism by which microtubule rearrangement leads to NF-κB activation largely remains to be identified. However microtubule disrupting agents may possibly act in synergy or antagonism against apoptotic cell death in response to conventional chemotherapy targeting DNA damage such as adriamycin or comptothecin in cancer cells. Interestingly pretreatment of microtubule disrupting agents (colchicine, vinblastine and nocodazole) was observed to lead to paradoxical suppression of DNA damage-induced NF-κB binding activity, even though these could enhance NF-κB signaling in the absence of other stimuli. Moreover this suppressed NF-κB binding activity subsequently resulted in synergic apoptotic response, as evident by the combination with Adr and low doses of microtubule disrupting agents was able to potentiate the cytotoxic action through caspase-dependent pathway. Taken together, these results suggested that inhibition of microtubule network chemosensitizes the cancer cells to die by apoptosis through suppressing NF-κB DNA binding activity. Therefore, our study provided a possible anti-cancer mechanism of microtubule disrupting agent to overcome resistance against to chemotherapy such as DNA damaging agent.
Collapse
Affiliation(s)
- Hyunji Lee
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Juhee Jeon
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Young Sue Ryu
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Jae Eun Jeong
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Sanghee Shin
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Tiejun Zhang
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Seong Wook Kang
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jang Hee Hong
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| | - Gang Min Hur
- Department of Pharmacology, Research Institute for Medical Science, Daejeon Regional Cancer Center, Daejeon, Korea
| |
Collapse
|
14
|
Cho SO, Lim JW, Jun JH, Kim KH, Kim H. Helicobacter pylori in a Korean isolate expressed proteins differentially in human gastric epithelial cells. Dig Dis Sci 2010; 55:1550-64. [PMID: 19672714 DOI: 10.1007/s10620-009-0908-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Accepted: 07/05/2009] [Indexed: 12/19/2022]
Abstract
PURPOSE The proteins expressed in gastric epithelial cells infected with Helicobacter pylori (H. pylori) may determine the clinical outcome such as chronic gastritis, peptic ulcer, and gastric carcinoma. The present study aims to determine the differentially expressed proteins in human gastric epithelial AGS cells that were infected with H. pylori in a Korean isolate, a cagA+, vacA s1b m2 iceA1 H. pylori by proteomic analysis. The differentially expressed proteins, whose expression levels were more or less than twofold in H. pylori-infected cells, were analyzed. RESULTS Ten proteins (chromatin assembly factor-1, proliferating cell nuclear antigen, 14-3-3 protein tau, eukaryotic translation initiation factor 6, heat-shock protein 90beta, dimethylarginine dimethylaminohydrolase-1, L-lactate dehydrogenase B chain, prohibitin, triosephosphate isomerase, protein disulfide isomerase) were up-regulated while eight proteins (heat-shock gp96 precursor, nucleophosmin, ornithine aminotransferase, Ku70, L-arginine-glycine amidinotransferase, Smad anchor for receptor activation, ADP-ribosylation factor, WD repeat-containing protein isoform 1) were down-regulated by H. pylori infection in AGS cells. These proteins are related to cell proliferation, cell adhesion, carcinogenesis, cell-defense mechanisms against oxidative stress, membrane trafficking, and energy metabolism. CONCLUSIONS Oxidative stress, cell proliferation, cell adhesion, and membrane trafficking may be involved in the pathogenesis of gastric diseases including cancer associated with H. pylori in a Korean isolate.
Collapse
Affiliation(s)
- Soon Ok Cho
- Department of Pharmacology, Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
15
|
Kang MJ, Jung SM, Kim MJ, Bae JH, Kim HB, Kim JY, Park SJ, Song HS, Kim DW, Kang CD, Kim SH. DNA-dependent protein kinase is involved in heat shock protein-mediated accumulation of hypoxia-inducible factor-1alpha in hypoxic preconditioned HepG2 cells. FEBS J 2009; 275:5969-81. [PMID: 19021771 DOI: 10.1111/j.1742-4658.2008.06725.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hypoxic preconditioning may afford protection against subsequent lethal hypoxia. As hypoxic tolerance induces changes in the expression of genes involved in DNA damage and repair response pathways, we investigated whether DNA-dependent protein kinase (DNA-PK), one of the DNA double-strand break repair proteins, could be involved in hypoxic preconditioning-induced protective signaling cascades. We showed that induction of hypoxia-inducible factor-1alpha expression during hypoxic preconditioning by repeated hypoxic exposure was associated with increased mRNA and protein levels of DNA-PK catalytic subunit (DNA-PKcs) and Ku70/Ku80, the DNA-PK components, in human hepatoma HepG2 cells, followed by upregulation of Hsp70/Hsp90 and Bcl-2 and concurrent downregulation of Bax. Additionally, loss of DNA-PKcs led to attenuated expression of Hsp70/Hsp90, accelerated hypoxia-inducible factor-1alpha degradation, and increased susceptibility to hypoxia-induced cell death. We also found that the mRNA and protein levels of heat shock factor-1 (HSF1) were progressively increased with DNA-PK activation during hypoxic preconditioning, and inhibition of HSF1 function by KNK437 resulted in a significant decrease in the level of protein kinase Akt as well as of DNA-PKcs, with downregulation of Hsp70/Hsp90 and HIF-1alpha. Our results suggest the possibility that DNA-PK-mediated signaling pathway is required for the increase in HIF-1alpha expression through activation of HSF1 and subsequent upregulation of heat shock proteins after hypoxic reconditioning.
Collapse
Affiliation(s)
- Moon Jung Kang
- Department of Biochemistry, Pusan National University School of Medicine, Busan, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
In vitro anticancer property of a novel thalidomide analogue through inhibition of NF-kappaB activation in HL-60 cells. Acta Pharmacol Sin 2009; 30:134-40. [PMID: 19098937 DOI: 10.1038/aps.2008.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
AIM To investigate the anticancer property and possible mechanism of action of a novel sugar-substituted thalidomide derivative (STA-35) on HL-60 cells in vitro. METHODS TNF-alpha-induced NF-kappaB activation was determined using a reporter gene assay. The MTT assay was used to measure cytotoxicity of the compound. The appearance of apoptotic Sub-G1 cells was detected by flow cytometry analysis. PARP cleavage and protein expression of NF-kappaB p65 and its inhibitor IkappaB were viewed by Western blotting. RESULTS TA-35 (1-20 micromol/L) suppressed TNF-alpha-induced NF-kappaB activation in transfected cells (HEK293/pNiFty-SEAP) in a dose- (1-20 micromol/L) and time-dependent (0-48 h) manner. It was also shown that STA-35 exerted a dose-dependent inhibitory effect on HL-60 cell proliferation with an IC(50) value of 9.05 micromol/L. In addition, STA-35 induced apoptosis in HL-60 cells, as indicated by the appearance of a Sub-G1 peak in the cell cycle distribution, as well as poly ADP-ribose polymerase (PARP) cleavage. Subsequently, both NF-kappaB p65 and its inhibitor IkappaB gradually accumulated in cytoplasmic extracts in a dose- and time-dependent manner, indicating the blockage of NF-kappaB translocation induced by TNF-alpha from the cytoplasm to the nucleus. CONCLUSION A novel sugar-substituted thalidomide derivative, STA-35, is potent toward HL-60 cells in vitro and induces apoptosis by the suppression of NF-kappaB activation.
