1
|
Kim R, Kin T, Beck WT. Impact of Complex Apoptotic Signaling Pathways on Cancer Cell Sensitivity to Therapy. Cancers (Basel) 2024; 16:984. [PMID: 38473345 DOI: 10.3390/cancers16050984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer drugs induce apoptotic and non-apoptotic cell death in various cancer types. The signaling pathways for anticancer drug-induced apoptotic cell death have been shown to differ between drug-sensitive and drug-resistant cells. In atypical multidrug-resistant leukemia cells, the c-Jun/activator protein 1 (AP-1)/p53 signaling pathway leading to apoptotic death is altered. Cancer cells treated with anticancer drugs undergo c-Jun/AP-1-mediated apoptotic death and are involved in c-Jun N-terminal kinase activation and growth arrest- and DNA damage-inducible gene 153 (Gadd153)/CCAAT/enhancer-binding protein homologous protein pathway induction, regardless of the p53 genotype. Gadd153 induction is associated with mitochondrial membrane permeabilization after anticancer drug treatment and involves a coupled endoplasmic reticulum stress response. The induction of apoptosis by anticancer drugs is mediated by the intrinsic pathway (cytochrome c, Cyt c) and subsequent activation of the caspase cascade via proapoptotic genes (e.g., Bax and Bcl-xS) and their interactions. Anticancer drug-induced apoptosis involves caspase-dependent and caspase-independent pathways and occurs via intrinsic and extrinsic pathways. The targeting of antiapoptotic genes such as Bcl-2 enhances anticancer drug efficacy. The modulation of apoptotic signaling by Bcl-xS transduction increases the sensitivity of multidrug resistance-related protein-overexpressing epidermoid carcinoma cells to anticancer drugs. The significance of autophagy in cancer therapy remains to be elucidated. In this review, we summarize current knowledge of cancer cell death-related signaling pathways and their alterations during anticancer drug treatment and discuss potential strategies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - William T Beck
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Kurimchak AM, Herrera-Montávez C, Montserrat-Sangrà S, Araiza-Olivera D, Hu J, Neumann-Domer R, Kuruvilla M, Bellacosa A, Testa JR, Jin J, Duncan JS. The drug efflux pump MDR1 promotes intrinsic and acquired resistance to PROTACs in cancer cells. Sci Signal 2022; 15:eabn2707. [PMID: 36041010 PMCID: PMC9552188 DOI: 10.1126/scisignal.abn2707] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Proteolysis-targeting chimeras (PROTACs) are a promising new class of drugs that selectively degrade cellular proteins of interest. PROTACs that target oncogene products are avidly being explored for cancer therapies, and several are currently in clinical trials. Drug resistance is a substantial challenge in clinical oncology, and resistance to PROTACs has been reported in several cancer cell models. Here, using proteomic analysis, we found intrinsic and acquired resistance mechanisms to PROTACs in cancer cell lines mediated by greater abundance or production of the drug efflux pump MDR1. PROTAC-resistant cells were resensitized to PROTACs by genetic ablation of ABCB1 (which encodes MDR1) or by coadministration of MDR1 inhibitors. In MDR1-overexpressing colorectal cancer cells, degraders targeting either the kinases MEK1/2 or the oncogenic mutant GTPase KRASG12C synergized with the dual epidermal growth factor receptor (EGFR/ErbB)/MDR1 inhibitor lapatinib. Moreover, compared with single-agent therapies, combining MEK1/2 degraders with lapatinib improved growth inhibition of MDR1-overexpressing KRAS-mutant colorectal cancer xenografts in mice. Together, our findings suggest that concurrent blockade of MDR1 will likely be required with PROTACs to achieve durable protein degradation and therapeutic response in cancer.
Collapse
Affiliation(s)
- Alison M. Kurimchak
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Carlos Herrera-Montávez
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sara Montserrat-Sangrà
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Daniela Araiza-Olivera
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jianping Hu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York 10029, USA
| | - Ryan Neumann-Domer
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Mathew Kuruvilla
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA,Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA 19111, USA,Biomedical Sciences Graduate Program, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Alfonso Bellacosa
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA,Biomedical Sciences Graduate Program, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Joseph R. Testa
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York 10029, USA
| | - James S. Duncan
- Cancer Signaling & Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA,Correspondence:
| |
Collapse
|
3
|
Phytol and Heptacosane Are Possible Tools to Overcome Multidrug Resistance in an In Vitro Model of Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2022; 15:ph15030356. [PMID: 35337153 PMCID: PMC8952646 DOI: 10.3390/ph15030356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 12/19/2022] Open
Abstract
Drug resistance is the ability of cancer cells to gain resistance to both conventional and novel chemotherapy agents, and remains a major problem in cancer therapy. Resistance mechanisms are multifactorial and involve more strictly pharmacological factors, such as P-glycoprotein (P-gp) and biological factors such as inhibitor of apoptosis proteins (IAPs) and the nuclear factor-kappa B (NF-κB) pathway. Possible therapeutic strategies for the treatment of acute myeloid leukemia (AML) have increased in recent years; however, drug resistance remains a problem for most pa-tients. Phytol and heptacosane are the major compounds of Euphorbia intisy essential oil (EO) which were demonstrated to inhibit P-gp in a multidrug resistant in vitro model of AML. This study investigated the mechanism by which phytol and heptacosane improve P-gp-mediated drug transport. Phytol suppresses the P-gp expression via NF-κB inhibition and does not seem to act on the efflux system. Heptacosane acts as a substrate and potent P-gp inhibitor, demonstrating the ability to retain the substrate doxorubicin inside the cell and enhancing its cytotoxic effects. Our results suggest that these compounds act as non-toxic modulators of P-gp through different mechanisms and are able to revert P-gp-mediated drug resistance in tumor cells.
Collapse
|
4
|
Gong C, Yang H, Wang S, Liu J, Li Z, Hu Y, Chen Y, Huang Y, Luo Q, Wu Y, Liu E, Xiao Y. hTERT Promotes CRC Proliferation and Migration by Recruiting YBX1 to Increase NRF2 Expression. Front Cell Dev Biol 2021; 9:658101. [PMID: 34079797 PMCID: PMC8165255 DOI: 10.3389/fcell.2021.658101] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
High human telomerase reverse transcriptase (hTERT) expression is related to severe Colorectal Cancer (CRC) progression and negatively related to CRC patient survival. Previous studies have revealed that hTERT can reduce cancer cellular reactive oxygen species (ROS) levels and accelerate cancer progression; however, the mechanism remains poorly understood. NFE2-related factor 2 (NRF2) is a molecule that plays a significant role in regulating cellular ROS homeostasis, but whether there is a correlation between hTERT and NRF2 remains unclear. Here, we showed that hTERT increases CRC proliferation and migration by inducing NRF2 upregulation. We further found that hTERT increases NRF2 expression at both the mRNA and protein levels. Our data also revealed that hTERT primarily upregulates NRF2 by increasing NRF2 promoter activity rather than by regulating NRF2 mRNA or protein stability. Using DNA pull-down/MS analysis, we found that hTERT can recruit YBX1 to upregulate NRF2 promoter activity. We also found that hTERT/YBX1 may localize to the P2 region of the NRF2 promoter. Taken together, our results demonstrate that hTERT facilitates CRC proliferation and migration by upregulating NRF2 expression through the recruitment of the transcription factor YBX1 to activate the NRF2 promoter. These results provide a new theoretical basis for CRC treatment.
Collapse
Affiliation(s)
- Chunli Gong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Huan Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Sumin Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jiao Liu
- Department of Endoscope, General Hospital of Shenyang Military Region, Shenyang, China
| | - Zhibin Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yiyang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yang Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yu Huang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiang Luo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuyun Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - En Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yufeng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
5
|
The "Janus" Role of C/EBPs Family Members in Cancer Progression. Int J Mol Sci 2020; 21:ijms21124308. [PMID: 32560326 PMCID: PMC7352866 DOI: 10.3390/ijms21124308] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
CCAAT/enhancer-binding proteins (C/EBPs) constitute a family of transcription factors composed of six members that are critical for normal cellular differentiation in a variety of tissues. They promote the expression of genes through interaction with their promoters. Moreover, they have a key role in regulating cellular proliferation through interaction with cell cycle proteins. C/EBPs are considered to be tumor suppressor factors due to their ability to arrest cell growth (contributing to the terminal differentiation of several cell types) and for their role in cellular response to DNA damage, nutrient deprivation, hypoxia, and genotoxic agents. However, C/EBPs can elicit completely opposite effects on cell proliferation and cancer development and they have been described as both tumor promoters and tumor suppressors. This "Janus" role of C/EBPs depends on different factors, such as the type of tumor, the isoform/s expressed in cells, the type of dimerization (homo- or heterodimerization), the presence of inhibitory elements, and the ability to inhibit the expression of other tumor suppressors. In this review, we discuss the implication of the C/EBPs family in cancer, focusing on the molecular aspects that make these transcription factors tumor promoters or tumor suppressors.