Collapse
|
17
|
Scanlon TC, Gottlieb B, Durcan TM, Fon EA, Beitel LK, Trifiro MA. Isolation of human proteasomes and putative proteasome-interacting proteins using a novel affinity chromatography method. Exp Cell Res 2008; 315:176-89. [PMID: 19013454 DOI: 10.1016/j.yexcr.2008.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 10/24/2008] [Accepted: 10/24/2008] [Indexed: 02/02/2023]
Abstract
The proteasome is the primary subcellular organelle responsible for protein degradation. It is a dynamic assemblage of 34 core subunits and many differentially expressed, transiently interacting, modulatory proteins. This paper describes a novel affinity chromatography method for the purification of functional human holoproteasome complexes using mild conditions. Human proteasomes purified by this simple procedure maintained the ability to proteolytically process synthetic peptide substrates and degrade ubiquitinated parkin. Furthermore, the entire purification fraction was analyzed by mass spectrometry in order to identify proteasomal proteins and putative proteasome-interacting proteins. The mild purification conditions maintained transient physical interactions between holoproteasomes and a number of known modulatory proteins. In addition, several classes of putative interacting proteins co-purified with the proteasomes, including proteins with a role in the ubiquitin proteasome system for protein degradation or DNA repair. These results demonstrate the efficacy of using this affinity purification strategy for isolating functional human proteasomes and identifying proteins that may physically interact with human proteasomes.
Collapse
Affiliation(s)
- Thomas C Scanlon
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Montreal, Canada
| | | | | | | | | | | |
Collapse
|
18
|
Assef Y, Rubio F, Coló G, del Mónaco S, Costas MA, Kotsias BA. Imatinib resistance in multidrug-resistant K562 human leukemic cells. Leuk Res 2008; 33:710-6. [PMID: 18977528 DOI: 10.1016/j.leukres.2008.09.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2008] [Revised: 09/16/2008] [Accepted: 09/22/2008] [Indexed: 12/31/2022]
Abstract
The multidrug resistance phenotype (MDR) is one of the major causes of failure in cancer chemotherapy and it is associated with the over-expression of P-glycoprotein (P-gp or MDR1) in tumor cell membranes. A constitutive NF-kappaB activity has been observed in several haematological malignancies and this is associated with its anti-apoptotic role. In the present work, the relationship between NF-kappaB and MDR phenotype was evaluated in wild type K562 human leukemic cells (K562-WT) and in its vincristine-resistant counterpart, K562-Vinc cells. These data showed that K562-Vinc cells, which express an active P-gp, exhibited MDR phenotype. The resistant indexes (IC(50)(K562-Vinc)/IC(50)(K562-WT)) for structurally unrelated drugs like imatinib, doxorubicin and colchicine were 8.0+/-0.3, 2.8+/-0.4 and 44.8+/-8.8, respectively. The imatinib resistance was reversed by P-gp blockade suggesting the involvement of P-gp in imatinib transport. We observed that NF-kappaB was constitutively activated in both cell lines but in a lesser extent in K562-Vinc. The inhibition of NF-kappaB with BAY 11-7082 increased the cytotoxicity of imatinib in K562-Vinc cells but not in K562-WT. Further, the co-administration of imatinib and BAY 11-7082 sensitized multidrug-resistant K562 cells to cell death as detected by increased percentage of annexin V positive cells. The induced cell death in K562-Vinc cells was associated with activation of caspases 9 and 3. Finally, we provide data showing that BAY 11-7082 down-regulates the expression of P-gp suggesting that the activity of NF-kappaB could be functionally associated to this protein in K562 cells. Our results indicate that the vincristine-resistant K562 cells which developed MDR phenotype, exhibited resistance to imatinib associated with a functional P-gp over-expression. This resistance could be partially overcome by the inhibition of NF-kappaB pathway.
Collapse
Affiliation(s)
- Yanina Assef
- Laboratorio de Neurofisiología, Instituto de Investigaciones Médicas Alfredo Lanari, Universidad de Buenos Aires, Conicet, Argentina
| | | | | | | | | | | |
Collapse
|
19
|
Lim JW, Kim KH, Kim H. NF-kappaB p65 regulates nuclear translocation of Ku70 via degradation of heat shock cognate protein 70 in pancreatic acinar AR42J cells. Int J Biochem Cell Biol 2008; 40:2065-77. [PMID: 18378183 DOI: 10.1016/j.biocel.2008.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Revised: 01/17/2008] [Accepted: 02/11/2008] [Indexed: 12/26/2022]
Abstract
Ku proteins such as Ku70 and Ku80 play key roles in multiple nuclear processes. Nuclear translocation of Ku70 is independent of Ku80 translocation and mediated by nuclear localization signal (NLS) receptors including importin-alpha. In the present study using pancreatic acinar AR42J cells, heat shock cognate protein 70 (Hsc70) was identified as the protein associated with NLS of Ku70. Interaction of Ku70 with importin-alpha and nuclear translocation of Ku70 was suppressed by overexpression of Hsc70, but enhanced by downregulation of Hsc70. The results suggest that the formation of Ku70 complex with Hsc70 prevents NLS of Ku70 from access of importin-alpha and inhibits nuclear translocation of Ku70. Since NF-kappaB p65 activation induced the decrease of Hsc70 level, the interaction of Ku70 with importin-alpha and nuclear translocation of Ku70 increased upon the activation of NF-kappaB p65. NF-kappaB p65 induced cell proliferation through decrease of Hsc70 levels and increase of nuclear translocation of Ku70. In the cells treated with cerulein as a physiological stimulus to activate NF-kappaB p65, nuclear translocation of Ku70 increased through NF-kappaB p65-mediated decrease of Hsc70 level. The results suggest that the involvement of NF-kappaB p65 in nuclear translocation of Ku70 may be mediated by Hsc70 degradation, which may play a key role in cell proliferation of pancreatic acinar AR42J cells.
Collapse
Affiliation(s)
- Joo Weon Lim
- Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | | | | |
Collapse
|
20
|
Huang X, Li M, Xu B, Zhu X, Deng Z, Lin W. Proteasome and NF-kappaB inhibiting phaeophytins from the green alga Cladophora fascicularis. Molecules 2007; 12:582-92. [PMID: 17851413 PMCID: PMC6149400 DOI: 10.3390/12030582] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2007] [Revised: 03/18/2007] [Accepted: 03/21/2007] [Indexed: 11/17/2022] Open
Abstract
Chemical examination of the green alga Cladophora fascicularis resulted in the isolation and characterization of a new porphyrin derivative, porphyrinolactone (1), along with five known phaeophytins 2-6 and fourteen sterols and cycloartanes. The structure of 1 was determined on the basis of spectroscopic analyses and by comparison of its NMR data with those of known phaeophytins. Compounds 1-6 displayed moderate inhibition of tumor necrosis factor alpha (TNF-alpha) induced nuclear factor-kappaB (NF-kappaB) activation, while 2 and 4 displayed potential inhibitory activity toward proteasome chymotripsin-like activation. The primary structure-activity relationship was also discussed.
Collapse
Affiliation(s)
- Xinping Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, P.R. China, Tel: (+86) 10-82806188; Fax: (+86) 10-82802724
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, P.R. China
| | - Min Li
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, P.R. China, Tel: (+86) 10-82806188; Fax: (+86) 10-82802724
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, P.R. China, Tel: (+86) 10-82806188; Fax: (+86) 10-82802724
| | - Xiaobin Zhu
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, P.R. China
| | - Zhiwei Deng
- Analytic and Testing Center, Beijing Normal University, Beijing 100073, P.R. China
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, P.R. China, Tel: (+86) 10-82806188; Fax: (+86) 10-82802724
| |
Collapse
|
21
|
Ma YX, Fu HZ, Li M, Sun W, Xu B, Cui JR. An anticancer effect of a new saponin component from Gymnocladus chinensis Baillon through inactivation of nuclear factor-κB. Anticancer Drugs 2007; 18:41-6. [PMID: 17159501 DOI: 10.1097/01.cad.0000236317.04199.69] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gymnocladus chinensis Baillon is widely distributed in China, and its fruits have been used in the treatment of rheumatism, furunculosis, soreness and swelling in traditional Chinese medicine for a long time. Few biological components were, however, isolated. In this study, a new triterpenoid saponin (GC-1) was extracted from the fruit of Gymnocladus chinensis Baillon and its biological actions were investigated. The results showed that GC-1 inhibited growth of a panel of human cancer cell lines in vitro by 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide and sulforhodamine B assays. Furthermore, GC-1 was demonstrated to induce apoptosis in HL-60 cells in a dose-dependent manner. By using a reporter gene assay, nuclear factor-kappaB activity induced by tumor necrosis factor-alpha was decreased gradually by addition of increasing concentration of GC-1 (1-40 micromol/l). In parallel, the blockage of nuclear factor-kappaB translocation from cytoplasm to nucleus was determined by Western blotting. This is the first study investigating the link of antiproliferative action of the compound with the inhibition of nuclear factor-kappaB activation. The mechanism of the actions of GC-1 might be due to the interruption of nuclear factor-kappaB translocation in the signaling pathway, which contributes to the chemotherapy potential.
Collapse
Affiliation(s)
- Yan-Xia Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, PRC
| | | | | | | | | | | |
Collapse
|
22
|
Kim H. DNA repair Ku proteins in gastric cancer cells and pancreatic acinar cells. Amino Acids 2006; 34:195-202. [PMID: 17031478 DOI: 10.1007/s00726-006-0411-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Accepted: 08/29/2006] [Indexed: 12/18/2022]
Abstract
The DNA repair protein Ku acts as a heterodimer of Ku70 and Ku80 that binds to the DNA ends, nicks, or single-to-double-strand transition. It has a crucial role for DNA double-strand-break repair. Either Ku70 or Ku80 itself may have a unique function that is independent of the other Ku subunit. In this review, the role of Ku on cell proliferation and apoptosis will be discussed. Ku acts as a regulator of transcription by interacting with the recombination signal binding protein Jkappa and the NF-kappaB p50 homodimer to up-regulate p50 expression, which may regulate the proliferation of gastric cancer cells. Both Ku70 and Ku80 expressions are mediated by constitutively activated NF-kappaB and constitutively expressed cyclooxygenase-2 in gastric cancer cells, which may be related to gastric cell proliferation and carcinogenesis. In addition, nuclear loss of Ku may underlie the mechanism of apoptosis in pancreatic acinar cells after oxidative stress.
Collapse
Affiliation(s)
- H Kim
- Department of Food and Nutrition, Brain Korea 21 Project, College of Human Ecology and Biomolecule Secretion Research Center, Institute of Gastroenterology, College of Medicine, Yonsei University, Seoul, Korea.
| |
Collapse
|
23
|
Song JY, Lim JW, Kim H, Kim KH. Role of NF-κB and DNA Repair Protein Ku on Apoptosis in Pancreatic Acinar Cells. Ann N Y Acad Sci 2006; 1010:259-63. [PMID: 15033730 DOI: 10.1196/annals.1299.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Reactive oxygen species have been known to cause DNA damage and induce apoptosis. During DNA damage, DNA repair proteins Ku70 and Ku80 prevent cell death, but severe DNA damage beyond the repair capacity of the DNA repair proteins triggers necrosis or apoptosis. Recent reports have shown that NF-kappaB plays a critical role in protecting the cells from apoptosis. We investigated whether glucose oxidase acting on beta-D-glucose (G/GO), which continuously produces H(2)O(2), induces apoptosis, and whether NF-kappaB and Ku are involved in G/GO-induced apoptosis in pancreatic acinar AR42J cells. Electron microscopic observation showed that apoptotic cells with characteristic nuclear condensation and shrinkage as well as large vacuoles were detected after G/GO treatment. G/GO treatment induced apoptotic cell death, as determined by viable cell count and DNA fragmentation. G/GO-induced apoptosis was increased in the cells transfected with the Ku-dominant negative mutant (Ku D/N) and mutated IkappaBalpha gene (IkappaB mt) as compared to the wild-type cells (Wild) and the cells transfected with the control pcDNA3 vector (pcN-3). G/GO treatment caused nuclear loss of both Ku70 and Ku80 in Wild cells and pcN-3 cells. Even without G/GO treatment, nuclear loss of Ku proteins was observed in IkappaB mt cells. These results suggest that oxidative stress-induced reduction of nuclear Ku proteins may cause loss of defense against DNA damage and thus induce apoptosis in pancreatic acinar cells. The novel finding is that nuclear translocation of Ku proteins may be mediated by NF-kappaB.