Collapse
|
6
|
Robinson K, Tiriveedhi V. Perplexing Role of P-Glycoprotein in Tumor Microenvironment. Front Oncol 2020; 10:265. [PMID: 32195185 PMCID: PMC7066112 DOI: 10.3389/fonc.2020.00265] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
Development of multidrug resistance (MDR) still remains a major obstacle to the long-term success of cancer therapy. P-glycoprotein (P-gp) is a well-identified membrane transporter with capability to efflux drug molecules out of the cancer cell leading to reduced efficiency of chemotherapy. Cancer cells upregulate P-gp expression as an adaptive response to evade chemotherapy mediated cell death. While several P-gp inhibitors have been discovered by in silico and pre-clinical studies, very few have successfully passed all phases of the clinical trials. Studies show that application of P-gp inhibitors in cancer therapy regimen following development of MDR achieved limited beneficial outcomes. While, the non-specific substrate binding to P-gp has made the drug-design a challenge, a bigger perplexing challenge comes from its role in tumor immunology. Expression of P-gp was noted immune cell phenotypes with apparently antagonistic functionality. Both pro-tumor MΦ2-macrophages and, anti-tumor NK-cell and Th17/CD4+T cell subsets have shown enhanced expression of P-gp. While drug based inhibition of P-gp in pro-tumor immune cell phenotypes could promote tumor elimination, however, it would not be a rational choice to exert inhibition of P-gp on anti-tumor immune cell phenotypes. This mutually exclusive paradigm of P-gp functionality requires a more comprehensive and detailed understanding of its role in tumor microenvironment with active interplay of cancer and immune cells in the tumor mileu. In this review, we focus on the current understanding of the role of P-gp in cancer cells and immune cells and finally attempt to highlight some caveats in the current understanding of its role in comprehensive tumor microenvironment along with challenges in the development of P-gp inhibitors toward anti-cancer therapy.
Collapse
Affiliation(s)
- Kianna Robinson
- Department of Biological Sciences, Tennessee State University, Nashville, TN, United States
| | - Venkataswarup Tiriveedhi
- Department of Biological Sciences, Tennessee State University, Nashville, TN, United States.,Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
7
|
Mirzaei SA, Dinmohammadi F, Alizadeh A, Elahian F. Inflammatory pathway interactions and cancer multidrug resistance regulation. Life Sci 2019; 235:116825. [PMID: 31494169 DOI: 10.1016/j.lfs.2019.116825] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Multidrug resistances against chemotherapeutics are among the major challenges related to cancer treatment. Recent studies have demonstrated that different conditions may tune the expression and activity of MDR transporters. For instance, inflammation occurs through a complex cytological process and chemical reactions in the most tumor microenvironment; it can play a critical role in cancer development and is capable of altering the expression and function of MDR transporters. Cytokines, interleukins, and prostaglandins are potent inflammatory mediators that can modulate the expression of MDRs at transcriptional and post-transcriptional levels in the most human cancer cells and tissues and potentially contribute to balance bioavailability of chemotherapeutic agents. Since cancer cases are usually accompanied by inflammatory responses, glucocorticoids and NSAIDs are the primary useful combination chemotherapies in a variety of cancer treatment protocols. In addition to the anti-inflammatory activities of these agents, they exert diverse modulatory effects on MDR-mediated drug resistance via specific mechanisms. Several factors, including cell and MDR-protein types, pharmacokinetics, and pharmacogenetics, mainly influence the regulatory mechanisms. Uncovering the networks between inflammation and multidrug resistance will be clinically helpful in the treatment of malignant cancers and decreasing the cancer mortality rates.
Collapse
Affiliation(s)
- Seyed Abbas Mirzaei
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farideh Dinmohammadi
- Department of Food and Drug Control, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
8
|
Salaroglio IC, Gazzano E, Abdullrahman A, Mungo E, Castella B, Abd-Elrahman GEFAE, Massaia M, Donadelli M, Rubinstein M, Riganti C, Kopecka J. Increasing intratumor C/EBP-β LIP and nitric oxide levels overcome resistance to doxorubicin in triple negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:286. [PMID: 30482226 PMCID: PMC6258159 DOI: 10.1186/s13046-018-0967-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Triple negative breast cancer (TNBC) easily develops resistance to the first-line drug doxorubicin, because of the high levels of the drug efflux transporter P-glycoprotein (Pgp) and the activation of pro-survival pathways dependent on endoplasmic reticulum (ER). Interfering with these mechanisms may overcome the resistance to doxorubicin, a still unmet need in TNBC. METHODS We analyzed a panel of human and murine breast cancer cells for their resistance to doxorubicin, Pgp expression, lysosome and proteasome activity, nitrite production, ER-dependent cell death and immunogenic cell death parameters. We evaluated the efficacy of genetic (C/EBP-β LIP induction) and pharmacological strategies (lysosome and proteasome inhibitors), in restoring the ER-dependent and immunogenic-dependent cell death induced by doxorubicin, in vitro and in syngeneic mice bearing chemoresistant TNBC. The results were analyzed by one-way analysis of variance test. RESULTS We found that TNBC cells characterized by high levels of Pgp and resistance to doxorubicin, had low induction of the ER-dependent pro-apoptotic factor C/EBP-β LIP upon doxorubicin treatment and high activities of lysosome and proteasome that constitutively destroyed LIP. The combination of chloroquine and bortezomib restored doxorubicin sensitivity by activating multiple and interconnected mechanisms. First, chloroquine and bortezomib prevented C/EBP-β LIP degradation and activated LIP-dependent CHOP/TRB3/caspase 3 axis in response to doxorubicin. Second, C/EBP-β LIP down-regulated Pgp and up-regulated calreticulin that triggered the dendritic cell (DC)-mediated phagocytosis of tumor cell, followed by the activation of anti-tumor CD8+T-lymphocytes upon doxorubicin treatment. Third, chloroquine and bortezomib increased the endogenous production of nitric oxide that further induced C/EBP-β LIP and inhibited Pgp activity, enhancing doxorubicin's cytotoxicity. In orthotopic models of resistant TNBC, intratumor C/EBP-β LIP induction - achieved by a specific expression vector or by chloroquine and bortezomib - effectively reduced tumor growth and Pgp expression, increased intra-tumor apoptosis and anti-tumor immune-infiltrate, rescuing the efficacy of doxorubicin. CONCLUSIONS We suggest that preventing C/EBP-β LIP degradation by lysosome and proteasome inhibitors triggers multiple virtuous circuitries that restore ER-dependent apoptosis, down-regulate Pgp and re-activate the DC/CD8+T-lymphocytes response against TNBC. Lysosome and proteasome inhibitors associated with doxorubicin may overcome the resistance to the drug in TNBC.
Collapse
Affiliation(s)
- Iris C Salaroglio
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Ahmad Abdullrahman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Eleonora Mungo
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy
| | - Barbara Castella
- Laboratory of Blood Tumor Immunology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Gamal Eldein Fathy Abd-Ellatef Abd-Elrahman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.,Pharmaceutical and Drug Industries Research Division, Therapeutic Chemistry Department, National Research Centre, Cairo, Egypt
| | - Massimo Massaia
- Laboratory of Blood Tumor Immunology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,Hematology Division, AO S Croce e Carle, Cuneo, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Menachem Rubinstein
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Turin, Italy.
| |
Collapse
|
9
|
Xia YQ, Hua RJ, Juan C, Zhong ZH, Tao CS, Fang R, Lin H, Rui G, Yong C. SIRT6 Depletion Sensitizes Human Hepatoma Cells to Chemotherapeutics by Downregulating MDR1 Expression. Front Pharmacol 2018; 9:194. [PMID: 29563873 PMCID: PMC5845756 DOI: 10.3389/fphar.2018.00194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/21/2018] [Indexed: 12/13/2022] Open
Abstract
Multidrug resistance (MDR) due to overexpression of MDR1 is a major obstacle that hinders the treatment of hepatocellular carcinoma (HCC). In this study, we explored the function and underlying molecular mechanism of SIRT6 in MDR of HCC. Chemotherapeutic agents (doxorubicin, cisplatin, and sorafenib) treatment increased SIRT6 mRNA and protein level in two HCC cell lines in a dose-dependent manner. SIRT6 depletion resulted in decreased cell viability and increased apoptosis in HCC cells treated with chemotherapeutic agents. Mechanistically, SIRT6 depletion reduced MDR1 transcription by targeting its promoter in HCC cells treated with chemotherapeutic agents. Consistently, the protein level of MDR1 was also reduced in SIRT6-depleted HCC cells. Further studies indicated that SIRT6 depletion may suppress CCAAT/enhancer binding protein β (C/EBPβ), to act as a transcriptional activator of MDR1 in HCC cells treated with chemotherapeutic agents. Importantly, forced expression of MDR1 could attenuate the apoptosis induced by chemotherapeutic agents in SIRT6-depleted cells. Taken together, these results indicated SIRT6 depletion enhanced chemosensitivity of human hepatoma cells by downregulating MDR1 expression through suppressing C/EBPβ. SIRT6 may serve as a novel target to enhance chemosensitivity in HCC cells.