Collapse
Affiliation(s)
- Ji Yeon Song
- Department of Pharmacology and Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | |
Collapse
|
24
|
Abdel-Latif MMM, Raouf AA, Sabra K, Kelleher D, Reynolds JV. Vitamin C enhances chemosensitization of esophageal cancer cells in vitro. J Chemother 2006; 17:539-49. [PMID: 16323444 DOI: 10.1179/joc.2005.17.5.539] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Chemotherapy is increasingly utilised in multimodal protocols to try and improve outcomes. Cisplatin and 5-fluorouracil (5-Fu) are the mainstay of chemotherapeutic regimens, and an understanding of sensitivity and resistance of esophageal cancer to these agents is of considerable clinical importance. Antioxidants may modulate the response to chemotherapy, and in this study we examined the effect of vitamin C on 5-Fu and cisplatin cytotoxicity and related pathways in the esophageal cancer cell lines OE33 and SKGT-4. The antiproliferative effect of antitumor agents was measured by the MTT assay, and the transcription factors NF-kappaB and AP-1 pathways were assessed by electrophoretic mobility gel shift assay. 5-Fu and cisplatin demonstrated marked morphological changes and decreased cell proliferation. A combination of vitamin C with 5-Fu or cisplatin exerted a significantly enhanced cytotoxic effect compared to both drugs individually. Treatment of esophageal cancer cells with 5-Fu and cisplatin induced NF-kappaB and AP-1 activation. Pretreatment with vitamin C inhibited 5-Fu or cisplatin induced NF-kappaB nuclear translocation and DNA binding activity, but vitamin C had no effect on IkappaB-alpha protein levels. Vitamin C also inhibited 5-Fu- and cisplatin-induced AP-1 activation. Our data demonstrate that vitamin C enhances the antitumor activity of 5-Fu and cisplatin, in part by inhibiting translocation of NF-kappaB and AP-1, and sensitizes cancer cells to drug-induced cell death. The data suggest that vitamin C supplementation may improve the efficacy of chemotherapy for esophageal cancer.
Collapse
Affiliation(s)
- M M M Abdel-Latif
- Department of Surgery, Trinity Centre for Health Sciences, St James's Hospital, Dublin, Ireland.
| | | | | | | | | |
Collapse
|
25
|
Abstract
Nuclear Factor-kappa B (NF-kappa B) is an inducible transcription factor that regulates the expression of many genes involved in the immune response. Recently, NF-kappa B activity has been shown to be upregulated in many cancers, including melanoma. Data indicate that the enhanced activation of NF-kappa B may be due to deregulations in upstream signaling pathways such as Ras/Raf, PI3K/Akt, and NIK. Multiple studies have shown that NF-kappa B is involved in the regulation of apoptosis, angiogenesis, and tumor cell invasion, all of which indicate the important role of NF-kappa B in tumorigenesis. Thus, understanding the molecular mechanism of melanoma progression will aid in designing new therapeutic approaches for melanoma. In this review, the association between NF-kappa B and melanoma tumorigenesis are discussed. Additionally, the potential of emerging selective NF-kappa B inhibitors for the treatment of melanoma is reviewed.
Collapse
|
26
|
Whang PG, Gamradt SC, Gates JJ, Lieberman JR. Effects of the proteasome inhibitor bortezomib on osteolytic human prostate cancer cell metastases. Prostate Cancer Prostatic Dis 2005; 8:327-34. [PMID: 16130017 DOI: 10.1038/sj.pcan.4500823] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prostate adenocarcinoma is the most common malignancy diagnosed in males, and bone metastases remain a significant source of morbidity and mortality in this population. The ubiquitin-proteasome cascade is responsible for the degradation of intracellular proteins, and this pathway is thought to play an essential role in the development of malignancies by altering the levels of various proteins involved in the regulation of cell division. Proteasome inhibitors represent a class of chemotherapeutic agents that have been shown to inhibit tumor growth by a number of different mechanisms. Using a murine intratibial injection model, we examined the effects of the proteasome inhibitor bortezomib on the establishment and progression of osteolytic bone lesions induced by human CaP cells (PC-3 cell line). In this study, the intravenous administration of bortezomib (1 mg/kg) did not prevent the initial formation of osteolytic lesions but did appear to inhibit their growth in a time-dependent fashion. In contrast, bortezomib therapy effectively inhibited the establishment and progression of subcutaneous PC-3 tumors, which served as a positive control. These results suggest that proteasome inhibitors such as bortezomib may represent a novel adjunctive therapy for the treatment of osteolytic skeletal metastases, especially when treatment is initiated early during the disease process.
Collapse
Affiliation(s)
- P G Whang
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
27
|
Sharma V, Lansdell TA, Peddibhotla S, Tepe JJ. Sensitization of tumor cells toward chemotherapy: enhancing the efficacy of camptothecin with imidazolines. ACTA ACUST UNITED AC 2005; 11:1689-99. [PMID: 15610853 DOI: 10.1016/j.chembiol.2004.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2003] [Revised: 09/16/2004] [Accepted: 10/04/2004] [Indexed: 11/21/2022]
Abstract
Activation of nuclear transcription factor kappaB (NF-kappaB) by chemotherapeutic agents was found to protect cells from apoptosis. In light of its central role in regulating the cellular resistance to apoptotic agents, inhibition of NF-kappaB-mediated gene transcription may sensitize tumor cells to chemotherapeutic agents and enhance their efficacy. We describe herein a noncytotoxic imidazoline scaffold that sensitizes leukemia T cells to the chemotherapeutic agent camptothecin. No significant induction of apoptosis was found when cells were treated with the imidazoline; however, pretreatment of cells with this agent resulted in a drastic enhancement in efficacy of camptothecin (approximately 75-fold). Elucidation of the potential cellular mechanism revealed that the imidazoline prevents nuclear translocation of NF-kappaB. These findings indicate that inhibition of NF-kappaB by this imidazoline may present improved strategies in the chemotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- Vasudha Sharma
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | |
Collapse
|
28
|
Chen TY, Chen JS, Su WC, Wu MS, Tsao CJ. Expression of DNA repair gene Ku80 in lymphoid neoplasm. Eur J Haematol 2005; 74:481-8. [PMID: 15876251 DOI: 10.1111/j.1600-0609.2005.00428.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Ku, a heterodimer of KU70 and Ku80 that binds to double-strand DNA breaks (DSBs) and activates the catalytic subunit (DNA-PKcs) when DNA is bound, is essential in DSB repair and V(D)J recombination. Ku80 is a putative tumor suppressor gene that might play an important role in drug resistance. Our aim was to determine the role of Ku80 in lymphoid malignancy. PATIENTS AND METHODS Competitive reverse transcription-polymerase chain reaction assays were performed and the expression levels of Ku80 were measured in normal peripheral blood mononuclear cells (n = 9) and malignant cells from 25 patients with acute lymphoblastic leukemia (ALL) (14 children, 11 adults), and chronic lymphoproliferative disorders (n = 6). The Ku80 transcripts were sequencing for the possibility of mutation. RESULTS No mutation or Ku80 variant at the RNA level was seen in any patient samples or in the Raji or CCRF-CEM cell lines. In Ku80 expression, 8.8-, 1.9-, and 6.2-fold mean increases were seen in adult, pediatric ALL, and chronic lymphoid malignancies compared with the control. The Ku80 was significantly higher in adult than in pediatric ALL (P = 0.02). The amount of Ku80 expression in ALL was moderately correlated with peripheral white blood cell counts, but not with Ki67 labeling index. High Ku80 expressers (higher than the mean of all patients with ALL) tended to respond poorly to therapy: Only 22% of high Ku80 expressers achieved durable complete remission compared to 62% of low expressers. CONCLUSIONS Our study suggests that Ku80 might contribute to generally poor prognoses in adult ALL.
Collapse
MESH Headings
- Adolescent
- Adult
- Antigens, Nuclear/biosynthesis
- Antigens, Nuclear/genetics
- Cell Line, Tumor
- Child
- Child, Preschool
- DNA Repair/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Gene Expression Regulation, Leukemic
- Humans
- Infant
- Ki-67 Antigen/biosynthesis
- Ki-67 Antigen/genetics
- Ku Autoantigen
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukocytes, Mononuclear/metabolism
- Male
- Middle Aged
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Prognosis
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
Collapse
Affiliation(s)
- Tsai-Yun Chen
- Section of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.
| | | | | | | | | |
Collapse
|
29
|
|
30
|
Ryu JS, Um JH, Kang CD, Bae JH, Kim DU, Lee YJ, Kim DW, Chung BS, Kim SH. Fractionated irradiation leads to restoration of drug sensitivity in MDR cells that correlates with down-regulation of P-gp and DNA-dependent protein kinase activity. Radiat Res 2005; 162:527-35. [PMID: 15624307 DOI: 10.1667/rr3260] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We showed that the drug sensitivity of multidrug-resistant (MDR) cells could be enhanced by fractionated irradiation. The molecular changes associated with fractionated radiation-induced chemosensitization were characterized. Irradiated cells of the multidrug-resistant CEM/MDR sublines (CEM/MDR/IR1, 2 and 3) showed a loss of P-glycoprotein (P-gp) and concurrent reduction of Ku DNA binding and DNA-PK activities with decreased level of Ku70/80 and increased level of DNA-PKcs, and these changes were followed by an increased susceptibility to anticancer drugs. These irradiated MDR cells also exhibited the reduction of other chemoresistance-related proteins, including BCL2, NF-kappaB, EGFR, MDM2 and Ku70/80, and the suppression of HIF-1alpha expression induced by hypoxia. In contrast, fractionated irradiation increased the levels of these proteins and induced drug resistance in the parental drug-sensitive CEM cells. These results suggest that the chemoresistance-related proteins are differentially modulated in drug-sensitive and MDR cells by fractionated irradiation, and the optimized treatment with fractionated radiation could lead to new chemoradiotherapeutic strategies to treat multidrug-resistant tumors.
Collapse
Affiliation(s)
- Jun Sun Ryu
- Department of Biochemistry, College of Medicine, Pusan National University, Pusan 602-739, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Um JH, Kwon JK, Kang CD, Kim MJ, Ju DS, Bae JH, Kim DW, Chung BS, Kim SH. Relationship between Antiapoptotic Molecules and Metastatic Potency and the Involvement of DNA-Dependent Protein Kinase in the Chemosensitization of Metastatic Human Cancer Cells by Epidermal Growth Factor Receptor Blockade. J Pharmacol Exp Ther 2004; 311:1062-70. [PMID: 15273254 DOI: 10.1124/jpet.104.070938] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The failure to treat metastatic cancer with multidrug resistance is a major problem for successful cancer therapy, and the molecular basis for the association of metastatic phenotype with resistance to therapy is still unclear. In this study, we revealed that various metastatic cancer cells showed consistently higher levels of antiapoptotic proteins, including Bcl-2, nuclear factor-kappaB, MDM2, DNA-dependent protein kinase (DNA-PK), and epidermal growth factor receptor (EGFR), and lower levels of proapoptotic proteins, including Bax and p53 than low metastatic parental cells. This was followed by chemo- and radioresistance in metastatic cancer cells compared with their parental cells. EGFR and DNA-PK activity, which are known to be associated with chemo- and radioresistance, were demonstrated to be mutually regulated by each other. Treatment with PKI166, an EGFR inhibitor, suppressed etoposide-induced activation of DNA-PK in A375SM metastatic melanoma cells. In addition, PKI166 enhanced markedly the chemosensitivities of metastatic cancer cell sublines to various anticancer drugs in comparison with those of low metastatic cancer cells. These results suggest that the activities of DNA-PK and EGFR, which is positively correlated with each other, may contribute to metastatic phenotype as well as therapy resistance, and the EGFR inhibitor enhances the effect of anticancer drugs against therapy-resistant metastatic cancer cells via suppression of stress responses, including activation of DNA-PK.
Collapse
Affiliation(s)
- Jee Hyun Um
- Department of Biochemistry, College of Medicine, Pusan National University, Pusan 602-739, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dhar SK, Lynn BC, Daosukho C, St Clair DK. Identification of nucleophosmin as an NF-kappaB co-activator for the induction of the human SOD2 gene. J Biol Chem 2004; 279:28209-19. [PMID: 15087454 PMCID: PMC2651640 DOI: 10.1074/jbc.m403553200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Manganese superoxide dismutase (MnSOD) is an antioxidant enzyme essential for the survival of life. We have reported that NF-kappaB is essential but not sufficient for the synergistic induction of MnSOD by phorbol 12-myristate 13-acetate and cytokines. To further identify transcription factors and co-activators that participate in the induction of MnSOD, we used NF-kappaB affinity chromatography to isolate potential NF-kappaB interacting proteins. Proteins eluted from the NF-kappaB affinity column were subjected to proteomic analysis and verified by Western analysis. Nucleophosmin (NPM), a nucleolar phosphoprotein, is the most abundant single protein identified. Co-immunoprecipitation studies suggest a physical interaction between NPM and NF-kappaB proteins. To verify the role of NPM on MnSOD gene transcription, cells were transfected with constructs expressing NPM in sense or antisense orientation as well as interference RNA. The results indicate that an increase NPM expression leads to increased MnSOD gene transcription in a dose-dependent manner. Consistent with this, expression of small interfering RNA for NPM leads to inhibition of MnSOD gene transcription but does not have any effect on the expression of interleukin-8, suggesting that the effect of NPM is selective. These results identify NPM as a partner of the NF-kappaB transcription complex in the induction of MnSOD by phorbol 12-myristate 13-acetate and cytokines.