Collapse
Affiliation(s)
- Yang Q Xia
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, China.,The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ren J Hua
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Chen Juan
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Zhou H Zhong
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Cheng S Tao
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ren Fang
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - He Lin
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Gong Rui
- The Key Laboratory of Molecular Biology of Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Chen Yong
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Roles of tumor heterogeneity in the development of drug resistance: A call for precision therapy. Semin Cancer Biol 2017; 42:13-19. [DOI: 10.1016/j.semcancer.2016.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 11/08/2016] [Indexed: 12/13/2022]
|
11
|
Hansberg-Pastor V, González-Arenas A, Camacho-Arroyo I. CCAAT/enhancer binding protein β negatively regulates progesterone receptor expression in human glioblastoma cells. Mol Cell Endocrinol 2017; 439:317-327. [PMID: 27663075 DOI: 10.1016/j.mce.2016.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/29/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Many progesterone (P4) actions are mediated by its intracellular receptor (PR), which has two isoforms (PR-A and PR-B) differentially transcribed from separate promoters of a single gene. In glioblastomas, the most frequent and aggressive brain tumors, PR-B is the predominant isoform. In an in silico analysis we showed putative CCAAT/Enhancer Binding Protein (C/EBP) binding sites at PR-B promoter. We evaluated the role of C/EBPβ in PR-B expression regulation in glioblastoma cell lines, which expressed different ratios of PR and C/EBPβ isoforms (LAP1, LAP2, and LIP). ChIP assays showed a significant basal binding of C/EBPβ, specific protein 1 (Sp1) and estrogen receptor alpha (ERα) to PR-B promoter. C/EBPβ knockdown increased PR-B expression and treatment with estradiol (E2) reduced C/EBPβ binding to the promoter and up-regulated PR-B expression. P4 induced genes were differently regulated when CEBP/β was silenced. These data show that C/EBPβ negatively regulates PR-B expression in glioblastoma cells.
Collapse
Affiliation(s)
- Valeria Hansberg-Pastor
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México (UNAM), Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, UNAM, Ciudad de México, Mexico.
| |
Collapse
|
12
|
Mikulska JE. Analysis of Response Elements Involved in the Regulation of the Human Neonatal Fc Receptor Gene (FCGRT). PLoS One 2015; 10:e0135141. [PMID: 26252948 PMCID: PMC4529178 DOI: 10.1371/journal.pone.0135141] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/19/2015] [Indexed: 12/26/2022] Open
Abstract
Human epithelial, endothelial and PMA-differentiated THP-1 cell lines were used as model systems to study the transcriptional regulation of the human FCGRT gene encoding the alpha chain of hFcRn. The data obtained from site-directed mutagenesis in transient transfection experiments indicate that the Sp1 sites at positions -641, -635, and -313, CF1/YY1 elements at positions -586 and -357, and the AP-1 motif at -276 within the-660/-233 fragment of the human FCGRT promoter (hFCGRT) participate in the regulation of human FCGRT in all selected cell lines. However, their individual contribution to promoter activity is not equivalent. The Sp1 binding site at -313 and the AP-1 site at -276 are critical for the activity of the hFCGRT promoter in epithelial and endothelial cells. Moreover, the CF1/YY1 site at -586 in differentiated THP-1 cells, plays an essential role in the transcriptional activity of the promoter. In addition, the C/EBPbeta binding site at -497 of the hFCGRT promoter in epithelial and endothelial cells, and the C/EBPbeta motif located at -497 and -233 within the hFCGRT promoter in differentiated THP-1 cells may function as positive regulatory sequences in response to LPS or PMA stimulation. EMSA and supershift analyses showed that the functionally identified binding motifs in the hFCGRT promoter were able to specifically interact with their corresponding (Sp1, Sp2, Sp3, c-Fos, c-Jun, YY1, and C/EBPbeta or C/EBPdelta) transcription factors (TFs), suggesting their possible involvement in the regulation of the human FCGRT gene expression.
Collapse
Affiliation(s)
- Joanna E. Mikulska
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- * E-mail:
| |
Collapse
|
13
|
Cao W, Liu Y, Zhang R, Zhang B, Wang T, Zhu X, Mei L, Chen H, Zhang H, Ming P, Huang L. Homoharringtonine induces apoptosis and inhibits STAT3 via IL-6/JAK1/STAT3 signal pathway in Gefitinib-resistant lung cancer cells. Sci Rep 2015; 5:8477. [PMID: 26166037 PMCID: PMC4499885 DOI: 10.1038/srep08477] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/22/2015] [Indexed: 12/25/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are mostly used in non-small cell lung cancer (NSCLC) treatment. Unfortunately, treatment with Gefitinib for a period of time will result in drug resistance and cause treatment failure in clinic. Therefore, exploring novel compounds to overcome this resistance is urgently required. Here we investigated the antitumor effect of homoharringtonine (HHT), a natural compound extracted from Cephalotaxus harringtonia, on Gefitinib-resistant NSCLC cell lines in vitro and in vivo. NCI-H1975 cells with EGFR T790M mutation are more sensitive to HHT treatment compared with that of A549 cells with wild type EGFR. HHT inhibited cells growth, cell viability and colony formation, as well as induced cell apoptosis through mitochondria pathway. Furthermore, we explored the mechanism of HHT inhibition on NSCLC cells. Higher level of interleukin-6 (IL-6) existed in lung cancer patients and mutant EGFR and TGFβ signal requires the upregulation of IL-6 through the gp130/JAK pathway to overactive STAT3, an oncogenic protein which has been considered as a potential target for cancer therapy. HHT reversiblely inhibited IL-6-induced STAT3 Tyrosine 705 phosphorylation and reduced anti-apoptotic proteins expression. Gefitinib-resistant NSCLC xenograft tests also confirmed the antitumor effect of HHT in vivo. Consequently, HHT has the potential in Gefitinib-resistant NSCLC treatment.
Collapse
Affiliation(s)
- Wei Cao
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Ying Liu
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Ran Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Bo Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Teng Wang
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xianbing Zhu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Lin Mei
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Hongbo Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Hongling Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Pinghong Ming
- Laboratory of Zhuhai People’s Hospital, Zhuhai, Guangdong, 519000, China
| | - Laiqiang Huang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Health Science and Technology (prep), Center for Biotechnology & Biomedicine and Division of Life & Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
14
|
Crawford RR, Prescott ET, Sylvester CF, Higdon AN, Shan J, Kilberg MS, Mungrue IN. Human CHAC1 Protein Degrades Glutathione, and mRNA Induction Is Regulated by the Transcription Factors ATF4 and ATF3 and a Bipartite ATF/CRE Regulatory Element. J Biol Chem 2015; 290:15878-15891. [PMID: 25931127 DOI: 10.1074/jbc.m114.635144] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Indexed: 11/06/2022] Open
Abstract
Using an unbiased systems genetics approach, we previously predicted a role for CHAC1 in the endoplasmic reticulum stress pathway, linked functionally to activating transcription factor 4 (ATF4) following treatment with oxidized phospholipids, a model for atherosclerosis. Mouse and yeast CHAC1 homologs have been shown to degrade glutathione in yeast and a cell-free system. In this report, we further defined the ATF4-CHAC1 interaction by cloning the human CHAC1 promoter upstream of a luciferase reporter system for in vitro assays in HEK293 and U2OS cells. Mutation and deletion analyses defined two major cis DNA elements necessary and sufficient for CHAC1 promoter-driven luciferase transcription under conditions of ER stress or ATF4 coexpression: the -267 ATF/cAMP response element (CRE) site and a novel -248 ATF/CRE modifier (ACM) element. We also examined the ability of the CHAC1 ATF/CRE and ACM sequences to bind ATF4 and ATF3 using immunoblot-EMSA and confirmed ATF4, ATF3, and CCAAT/enhancer-binding protein β binding at the human CHAC1 promoter in the proximity of the ATF/CRE and ACM using ChIP. To further validate the function of CHAC1 in a human cell model, we measured glutathione levels in HEK293 cells with enhanced CHAC1 expression. Overexpression of CHAC1 led to a robust depletion of glutathione, which was alleviated in a CHAC1 catalytic mutant. These results suggest an important role for CHAC1 in oxidative stress and apoptosis with implications for human health and disease.
Collapse
Affiliation(s)
- Rebecca R Crawford
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Eugenia T Prescott
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Charity F Sylvester
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Ashlee N Higdon
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Jixiu Shan
- Department of Biochemistry and Molecular Biology, Shands Cancer Center and Center for Nutritional Sciences, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Michael S Kilberg
- Department of Biochemistry and Molecular Biology, Shands Cancer Center and Center for Nutritional Sciences, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Imran N Mungrue
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112.
| |
Collapse
|
15
|
Riganti C, Kopecka J, Panada E, Barak S, Rubinstein M. The role of C/EBP-β LIP in multidrug resistance. J Natl Cancer Inst 2015; 107:djv046. [PMID: 25766403 DOI: 10.1093/jnci/djv046] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chemotherapy triggers endoplasmic reticulum (ER) stress, which in turn regulates levels of the active (LAP) and the natural dominant-negative (LIP) forms of the transcription factor C/EBP-β. LAP upregulates and LIP downregulates the multidrug resistance (MDR) protein P-glycoprotein (Pgp), but it is not known how critical is their role in establishing MDR. METHODS Cell viability was quantitated by crystal violet staining and measuring absorbance at 540nm. Expression of various proteins was determined by immunoblotting. mRNA levels were determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR). LIP and LAP were overexpressed using expression plasmids followed by selection with blasticidin. Tumor cells expressing doxycycline-inducible LIP were orthotopically implanted in mice (n = 15 mice per group), and tumor size was measured daily by caliper. Tumor sections were stained with hematoxylin and eosin and immunostained for Pgp, proliferation, and ER stress markers. RESULTS MDR cells do not express basal, chemotherapy-triggered, or ER stress-triggered LIP and fail to activate the CHOP-caspase-3 death-triggering axis upon ER stress or chemotherapy challenge. Overexpression of LIP reversed the MDR phenotype in vitro and in tumors implanted in mice. LIP was undetectable in MDR cells, probably due to its ubiquitination, which was 3.56-fold higher, resulting in lysosomal and proteasomal degradation of LIP. CONCLUSIONS Spontaneous and drug-selected MDR cells lack LIP, which is eliminated by ubiquitin-mediated degradation. Loss of LIP drives MDR not only by increasing Pgp expression but also by a two-fold attenuation of ER stress-triggered cell death.