Collapse
MESH Headings
- Blotting, Northern
- Blotting, Western
- Cell Line
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Chromatography, Affinity
- Cytokines/metabolism
- DNA, Complementary/metabolism
- Dose-Response Relationship, Drug
- Exons
- Humans
- Interleukin-8/biosynthesis
- Introns
- Luciferases/metabolism
- Mass Spectrometry
- Models, Genetic
- Mutagenesis, Site-Directed
- NF-kappa B/metabolism
- Nuclear Proteins/metabolism
- Nuclear Proteins/physiology
- Nucleophosmin
- Oligonucleotides, Antisense/pharmacology
- Peptides/chemistry
- Plasmids/metabolism
- Precipitin Tests
- Protein Binding
- RNA/chemistry
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Silver Staining
- Superoxide Dismutase/biosynthesis
- Tetradecanoylphorbol Acetate/metabolism
- Time Factors
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Sanjit K Dhar
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
33
|
Wang W, Luo HS. Involvement of NF-kappa B activation in P-glycoprotein expression in vincristine-resistant human gastric cancer cells. Shijie Huaren Xiaohua Zazhi 2004; 12:537-541. [DOI: 10.11569/wcjd.v12.i3.537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of vincristine on activation of nuclear factor-kappa B (NF-κB) and expression of p-glycoprotein (P-gp) in vincristine-resistant human gastric cancer SGC7901 cells (SGC7901/VCR) and the parent sensitive clone (SGC7901).
METHODS: NF-κB-DNA binding activity was determined by electrophoretic mobility shift assay (EMSA). Levels of inhibitory B (IB-) and P-gp were measured by cellular-ELISA. And immunocytochemistry was used to detect the translocation of P65.
RESULTS: IN comparison with the parent SGC7901 cells, the basal and vincristine-induced NF-κB-DNA binding activity at various concentrations were all higher in the SGC7901/VCR cells. Concurrent with the NF-κB activation, vincristine-induced IB- degradation and P-gp upregulation were also found to be highly enhanced in the SGC7901/VCR cells than that in the sensitive SGC7901 cells. In the SGC7901/VCR cells, the activation of NF-κB induced by vincristine showed a positive relation with the level of P-gp expression (r = 0.977, P < 0.01). And nuclear translocation of P65 occurred concomitantly with NF-κB activation. Furthermore, the inhibition of NF-κB by MG-132, a proteasome inhibitor, could reduce NF-κB activation, IB- degradation and P-gp upregulation.
CONCLUSION: The enhancement of NF-κB activation correlates with vincristine resistance and NF-κB may play an important role in the regulation of the P-gp expression.
Collapse
|
34
|
Lim JW, Kim H, Kim KH. The Ku antigen-recombination signal-binding protein Jkappa complex binds to the nuclear factor-kappaB p50 promoter and acts as a positive regulator of p50 expression in human gastric cancer cells. J Biol Chem 2003; 279:231-7. [PMID: 14570916 DOI: 10.1074/jbc.m308609200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The p50 subunit of NF-kappaB is a transcription factor that regulates the expression of a variety of genes. Previously, we showed that the expression of Ku antigen, a DNA repair protein, is mediated by NF-kappaB in gastric cancer AGS cells (Lim, J. W., Kim, H., and Kim, K. H. (2002) J. Biol. Chem. 277, 46093-46100). In this study, we report that the inhibition of Ku activity reduced both p50 expression and nuclear NF-kappaB activity in AGS cells. A co-immunoprecipitation experiment demonstrated that Ku antigen interacted with recombination signal-binding protein Jkappa (RBP-Jkappa), a DNA-binding protein. Ku antigen, RBP-Jkappa, and p50 were found to bind to the DNA region containing the kappaB element in the p50 promoter. Supershift and competition experiments demonstrated that Ku antigen and RBP-Jkappa bound sequence-specifically to downstream elements of kappaB at GCTTC and TGGGGG. mRNA expression and de novo synthesis of p50 were inhibited in cells transfected with the mutant gene expression constructs for IkappaBalpha, Ku80, and RBP-Jkappa. A reporter assay demonstrated that p50 transcription was positively mediated by NF-kappaB, Ku antigen, and RBP-Jkappa and that the binding elements for these proteins were required for optimal p50 expression. The interaction of Ku antigen with RBP-Jkappa and NF-kappaB p50 may act as a positive regulator of p50 expression in gastric cancer AGS cells.
Collapse
Affiliation(s)
- Joo Weon Lim
- Department of Pharmacology and the Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | |
Collapse
|
35
|
Yeh PY, Chuang SE, Yeh KH, Song YC, Cheng AL. Involvement of nuclear transcription factor-kappa B in low-dose doxorubicin-induced drug resistance of cervical carcinoma cells. Biochem Pharmacol 2003; 66:25-33. [PMID: 12818362 DOI: 10.1016/s0006-2952(03)00250-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Administration of suboptimal doses of anticancer drugs not only fails to control tumor but often results in increased drug resistance of tumor cells. However, little is known about the effects of transient exposure to a minimally cytotoxic dose of chemotherapy on the development of drug resistance of the tumors. Previous studies have shown that upregulation of drug-exporter proteins (ATP-binding-cassette proteins) may be one of the key mechanisms involved in inducible drug resistance. In this study, we demonstrated that upregulation of NF-kappa B is another possible mechanism. SiHa cells were exposed to low-dose doxorubicin (100 nM; IC(30)) for 3 hr, and then were continuously cultured in drug-free culture media (designated as SiHa/DR cells). SiHa/DR cells at up to 9 passages showed increased resistance to doxorubicin and cross-resistance to cisplatin. Results of quantitative real-time PCR and flow cytometry assay indicated that the increased drug-resistance in SiHa/DR cells was not due to upregulation of drug-exporter proteins or to the decrease of intracellular concentration of anticancer drugs. Both the basal and drug-induced NF-kappa B activity were shown to be increased in SiHa/DR cells by EMSA and NF-kappa B-driven luciferase reporter gene assay. Suppression of NF-kappa B activation by transfection of a dominant negative I kappa B alpha prevented the development of drug resistance, indicating that the upregulated NF-kappa B activity was positively correlated with the low-dose doxorubicin-induced drug resistance. These results suggest that even a transient exposure to a small dose of anticancer drugs may induce drug resistance in some cancer cells via upregulation of NF-kappa B activity.
Collapse
Affiliation(s)
- Pei-Yen Yeh
- Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
36
|
Arlt A, Kruse ML, Breitenbroich M, Gehrz A, Koc B, Minkenberg J, Fölsch UR, Schäfer H. The early response gene IEX-1 attenuates NF-kappaB activation in 293 cells, a possible counter-regulatory process leading to enhanced cell death. Oncogene 2003; 22:3343-51. [PMID: 12761504 DOI: 10.1038/sj.onc.1206524] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The early response gene IEX-1 is involved in the regulation of cellular growth and survival, and its expression is related to stress-, growth- and death-inducing signals. Addressing the role of IEX-1 in the promotion of apoptosis, we investigated the effect of IEX-1 on nuclear factor-kappaB (NF-kappaB) activation. Stably transfected HEK-293 cells conditionally overexpressing IEX-1 exhibit decreased levels of NF-kappaB activity, either basal or TNFalpha induced, as shown by gel-shift and luciferase reporter gene assay. Furthermore, activated p65 accumulated in the nuclei of 293 cells to a lower degree, if IEX-1 expression was increased. This inhibited NF-kappaB activation was preceded by an altered turnover of IkappaBalpha and phospho-IkappaBalpha. In addition, IEX-1 expression also inhibited the activity of the 26S-proteasome, as shown by a fluorometric proteasome assay. Conversely, disruption of IEX-1 expression in 293 cells by stable transfection with specific anti-IEX-1 hammerhead ribozymes increased NF-kappaB activity, and accelerated the degradation of IkappaBalpha. Along with these opposite effects of IEX-1 expression and IEX-1 disruption on NF-kappaB activation, the sensitivity of 293 cells towards various apoptotic stimuli also changed. In contrast to ribozyme-transduced 293 cells that were significantly less sensitive to apoptosis, this sensitivity was enhanced if IEX-1 expression was increased. Our data suggest that IEX-1 - itself an NF-kappaB target gene - inhibits the activation of this transcription factor, and hereby may counteract the antiapoptotic potential of NF-kappaB.
Collapse
Affiliation(s)
- Alexander Arlt
- Laboratory of Molecular Gastroenterology & Hepatology, 1st Department of Medicine, Christian-Albrechts University of Kiel, Schittenhelmstr. 12, D-24105 Kiel, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Amit S, Ben-Neriah Y. NF-kappaB activation in cancer: a challenge for ubiquitination- and proteasome-based therapeutic approach. Semin Cancer Biol 2003; 13:15-28. [PMID: 12507553 DOI: 10.1016/s1044-579x(02)00096-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nuclear factor-kappa B (NF-kappaB) activation relies primarily on ubiquitin-mediated degradation of its inhibitor IkappaB. NF-kappaB plays an important role in many aspects of tumor development, progression, and therapy. Some types of cancer are characterized by constitutive NF-kappaB activity, whereas in others such activity is induced following chemotherapy. NF-kappaB-harboring tumors are generally resistant to chemotherapy and their eradication requires NF-kappaB inhibition. Here we describe the mechanisms of NF-kappaB activation in normal and tumor cells, review prevalent notions regarding the factor's contribution to tumorigenicity and discuss present and future options for NF-kappaB inhibition in cancer. The ubiquitination-mediated activation of NF-kappaB is intersected by another cancer-associated protein, beta-catenin. We, therefore, compare the related activation pathways and discuss the possibility of differential targeting of the involved ubiquitination machinery.
Collapse
Affiliation(s)
- Sharon Amit
- The Lautenberg Center for Immunology, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | | |
Collapse
|
38
|
Sonis ST. The biologic role for nuclear factor-kappaB in disease and its potential involvement in mucosal injury associated with anti-neoplastic therapy. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2003; 13:380-9. [PMID: 12393757 DOI: 10.1177/154411130201300502] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oral mucosal barrier injury (mucositis) is a frequent, painful, serious, dose-limiting toxicity associated with many anti-neoplastic drugs and radiation to the head and neck. Results of recent studies suggest that mucositis is the result of a complex series of interactive biological events that take place in the submucosa and epithelium. The nuclear transcription factor NF-kappaB has been implicated in the control of a broad range of biological responses, the activation of a large number of specific cellular genes, and the determination of the fate of cells exposed to ionizing radiation and anti-neoplastic drugs. Of particular importance to mucositis is the fact that NF-kappaB regulates key elements in the apparent sequence that leads to normal tissue toxicity. Not the least of these is the effect that NF-kappaB activation has on apoptosis. In particular, a paradox exists between the potential pro-apoptotic effect NF-kappaB exerts on normal cells, and the anti-apoptotic and cytoprotective effect it causes in tumor cells. This paper provides a review of the structure and function of NF-kappaB and speculates how its apparent enigmatic effect on normal and tumor cells may occur.
Collapse
Affiliation(s)
- Stephen T Sonis
- Department of Oral Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Yin F, Shi YQ, Chen CP, Qiao TD, Chen BJ, Miao JY, Fan DM. Expression of cDNA fragment encoding MGr1-Ag1 detected by MDR related antibody MGr1 in gastric cancer. Shijie Huaren Xiaohua Zazhi 2003; 11:18-21. [DOI: 10.11569/wcjd.v11.i1.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the distribution and significance of the possible cDNA fragments encoding MGr1-Ag1 in human gastric cancer.
METHODS: We investigated MGr1-Ag1 expression in gastric cancer using in situ hybridization(ISH) techniques in 42 cases of gastric carcinomas in cryostatic sections collected from Xijing Hospital.
RESULTS: MGr1-Ag1 mRNA was positive in cytoplasm of gastric glandulous epithelia with intensive staining in 50.00% (21/42) cases with gastric cancer, and 71.42% (30/42) cases in paracancerous gastric tissues. Eleven of 16 (68.75%) cases were well differentiated, 8/14 (57.14%) cases were moderately differentiated and 1/10 (10.00%) cases were poorly differentiated. The positive rate of MGr1-Ag1 mRNA in gastric cancer was significantly different from that in paracancerous gastric tissues (71.42%). There was no significant difference in positive rate of MGr1-Ag1 mRNA among normal, paracancerous gastric tissues, well and moderately differentiated tissues, but the positive rate in the poorly differentiated tissues was low, being significantly different from that in well or moderately differentiated tissues.