Collapse
Affiliation(s)
- Chiara Riganti
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Elisa Panada
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Sara Barak
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Menachem Rubinstein
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR).
| |
Collapse
|
16
|
CCAAT/enhancer binding protein β in relation to ER stress, inflammation, and metabolic disturbances. BIOMED RESEARCH INTERNATIONAL 2015; 2015:324815. [PMID: 25699273 PMCID: PMC4324884 DOI: 10.1155/2015/324815] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/12/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
The prevalence of the metabolic syndrome and underlying metabolic disturbances increase rapidly in developed countries. Various molecular targets are currently under investigation to unravel the molecular mechanisms that cause these disturbances. This is done in attempt to counter or prevent the negative health consequences of the metabolic disturbances. Here, we reviewed the current knowledge on the role of C/EBP-β in these metabolic disturbances. C/EBP-β deletion in mice resulted in downregulation of hepatic lipogenic genes and increased expression of β-oxidation genes in brown adipose tissue. Furthermore, C/EBP-β is important in the differentiation and maturation of adipocytes and is increased during ER stress and proinflammatory conditions. So far, studies were only conducted in animals and in cell systems. The results found that C/EBP-β is an important transcription factor within the metabolic disturbances of the metabolic system. Therefore, it is interesting to examine the potential role of C/EBP-β at molecular and physiological level in humans.
Collapse
|
17
|
Schulze S, Reinhardt S, Freese C, Schmitt U, Endres K. Identification of trichlormethiazide as a Mdr1a/b gene expression enhancer via a dual secretion-based promoter assay. Pharmacol Res Perspect 2015; 3:e00109. [PMID: 25692026 PMCID: PMC4317239 DOI: 10.1002/prp2.109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/07/2014] [Indexed: 11/29/2022] Open
Abstract
Transporters of the ATP-binding cassette (ABC) family such as MDR1 play a pivotal role in persistence of brain homeostasis by contributing to the strict permeability properties of the blood–brain barrier. This barrier on one hand compromises treatment of central nervous system diseases by restricting access of drugs; on the other hand, an impaired or altered function of barrier building cells has been described in neurological disorders. The latter might contribute to increased vulnerability of the brain under pathological conditions or even enforce pathogenesis. Here, we present a novel approach for a systematic examination of drug impact on Mdr1 gene expression by establishing a dual reporter gene assay for the murine upstream core promoters of Mdr1a and b. We validated the time-resolved assay in comparison with single reporter gene constructs and applied it to analyze effects of a Food and Drug Administration (FDA)-approved drug library consisting of 627 substances. The chemo-preventive synthetic dithiolethione oltipraz was reidentified with our assay as an already known inducer of Mdr1 gene expression. Together with two newly characterized modifiers – gemcitabine and trichlormethiazide – we prove our findings in a blood–brain barrier culture model as well as in wild-type and Mdr1 knockout mice. In sum, we could demonstrate that our dual reporter gene assay delivers results, which also persist in the living animal and consequently is applicable for further analysis and prediction of Mdr1 regulation in vivo.
Collapse
Affiliation(s)
- Sarina Schulze
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Sven Reinhardt
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Christian Freese
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine Mainz, Germany
| | - Ulrich Schmitt
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Kristina Endres
- Clinic of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| |
Collapse
|
18
|
Munoz JL, Rodriguez-Cruz V, Greco SJ, Nagula V, Scotto KW, Rameshwar P. Temozolomide Induces the Production of Epidermal Growth Factor to Regulate MDR1 Expression in Glioblastoma Cells. Mol Cancer Ther 2014; 13:2399-411. [DOI: 10.1158/1535-7163.mct-14-0011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Singh R, Sharma MC, Sarkar C, Singh M, Chauhan SS. Transcription factor C/EBP-β mediates downregulation of dipeptidyl-peptidase III expression by interleukin-6 in human glioblastoma cells. FEBS J 2014; 281:1629-41. [PMID: 24472318 DOI: 10.1111/febs.12728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/28/2013] [Accepted: 01/22/2014] [Indexed: 11/30/2022]
Abstract
Dipeptidyl-peptidase III (DPP III) is a cytosolic metallo-aminopeptidase implicated in various physiological and pathological processes. A previous study from our laboratory indicated an elevated expression of DPP III in glioblastoma (U87MG) cells. In the present study we investigated the role of interleukin-6 (IL-6), a pleiotropic cytokine produced by glial tumors, in the regulation of DPP III expression. Immunohistochemistry, western blotting and quantitative RT-PCR were used for quantitation of DPP III and IL-6 in human glioblastoma cells and tumors. Cell transfections and DPP III promoter reporter assays were performed to study the transcriptional regulation of DPP III by IL-6. Promoter deletion analysis, site directed mutagenesis, chromatin immunoprecipitation assays and small interfering RNA (siRNA) technology was employed to elucidate the molecular mechanism of IL-6 mediated regulation of DPP III expression in glioblastoma cells. Our results for the first time demonstrate a negative correlation (r = 0.632, P = 0.01) between DPP III and IL-6 in both human tumors and cultured glioblastoma cells. Treatment of U87MG cells with IL-6 significantly decreased DPP III expression with a concomitant increase in the levels of transcription factor CCAAT/enhancer binding protein beta (C/EBP-β). Deletion/mutagenesis of C/EBP-β binding motif of DPP III promoter significantly increased its activity and abolished its responsiveness to IL-6. This effect could also be mimicked by C/EBP-β siRNA. In conclusion our study for the first time demonstrates C/EBP-β mediated transcriptional downregulation of DPP III by IL-6. Our results demonstrating a negative correlation between IL-6 and DPP III taken together with the previously reported prognostic significance of this cytokine in glioblastoma suggests that DPP III may prove useful as a prognostic marker.
Collapse
Affiliation(s)
- Ratnakar Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | |
Collapse
|
20
|
Henrique R, Oliveira AI, Costa VL, Baptista T, Martins AT, Morais A, Oliveira J, Jerónimo C. Epigenetic regulation of MDR1 gene through post-translational histone modifications in prostate cancer. BMC Genomics 2013; 14:898. [PMID: 24344919 PMCID: PMC3878566 DOI: 10.1186/1471-2164-14-898] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 12/11/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Multidrug resistance 1 (MDR1) gene encodes for an ATP binding cassette transporter--P-glycoprotein (P-gp)-- involved in chemoresistance to taxanes. MDR1 promoter methylation is frequent in prostate carcinoma (PCa), suggesting an epigenetic regulation but no functional correlation has been established. We aimed to elucidate the epigenetic mechanisms involved in MDR1 deregulation in PCa. RESULTS MDR1 promoter methylation and P-gp expression were assessed in 121 PCa, 39 high-grade prostatic intraepithelial neoplasia (HGPIN), 28 benign prostatic hyperplasia (BPH) and 10 morphologically normal prostate tissue (NPT) samples, using quantitative methylation specific PCR and immunohistochemistry, respectively. PCa cell lines were exposed to a DNA methyltransferases inhibitor 5-aza-2'deoxycytidine (DAC) and histone deacetylases inhibitor trichostatin A (TSA). Methylation and histone posttranscriptional modifications status were characterized and correlated with mRNA and protein expression. MDR1 promoter methylation levels and frequency significantly increased from NPTs, to HGPIN and to PCa. Conversely, decreased or absent P-gp immunoexpression was observed in HGPIN and PCa, inversely correlating with methylation levels. Exposure to DAC alone did not alter significantly methylation levels, although increased expression was apparent. However, P-gp mRNA and protein re-expression were higher in cell lines exposed to TSA alone or combined with DAC. Accordingly, histone active marks H3Ac, H3K4me2, H3K4me3, H3K9Ac, and H4Ac were increased at the MDR1 promoter after exposure to TSA alone or combined with DAC. CONCLUSION Our data suggests that, in prostate carcinogenesis, MDR1 downregulation is mainly due to histone post-translational modifications. This occurs concomitantly with aberrant promoter methylation, substantiating the association with P-gp decreased expression.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adult
- Aged
- Aged, 80 and over
- Azacitidine/pharmacology
- Cell Line, Tumor
- CpG Islands
- DNA Methylation
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Histones/metabolism
- Humans
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Promoter Regions, Genetic
- Prostate/metabolism
- Prostatic Hyperplasia/genetics
- Prostatic Hyperplasia/metabolism
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Processing, Post-Translational
Collapse
Affiliation(s)
- Rui Henrique
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| | - Ana Isabel Oliveira
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Vera L Costa
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Tiago Baptista
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Ana Teresa Martins
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - António Morais
- Department of Urology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Jorge Oliveira
- Department of Urology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| |
Collapse
|
21
|
JNK1/2 Activation by an Extract from the Roots of Morus alba L. Reduces the Viability of Multidrug-Resistant MCF-7/Dox Cells by Inhibiting YB-1-Dependent MDR1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:741985. [PMID: 23983799 PMCID: PMC3741934 DOI: 10.1155/2013/741985] [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: 04/05/2013] [Revised: 06/28/2013] [Accepted: 06/30/2013] [Indexed: 11/28/2022]
Abstract
Cancer cells acquire anticancer drug resistance during chemotherapy, which aggravates cancer disease. MDR1 encoded from multidrug resistance gene 1 mainly causes multidrug resistance phenotypes of different cancer cells. In this study, we demonstrate that JNK1/2 activation by an extract from the root of Morus alba L. (White mulberry) reduces doxorubicin-resistant MCF-7/Dox cell viability by inhibiting YB-1 regulation of MDR1 gene expression. When MCF-7 or MCF-7/Dox cells, where MDR1 is highly expressed were treated with an extract from roots or leaves of Morus alba L., respectively, the root extract from the mulberry (REM) but not the leaf extract (LEM) reduced cell viabilities of both MCF-7 and MCF-7/Dox cells, which was enhanced by cotreatment with doxorubicin. REM but not LEM further inhibited YB-1 nuclear translocation and its regulation of MDR1 gene expression. Moreover, REM promoted phosphorylation of c-Jun NH2-terminal kinase 1/2 (JNK1/2) and JNK1/2 inhibitor, SP600125 and rescued REM inhibition of both MDR1 expression and viabilities in MCF-7/Dox cells. Consistently, overexpression of JNK1, c-Jun, or c-Fos inhibited YB-1-dependent MDR1 expression and reduced viabilities in MCF-7/Dox cells. In conclusion, our data indicate that REM-activated JNK-cJun/c-Fos pathway decreases the viability of MCF-7/Dox cells by inhibiting YB-1-dependent MDR1 gene expression. Thus, we suggest that REM may be useful for treating multidrug-resistant cancer cells.
Collapse
|
22
|
Li T, Wang H, Sun Y, Zhao L, Gang Y, Guo X, Huang R, Yang Z, Pan Y, Wu K, Xu L, Liu Z, Fan D. Transcription factor CUTL1 is a negative regulator of drug resistance in gastric cancer. J Biol Chem 2012; 288:4135-47. [PMID: 23255599 DOI: 10.1074/jbc.m112.345942] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Gastric cancer is one of the leading causes of malignancy-related mortality worldwide, and drug resistance hampered the clinical efficacy of chemotherapy. To better understand the molecular mechanism causing drug resistance, we previously established an isogenic pair of doxorubicin-sensitive and -resistant gastric cancer cell lines, SGC7901 and SGC7901/ADR cells. Here, we investigated how modulation of CUTL1 activity affects the response of gastric cancer to frequently used chemotherapeutic agents. In this study, we demonstrated that CUTL1 transcription activity was significantly reduced in doxorubicin-resistant cells. Furthermore, decreased CUTL1 expression was strongly associated with intrinsic drug resistance in human gastric cancer tissues and could be used as a poor prognosis biomarker. Both gain-of-function (by overexpression of active CUTL1) and loss-of-function (by CUTL1-specific shRNA knockdown) studies showed that increased CUTL1 activity significantly enhanced cell sensitivity to drugs and led to increased apoptosis, whereas decreased CUTL1 expression dramatically reduced cell sensitivity to drugs and thus fewer apoptoses. Importantly, modulation of CUTL1 activity resulted in altered sensitivity to multiple drugs. In vivo mouse studies indicated that overexpression of active CUTL1 significantly resulted in increased cancer tissue response to chemotherapy and therefore inhibited growth, whereas knockdown of CUTL1 conferred resistance to chemotherapy. Taken together, our results strongly indicate that CUTL1 activity is inversely associated with drug resistance and thus is an attractive therapeutic target to modulate multidrug resistance in gastric cancer.
Collapse
Affiliation(s)
- Tingting Li
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gromnicova R, Romero I, Male D. Transcriptional control of the multi-drug transporter ABCB1 by transcription factor Sp3 in different human tissues. PLoS One 2012; 7:e48189. [PMID: 23133566 PMCID: PMC3485001 DOI: 10.1371/journal.pone.0048189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 09/24/2012] [Indexed: 11/13/2022] Open
Abstract
The ATP-binding cassette (ABC) transporter ABCB1, encoded by the multidrug resistance gene MDR1, is expressed on brain microvascular endothelium and several types of epithelium, but not on endothelia outside the CNS. It is an essential component of the blood-brain barrier. The aim of this study was to identify cell-specific controls on the transcription of MDR1 in human brain endothelium. Reporter assays identified a region of 500 bp around the transcription start site that was optimally active in brain endothelium. Chromatin immunoprecipitation identified Sp3 and TFIID associated with this region and EMSA (electrophoretic mobility shift assays) confirmed that Sp3 binds preferentially to an Sp-target site (GC-box) on the MDR1 promoter in brain endothelium. This result contrasts with findings in other cell types and with the colon carcinoma line Caco-2, in which Sp1 preferentially associates with the MDR1 promoter. Differences in MDR1 transcriptional control between brain endothelium and Caco-2 could not be explained by the relative abundance of Sp1:Sp3 nor by the ratio of Sp3 variants, because activating variants of Sp3 were present in both cell types. However differential binding of other transcription factors was also detected in two additional upstream regions of the MDR1 promoter. Identification of cell-specific controls on the transcription of MDR1 indicates that it may be possible to modulate multi-drug resistance on tumours, while leaving the blood brain barrier intact.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Blood-Brain Barrier
- Caco-2 Cells
- Cell Line, Tumor
- Cell Separation
- Endothelial Cells/cytology
- Endothelium, Vascular/cytology
- Flow Cytometry
- Gene Expression Regulation
- Humans
- Immunohistochemistry/methods
- Microscopy, Fluorescence/methods
- Promoter Regions, Genetic
- Protein Binding
- Sp3 Transcription Factor/metabolism
- Transcription Factor TFIID/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Radka Gromnicova
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, Bucks, United Kingdom
| | - Ignacio Romero
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, Bucks, United Kingdom
| | - David Male
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, Bucks, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Balaguer TM, Gómez-Martínez A, García-Morales P, Lacueva J, Calpena R, Reverte LR, Riquelme NL, Martinez-Lacaci I, Ferragut JA, Saceda M. Dual regulation of P-glycoprotein expression by trichostatin A in cancer cell lines. BMC Mol Biol 2012; 13:25. [PMID: 22846052 PMCID: PMC3441908 DOI: 10.1186/1471-2199-13-25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/27/2012] [Indexed: 02/05/2023] Open
Abstract
Background It has been reported that the histone deacetylase inhibitor (iHDAc) trichostatin A (TSA) induces an increase in MDR1 gene transcription (ABCB1). This result would compromise the use of iHDACs in combination with other cytotoxic agents that are substrates of P-glycoprotein (Pgp). It has also been reported the use of alternative promoters by the ABCB1 gene and the existence of a translational control of Pgp protein. Finally, the ABCB1 gene is located in a genetic locus with the nested gene RUNDC3B in the complementary DNA strand, raising the possibility that RUNDC3B expression could interfere with ABCB1 alternative promoter regulation. Methods A combination of RT-PCR, real time RT-PCR, Western blot and drug accumulation assays by flow cytometry has been used in this study. Results The iHDACs-induced increase in MDR1 mRNA levels is not followed by a subsequent increase in Pgp protein levels or activity in several pancreatic and colon carcinoma cell lines, suggesting a translational control of Pgp in these cell lines. In addition, the MDR1 mRNA produced in these cell lines is shorter in its 5′ end that the Pgp mRNA produced in cell lines expressing Pgp protein. The different size of the Pgp mRNA is due to the use of alternative promoters. We also demonstrate that these promoters are differentially regulated by TSA. The translational blockade of Pgp mRNA in the pancreatic carcinoma cell lines could be related to alterations in the 5′ end of the MDR1 mRNA in the Pgp protein expressing cell lines. In addition, we demonstrate that the ABCB1 nested gene RUNDC3B expression although upregulated by TSA is independent of the ABCB1 alternative promoter used. Conclusions The results show that the increase in MDR1 mRNA expression after iHDACs treatment is clinically irrelevant since this mRNA does not render an active Pgp protein, at least in colon and pancreatic cancer cell lines. Furthermore, we demonstrate that TSA in fact, regulates differentially both ABCB1 promoters, downregulating the upstream promoter that is responsible for active P-glycoprotein expression. These results suggest that iHDACs such as TSA may in fact potentiate the effects of antitumour drugs that are substrates of Pgp. Finally, we also demonstrate that TSA upregulates RUNDC3B mRNA independently of the ABCB1 promoter in use.