CONCLUSION: MGr1-Ag1 mRNA is mainly localized in epithelial cells, and the level of its expression is correlated with the tumor differentiation.
Collapse
|
40
|
Bentires-Alj M, Barbu V, Fillet M, Chariot A, Relic B, Jacobs N, Gielen J, Merville MP, Bours V. NF-kappaB transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene 2003; 22:90-7. [PMID: 12527911 DOI: 10.1038/sj.onc.1206056] [Citation(s) in RCA: 338] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ubiquitous NF-kappaB transcription factor has been reported to inhibit apoptosis and to induce drug resistance in cancer cells. Drug resistance is the major reason for cancer therapy failure and neoplastic cells often develop multiple mechanisms of drug resistance during tumor progression. We observed that NF-kappaB or P-glycoprotein inhibition in the HCT15 colon cancer cells led to increased apoptotic cell death in response to daunomycin treatment. Interestingly, NF-kappaB inhibition through transfection of a plasmid coding for a mutated IkappaB-alpha inhibitor increased daunomycin cell uptake. Indeed, the inhibition of NF-kappaB reduced mdr1 mRNA and P-glycoprotein expression in HCT15 cells. We identified a consensus NF-kappaB binding site in the first intron of the human mdr1 gene and demonstrated that NF-kappaB complexes could bind with this intronic site. Moreover, NF-kappaB transactivates an mdr1 promoter luciferase construct. Our data thus demonstrate a role for NF-kappaB in the regulation of the mdr1 gene expression in cancer cells and in drug resistance.
Collapse
|
41
|
Matheos D, Novac O, Price GB, Zannis-Hadjopoulos M. Analysis of the DNA replication competence of the xrs-5 mutant cells defective in Ku86. J Cell Sci 2003; 116:111-24. [PMID: 12456721 DOI: 10.1242/jcs.00156] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The radiosensitive mutant xrs-5, a derivative of the Chinese hamster ovary (CHO) K1 cell line, is defective in DNA double-strand break repair and V(D)J recombination. The defective phenotypes of xrs-5 cells are complemented by the 86 kDa subunit of Ku antigen. OBA is a protein, previously purified from HeLa cells, that binds in a sequence-specific manner to mammalian origins of DNA replication. The DNA-binding subunit of OBA has been identified as Ku86. We tested the xrs-5 cell line for its ability to replicate a mammalian origin-containing plasmid, p186, in vivo and in vitro. In vivo, the p186 episomal DNA replication in transfected xrs-5 cells was reduced by 45% when compared with the CHO K1 cells transfected with p186. In vitro, although total and cytoplasmic cell extracts from xrs-5 cells replicated the p186 with the same efficiency as the parental CHO K1 cell extracts, xrs-5 nuclear extracts did not possess any detectable replication activity. Addition of affinity-purified OBA/Ku restored replication in the xrs-5 nuclear extract reaction. Western blot analyses showed that the levels of other replication proteins (Orc2, PCNA, DNA polymerase epsilon and delta, Primase and Topoisomerase IIalpha) were comparable in both the xrs-5 mutant and CHO K1 wild-type cell lines. In addition, the in vivo association of Ku with the DHFR origin-containing sequence (oribeta) was examined in both the CHO K1 and xrs-5 cell lines by a chromatin immunoprecipitation (ChIP) assay. Anti-Ku antibodies did not immunoprecipitate a detectable amount of Ku from the xrs-5 cells in the origin-containing sequence, in contrast to the CHO K1 cells, wherein Ku was found to be associated with the oribeta origin. The data implicate Ku antigen in in vivo and in vitro DNA replication and suggest the existence of another protein with Ku-like functions in the xrs-5 cells.
Collapse
Affiliation(s)
- Diamanto Matheos
- McGill Cancer Centre, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | | | | | | |
Collapse
|
42
|
Lim JW, Kim H, Kim KH. Expression of Ku70 and Ku80 mediated by NF-kappa B and cyclooxygenase-2 is related to proliferation of human gastric cancer cells. J Biol Chem 2002; 277:46093-100. [PMID: 12324457 DOI: 10.1074/jbc.m206603200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) expression is mediated by constitutive NF-kappaB and regulates human gastric cancer cell growth and proliferation. Inactivating Ku70 or Ku80 suppresses cell growth and induces apoptosis. It has been hypothesized that Ku70 and Ku80 expression may be associated with NF-kappaB activation and COX-2 expression and is involved in cell proliferation. In this study, we found that inhibition of constitutive NF-kappaB (by transfecting a mutated IkappaBalpha gene) and of COX-2 (by treatment with indomethacin and NS-398) suppressed Ku70 and Ku80 expression in cells. Treatment with prostaglandin E(2) adenocarcinoma gastric (AGS) increased expression of these Ku proteins in cells with low constitutive NF-kappaB levels. Inhibition of the Ku DNA end-binding activity by transfection with the C-terminal Ku80 expression gene suppressed cell proliferation. Ku70 or Ku80 overexpression by transfection with the Ku70 or Ku80 expression gene, respectively, enhanced proliferation of cells with low NF-kappaB levels. These results demonstrate that Ku70 and Ku80 expression is mediated by constitutively activated NF-kappaB and constitutively expressed COX-2 in gastric cancer cells and that the high Ku DNA end-binding activity contributes to cell proliferation. Ku70 and Ku80 expression may be related to gastric cell proliferation and carcinogenesis.
Collapse
Affiliation(s)
- Joo Weon Lim
- Department of Pharmacology and the Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | |
Collapse
|
43
|
Abstract
The targeted degradation of key regulatory proteins is an essential element of cell cycle control. The proteasome plays a central role in the degradation of such proteins and has therefore become an important therapeutic target for diseases involving cell proliferation, notably cancer. This review summarizes numerous studies demonstrating that proteasome inhibition induces apoptosis and sensitizes cancer cells to traditional tumoricidal agents both and The potent and selective proteasome inhibitor, PS-341, is particularly promising from a therapeutic perspective, and it is the only such inhibitor that has progressed to clinical trials. Preliminary data indicate that the drug is well tolerated by patients with cancer, and further trials are underway to assess the safety and efficacy of proteasome inhibition in hematologic and solid tumors, both as a monotherapy and in combination with other chemotherapeutics.
Collapse
Affiliation(s)
- Julian Adams
- Millennium Pharmaceuticals, Cambridge, Massachusetts 02139, USA.
| |
Collapse
|