Collapse
Affiliation(s)
- Trinidad Mata Balaguer
- Fundación para la Investigación Biomédica del Hospital Universitario de Elche, Elche, Alicante, 03203, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen KG, Sikic BI. Molecular pathways: regulation and therapeutic implications of multidrug resistance. Clin Cancer Res 2012; 18:1863-9. [PMID: 22344233 DOI: 10.1158/1078-0432.ccr-11-1590] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multidrug transporters constitute major mechanisms of MDR in human cancers. The ABCB1 (MDR1) gene encodes a well-characterized transmembrane transporter, termed P-glycoprotein (P-gp), which is expressed in many normal human tissues and cancers. P-gp plays a major role in the distribution and excretion of drugs and is involved in intrinsic and acquired drug resistance of cancers. The regulation of ABCB1 expression is complex and has not been well studied in a clinical setting. In this review, we elucidate molecular signaling and epigenetic interactions that govern ABCB1 expression and the development of MDR in cancer. We focus on acquired expression of ABCB1 that is associated with genomic instability of cancer cells, including mutational events that alter chromatin structures, gene rearrangements, and mutations in tumor suppressor proteins (e.g., mutant p53), which guard the integrity of genome. In addition, epigenetic modifications of the ABCB1 proximal and far upstream promoters by either demethylation of DNA or acetylation of histone H3 play a pivotal role in inducing ABCB1 expression. We describe a molecular network that coordinates genetic and epigenetic events leading to the activation of ABCB1. These mechanistic insights provide additional translational targets and potential strategies to deal with clinical MDR.
Collapse
Affiliation(s)
- Kevin G Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5151., USA
| | | |
Collapse
|
26
|
Coward J, Balkwill F. Targeting Inflammatory Pathways in Epithelial Ovarian Cancer. EMERGING THERAPEUTIC TARGETS IN OVARIAN CANCER 2011:133-164. [DOI: 10.1007/978-1-4419-7216-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
27
|
Chen C, Shen HL, Yang J, Chen QY, Xu WL. Preventing chemoresistance of human breast cancer cell line, MCF-7 with celecoxib. J Cancer Res Clin Oncol 2011; 137:9-17. [PMID: 20229271 DOI: 10.1007/s00432-010-0854-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 02/19/2010] [Indexed: 12/01/2022]
Abstract
PURPOSE To investigate the preventive effect of celecoxib, a specific cyclooxygenase-2 (Cox-2) inhibitor, on the development of chemoresistance in breast cancer cell line, MCF-7, and explore the mechanism of the action. METHODS Chemoresistance phenotype was established by treating MCF-7 cells with 0.05 μg/ml doxorubicin for 7 days, and then the effect of preventive chemoresistance was investigated by the combination of same dose of doxorubicin with 10 μM celecoxib. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide assay was used to assess cytostatic efficacy of doxorubicin and 50% inhibiting concentration (IC(50)) of MCF-7 cells. RT-PCR was performed to examine mRNA expression of multidrug resistance gene 1 (MDR1) and its transcription factors c-Jun and NF-κB. Western blotting analysis was performed to detect the expression of protein. Flow cytometry (FCM) was applied to analyze the expression and function of P-glycoprotein (P-gp). Electrophoretic gel mobility shift assay (EMSA) was performed to determine the DNA-binding activity of nuclear transcription factors AP-1 and NF-κB. RESULTS Compared with sensitive MCF-7 cells, MDR1 and its transcription factors c-Jun and NF-κB were up-regulated at both mRNA level (P < 0.01) and protein level (P < 0.01) by treatment with 0.05 μg/ml doxorubicin for 7 days. After co-incubation with both the same dose of doxorubicin and 10 μM celecoxib for 7 days, both mRNA level and protein level of MDR1, c-Jun and NF-κB up-regulated by doxorubicin were partly reversed (P < 0.01); DNA-binding activity of nuclear transcription factors AP-1 and NF-κB were inhibited; and the function of P-gp was decreased (P < 0.01). When MCF-7 cells were treated with increasing doses of doxorubicin in the presence of 10 μM celecoxib, IC50 value of doxorubicin and doxorubicin plus 10 μM celecoxib was 0.67 ± 0.03 and 0.38 ± 0.04 μg/ml, respectively (P < 0.01). CONCLUSION Celecoxib effectively prevents the development of chemoresistance in breast cancer cell line MCF-7 induced by doxorubicin, which was partly involved in inhibiting the expression and DNA-binding activity of nuclear transcription factors AP-1 and NF-κB and downstream expression and function of P-gp. Furthermore, cytostatic efficacy of celecoxib and doxorubicin on MCF-7 cell was synergistic.
Collapse
Affiliation(s)
- Chen Chen
- The Affiliated People's Hospital, Jiangsu University, Zhenjiang, People's Republic of China
| | | | | | | | | |
Collapse
|
28
|
Yang J, Huang J, Chatterjee TK, Twait E, Fisher RA. A novel mechanism involving coordinated regulation of nuclear levels and acetylation of NF-YA and Bcl6 activates RGS4 transcription. J Biol Chem 2010; 285:29760-9. [PMID: 20630860 PMCID: PMC2943308 DOI: 10.1074/jbc.m110.121459] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 07/13/2010] [Indexed: 11/06/2022] Open
Abstract
Neuronally enriched RGS4 plays a critical role attenuating G protein signaling in brain, although the mechanisms regulating RGS4 expression are unknown. Here we describe a novel mechanism for transcriptional activation of RGS4 in neuron-like PC6 cells, where RGS4 is markedly induced during confluence-induced growth arrest. Transcriptional activation of RGS4 in confluent PC6 cells was accompanied by impaired G(i/o)-dependent MAPK activation. In the human RGS4 gene promoter, we identified three phylogenetically conserved cis-elements: an inverted CCAAT box element (ICE), a cAMP response element, and a B-cell lymphoma 6 (Bcl6)-binding site. The ICE and the cAMP response element mediate activation, and the Bcl6 site mediates repression of RGS4 transcription. Activation of RGS4 transcription in confluent PC6 cells is accompanied by increases in NF-YA and C/EBPβ and decreases in Bcl6 levels in the nucleus. Increases in NF-YA and C/EBPβ lead to their increased binding to the RGS4 promoter in vivo, and dominant negative forms of these proteins repressed RGS4 promoter activity. Acetylation of NF-YA and Bcl6 were increased in postconfluent cells. Trichostatin A stimulation of RGS4 promoter activity, accompanied by increased binding of NF-YA and decreased binding of Bcl6 to the promoter, was abolished by mutation of the ICE and enhanced by mutation of the Bcl6 site. These findings demonstrate a dynamic and coordinated regulation of nuclear levels and acetylation status of trans-acting factors critical in determining the off/on state of the RGS4 promoter.
Collapse
Affiliation(s)
| | - Jie Huang
- From the Departments of Pharmacology and
| | - Tapan K. Chatterjee
- the Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45221
| | - Erik Twait
- Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | | |
Collapse
|
29
|
Wu Z, Zhang W, Lu Y, Lu C. Transcriptome profiling of zebrafish infected with Streptococcus suis. Microb Pathog 2010; 48:178-87. [DOI: 10.1016/j.micpath.2010.02.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 02/23/2010] [Accepted: 02/25/2010] [Indexed: 10/19/2022]
|
30
|
Pokharel∗ YR, Kim∗ ND, Han HK, Oh WK, Kang KW. Increased Ubiquitination of Multidrug Resistance 1 by Ginsenoside Rd. Nutr Cancer 2010; 62:252-9. [DOI: 10.1080/01635580903407171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Kim SN, Kim NH, Lee W, Seo DW, Kim YK. Histone deacetylase inhibitor induction of P-glycoprotein transcription requires both histone deacetylase 1 dissociation and recruitment of CAAT/enhancer binding protein beta and pCAF to the promoter region. Mol Cancer Res 2009; 7:735-44. [PMID: 19435809 DOI: 10.1158/1541-7786.mcr-08-0296] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although histone deacetylase (HDAC) inhibitors are appreciated as a promising class of anticancer drugs, recent reports show that P-glycoprotein (P-gp) is induced by HDAC inhibitor treatment in cancer cells, resulting in multidrug resistance of cancer cells to other chemotherapeutic agents. In this study, we investigated the molecular mechanism of HDAC inhibitor induction of P-gp expression. HDAC inhibitor treatment causes cell type-specific induction of P-gp expression without changes in the CpG methylation status of the promoter region. In addition, our data show that HDAC inhibitor does not alter the DNA binding activity of Sp1 but facilitates both the recruitment of a coactivator complex that includes CAAT/enhancer binding protein beta and pCAF and the dissociation of the repressive complex, HDAC1, to the Sp1 binding region. Subsequently, the hyperacetylated histone H3 becomes enriched in the promoter region, leading to RNA polymerase II recruitment to activate P-gp gene transcription. Furthermore, specific down-regulation of HDAC1, but not HDAC2, by RNA silencing was enough to induce P-gp expression in HeLa cells, strongly supporting the essential role of HDAC1 in HDAC inhibitor induction of P-gp. Concomitantly, cell type-specific induction of P-gp expression seems to be dependent on phosphatidylinositol 3-kinase activity. Taken together, our findings show that HDAC inhibitor treatment leads to an increase in P-gp expression through dynamic changes in chromatin structure and transcription factor association within the promoter region.
Collapse
Affiliation(s)
- Su-Nam Kim
- KIST Gangneung Institute, Gangneung, Korea
| | | | | | | | | |
Collapse
|
32
|
CCAAT/enhancer-binding protein beta: its role in breast cancer and associations with receptor tyrosine kinases. Expert Rev Mol Med 2009; 11:e12. [PMID: 19351437 DOI: 10.1017/s1462399409001033] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The CCAAT/enhancer-binding proteins (C/EBPs) are a family of leucine-zipper transcription factors that regulate gene expression to control cellular proliferation, differentiation, inflammation and metabolism. Encoded by an intronless gene, C/EBPbeta is expressed as several distinct protein isoforms (LAP1, LAP2, LIP) whose expression is regulated by the differential use of several in-frame translation start sites. LAP1 and LAP2 are transcriptional activators and are associated with differentiation, whereas LIP is frequently elevated in proliferative tissue and acts as a dominant-negative inhibitor of transcription. However, emerging evidence suggests that LIP can serve as a transcriptional activator in some cellular contexts, and that LAP1 and LAP2 might also have unique actions. The LIP:LAP ratio is crucial for the maintenance of normal growth and development, and increases in this ratio lead to aggressive forms of breast cancer. This review discusses the regulation of C/EBPbeta activity by post-translational modification, the individual actions of LAP1, LAP2 and LIP, and the functions and downstream targets that are unique to each isoform. The role of the C/EBPbeta isoforms in breast cancer is discussed and emphasis is placed on their interactions with receptor tyrosine kinases.
Collapse
|
33
|
Han CY, Cho KB, Choi HS, Han HK, Kang KW. Role of FoxO1 activation in MDR1 expression in adriamycin-resistant breast cancer cells. Carcinogenesis 2008; 29:1837-44. [PMID: 18390843 DOI: 10.1093/carcin/bgn092] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The development of multidrug resistance 1 (MDR1) can be mediated by a number of different mechanisms but elevated gene expression of MDR1 (P-glycoprotein) has often been a major cause of chemoresistance in many cancer cells. Therefore, the present study aimed to investigate the role of forkhead box-containing protein, O subfamily (FoxO), transcription factors in regulating the MDR1 gene expression. The proximal promoter region of the human MDR1 contained a putative FoxO-binding site, which partially overlapped with the enhancer/enhancer-binding protein beta-binding region. Gel shift and immunoblot analysis of subcellular fractions revealed that nuclear levels of FoxO1 and its DNA-binding activity were selectively enhanced in MCF-7/ADR cells, which was reversed by a FoxO1 antibody. Reporter gene assays showed that the transcription of MDR1 gene is stimulated by FoxO1 overexpression. Moreover, both MDR1 expression and doxorubicin resistance in MCF-7/ADR cells were reversed by FoxO1 small interfering RNA (siRNA). The MDR1 expression in MCF-7/ADR cells was also inhibited by insulin, a functional FoxO1 inactivator. In conclusion, FoxO1 is a novel transcriptional activator of MDR1 and is crucial for MDR1 induction in MCF-7/ADR cells.
Collapse
Affiliation(s)
- Chang-Yeob Han
- BK21 Project Team, College of Pharmacy, Chosun University, Gwangju 501-759, South Korea
| | | | | | | | | |
Collapse
|
34
|
Harris KE, Jeffery EH. Sulforaphane and erucin increase MRP1 and MRP2 in human carcinoma cell lines. J Nutr Biochem 2008; 19:246-54. [PMID: 17618109 DOI: 10.1016/j.jnutbio.2007.02.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Revised: 01/24/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
Multidrug resistance (MDR) transporters have been termed the Phase III detoxification system because they not only export endogenous metabolites but provide protection from xenobiotic insult by actively secreting foreign compounds and their metabolites from tissues. However, MDR overexpression in tumors can lead to drug resistance, a major obstacle in the treatment of many cancers, including lung cancer. Isothiocyanates from cruciferous vegetables, such as sulforaphane (SF) and erucin (ER), are known to enhance the expression of Phase II detoxification enzymes. Here we evaluated the ability of SF and ER to modulate MDR mRNA and protein expressions, as well as transporter activity. The expression of P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1) and multidrug resistance protein 2 (MRP2) in liver (HepG2), colon (Caco-2) and lung (A549) cancer cells treated with ER or SF was analyzed by Western blotting. Neither SF nor ER affected P-gp expression in any of the cell lines tested. Both SF and ER increased the protein levels of MRP1 and MRP2 in HepG2 cells and of MRP2 in Caco-2 cells in a dose-dependent manner. In A549 lung cancer cells, SF increased MRP1 and MRP2 mRNA and protein levels; ER caused a similar yet smaller increase in MRP1 and MRP2 mRNA. In addition, SF and ER increased MRP1-dependent efflux of 5-carboxyfluorescein diacetate in A549 cells, although again the effect of SF was substantially greater than that of ER. The implication of these findings is that dietary components that modulate detoxification systems should be studied carefully before being recommended for use during chemotherapy, as these compounds may have additional influences on the disposition of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Kristin E Harris
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | | |
Collapse
|
35
|
Raguz S, Randle RA, Sharpe ER, Foekens JA, Sieuwerts AM, Meijer-van Gelder ME, Melo JV, Higgins CF, Yagüe E. Production of P-glycoprotein from the MDR1 upstream promoter is insufficient to affect the response to first-line chemotherapy in advanced breast cancer. Int J Cancer 2008; 122:1058-67. [PMID: 17955490 DOI: 10.1002/ijc.23149] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Multidrug resistance, the phenomenon by which cells treated with a drug become resistant to the cytotoxic effect of a variety of other structurally and functionally unrelated drugs, is often associated with the expression of P-glycoprotein, an efflux membrane pump coded by the MDR1 (ABCB1) gene. Transcription from MDR1 can start at 2 promoters: a well-characterized downstream promoter and an as yet uncharacterized upstream promoter (USP). We have previously determined that the USP is activated in some drug-resistant cell lines, in primary breast tumors and in metastatic epithelial cells isolated from the lymph nodes of breast cancer patients. In this study, we report the cloning and characterization of the MDR1 USP and studied its association with chemotherapy response in breast cancer patients. Deletion analysis indicated that a nearby endogenous retroviral long terminal repeat is not responsible for promoter activation, and that the region within the first 400 nucleotides upstream from the transcription start point contained all the elements necessary for promoter activity in drug-resistant cells. We identified an element recognized by the transcription factor NF-IL6 (activated upon interleukin-6 exposure) which is necessary for promoter activity in drug-resistant cells and plays a role in the activation of the promoter in response to interleukin-6 in breast cancer MCF-7 cells. Although transcripts from this promoter are associated with translating polyribosomes, their low abundance makes the amount of synthesized P-glycoprotein insufficient to affect the response to first-line chemotherapy in patients with advanced breast cancer.
Collapse
Affiliation(s)
- Selina Raguz
- Medical Research Council Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hong DS, Angelo LS, Kurzrock R. Interleukin-6 and its receptor in cancer: implications for translational therapeutics. Cancer 2007; 110:1911-28. [PMID: 17849470 DOI: 10.1002/cncr.22999] [Citation(s) in RCA: 310] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin-6 (IL-6) plays a major role in the response to injury or infection and is involved in the immune response, inflammation, and hematopoiesis. Its deregulation impacts numerous disease states, including many types of cancer. Consequently, modulating IL-6 may be an innovative therapeutic strategy in several diseases. A review of relevant published literature regarding IL-6 and its receptor was performed. In addition, a review of the relevance of this cytokine system to human illness, particularly in cancer, was undertaken. IL-6 is a pleiotropic cytokine that is involved in the physiology of virtually every organ system. Aberrant expression of this cytokine has been implicated in diverse human illnesses, most notably inflammatory and autoimmune disorders, coronary artery and neurologic disease, gestational problems, and neoplasms. In cancer, high levels of circulating IL-6 are observed in almost every type of tumor studied and predict a poor outcome. Furthermore, elevated IL-6 levels are associated strongly with several of the striking phenotypic features of cancer. Several molecules have been developed recently that target the biologic function of IL-6. Early results in the clinic suggest that this strategy may have a significant salutary impact on diverse tumors. The field of cytokine research has yielded a deep understanding of the fundamental role of IL-6 and its receptor in health and disease. Therapeutic targeting of IL-6 and its receptor in cancer has strong biologic rationale, and there is preliminary evidence suggesting that targeting of the IL-6 system may be beneficial in the treatment of cancer.
Collapse
Affiliation(s)
- David S Hong
- Phase I Program, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
37
|
Gómez-Martínez A, García-Morales P, Carrato A, Castro-Galache MD, Soto JL, Carrasco-García E, García-Bautista M, Guaraz P, Ferragut JA, Saceda M. Post-transcriptional Regulation of P-Glycoprotein Expression in Cancer Cell Lines. Mol Cancer Res 2007; 5:641-53. [PMID: 17579122 DOI: 10.1158/1541-7786.mcr-06-0177] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The present study of inhibitors shows that the histone deacetylase-induced increase in P-glycoprotein (Pgp) mRNA (MDR1 mRNA) does not parallel either an increase in Pgp protein or an increase in Pgp activity in several colon carcinoma cell lines. Furthermore, studying the polysome profile distribution, we show a translational control of Pgp in these cell lines. In addition, we show that the MDR1 mRNA produced in these cell lines is shorter in its 5' end that the MDR1 mRNA produced in the MCF-7/Adr (human breast carcinoma) and K562/Adr (human erythroleukemia) cell lines, both of them expressing Pgp. The different size of the MDR1 mRNA is due to the use of alternative promoters. Our data suggest that the translational blockade of MDR1 mRNA in the colon carcinoma cell lines and in wild-type K562 cells could be overcome by alterations in the 5' end of the MDR1 mRNA in the resistant variant of these cell lines, as in the case of the K562/Adr cell line. This is, to our knowledge, the first report demonstrating that the presence of an additional 5' untranslated fragment in the MDR1 mRNA improves the translational efficiency of this mRNA.
Collapse
MESH Headings
- 5' Untranslated Regions
- ATP Binding Cassette Transporter, Subfamily B/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Neoplastic
- Histone Deacetylase Inhibitors
- Humans
- K562 Cells
- Models, Genetic
- Nucleic Acid Conformation
- Promoter Regions, Genetic
- RNA Processing, Post-Transcriptional
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Angeles Gómez-Martínez
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, 03202 Elche (Alicante), Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Han HK, Han CY, Cheon EP, Lee J, Kang KW. Role of hypoxia-inducible factor-alpha in hepatitis-B-virus X protein-mediated MDR1 activation. Biochem Biophys Res Commun 2007; 357:567-73. [PMID: 17433259 DOI: 10.1016/j.bbrc.2007.04.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 04/04/2007] [Indexed: 02/06/2023]
Abstract
The transition from chemotherapy-responsive cancer cells to chemotherapy-resistant cancer cells is mainly accompanied by the increased expression of multi-drug resistance 1 (MDR1). We found that hepatitis-B-virus X protein (HBx) increases the transcriptional activity and protein level of MDR1 in a hepatoma cell line, H4IIE. In addition, HBx overexpression made H4IIE cells more resistant to verapamil-uptake. HBx stabilized hypoxia-inducible factor-1alpha (HIF-1alpha) and induced the nuclear translocation of C/EBPbeta. Reporter gene analyses showed that HBx increased the reporter activity in the cells transfected with the reporter containing MDR1 gene promoter. Moreover, the luciferase reporter gene activity was significantly inhibited by HIF-1alpha siRNA but not by overexpression of C/EBP dominant negative mutant. These results imply that HBx increases the MDR1 transporter activity through the transcriptional activation of the MDR1 gene with HIF-1alpha activation, and suggest HIF-1alpha for the therapeutic target of HBV-mediated chemoresistance.
Collapse
Affiliation(s)
- Hyo-Kyung Han
- BK21 Project Team, College of Pharmacy, Chosun University, Gwangju 501-759, South Korea
| | | | | | | | | |
Collapse
|
39
|
Merienne K, Friedman J, Akimoto M, Abou-Sleymane G, Weber C, Swaroop A, Trottier Y. Preventing polyglutamine-induced activation of c-Jun delays neuronal dysfunction in a mouse model of SCA7 retinopathy. Neurobiol Dis 2006; 25:571-81. [PMID: 17189700 PMCID: PMC1858671 DOI: 10.1016/j.nbd.2006.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2006] [Revised: 10/27/2006] [Accepted: 11/01/2006] [Indexed: 11/23/2022] Open
Abstract
We have approached the role of cellular stress in neurodegenerative diseases caused by polyglutamine expansion (polyQ) in the context of Spinocerebellar ataxia type 7 (SCA7) that includes retinal degeneration. Using the R7E mouse, in which polyQ-ataxin-7 is specifically over-expressed in rod photoreceptors, we previously showed that rod dysfunction correlated to moderate and prolonged activation of the JNK/c-Jun stress pathway. SCA7 retinopathy was also associated with reduced expression of rod-specific genes, including the transcription factor Nrl, which is essential for rod differentiation and function. Here, we report that R7E retinopathy is improved upon breeding with the JunAA knock-in mice, in which JNK-mediated activation of c-Jun is compromised. Expression of Nrl and its downstream targets, which are involved in phototranduction, are partially restored in the JunAA-R7E mice. We further show that c-Jun can directly repress the transcription of Nrl. Our studies suggest that polyQ-induced cellular stress leads to repression of genes necessary for neuronal fate and function.
Collapse
Affiliation(s)
- Karine Merienne
- Department of Molecular Pathology, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/ULP, BP10142, 67404 Illkirch Cédex, CU de Strasbourg
- Chaire de Génétique Humaine, Collège de France
| | - James Friedman
- Department of Ophthalmology & Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Masayuki Akimoto
- Department of Ophthalmology & Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
- Translational Research Center, Kyoto University Hospital, Japan
| | - Gretta Abou-Sleymane
- Department of Molecular Pathology, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/ULP, BP10142, 67404 Illkirch Cédex, CU de Strasbourg
- Chaire de Génétique Humaine, Collège de France
| | - Chantal Weber
- Department of Molecular Pathology, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/ULP, BP10142, 67404 Illkirch Cédex, CU de Strasbourg
- Chaire de Génétique Humaine, Collège de France
| | - Anand Swaroop
- Department of Ophthalmology & Visual Sciences, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Human Genetics, W.K. Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yvon Trottier
- Department of Molecular Pathology, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/ULP, BP10142, 67404 Illkirch Cédex, CU de Strasbourg
- Chaire de Génétique Humaine, Collège de France
| |
Collapse
|
40
|
Wang YC, Juric D, Francisco B, Yu RX, Duran GE, Chen KG, Chen X, Sikic BI. Regional activation of chromosomal arm 7q with and without gene amplification in taxane-selected human ovarian cancer cell lines. Genes Chromosomes Cancer 2006; 45:365-74. [PMID: 16382445 DOI: 10.1002/gcc.20300] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Taxanes are important drugs in the treatment of ovarian and other cancers, but their efficacy is limited by intrinsic and acquired drug resistance. Expression of the multidrug transporter P-glycoprotein, encoded by the MDR1 (ABCB1) gene, is one of the causes of clinical drug resistance to taxanes. To study the mechanisms of MDR1 activation related to taxanes, we established 11 multidrug-resistant variants from six ovarian cancer cell lines by continuous exposure to either paclitaxel or docetaxel. We profiled gene expression and gene copy number alterations in these cell lines using cDNA microarrays and identified a cluster of genes coactivated with MDR1 in 7q21.11-13. Regional activation was evident in nine resistant variants displaying a coexpression pattern of up to 22 genes over an 8-Mb area, including SRI, MGC4175, CLDN12, CROT, and CDK6. In six of these variants, regional activation was driven by gene copy number alterations, with low-level gains or high-level amplifications spanning the involved region. However, three variants displayed regional increases in gene expression even without concomitant gene copy number changes. These results suggest that regional gene activation may be a fundamental mechanism for acquired drug resistance, with or without changes in gene dosage. In addition to numerical and structural chromosomal changes driven by genome instability in cancer cells, other mechanisms might be involved in MDR1 regional activation, such as chromatin remodeling and DNA or histone modifications of the 7q21 region.
Collapse
Affiliation(s)
- Yan C Wang
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chu F, Chou PM, Zheng X, Mirkin BL, Rebbaa A. Control of Multidrug Resistance Gene mdr1 and Cancer Resistance to Chemotherapy by the Longevity Gene sirt1. Cancer Res 2005; 65:10183-7. [PMID: 16288004 DOI: 10.1158/0008-5472.can-05-2002] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Irreversible growth arrest (also called senescence) has emerged recently as a tumor suppressor mechanism and a key determinant of cancer chemotherapy outcome. Previous work from our laboratory suggested that the cellular ability to undergo or to escape senescence dictates its fate to become drug-sensitive or drug-resistant, respectively. In the present study, we made the hypothesis that longevity genes, by virtue of their ability to inhibit senescence, may contribute to the onset of drug resistance. We report that expression of the longevity gene sirt1 increased both at the RNA and protein levels in all the five drug-resistant cell lines tested when compared with their drug-sensitive counterparts. In addition, biopsies from cancer patients treated with chemotherapeutic agents also expressed high levels of this molecule. These changes were specific for sirt1 because the expression of other members of its family was not affected. More importantly, small interfering RNA-mediated down-regulation of sirt1 significantly reversed the resistance phenotype and reduced expression of the multidrug resistance molecule P-glycoprotein. This was further confirmed by ectopic overexpression of sirt1, which induced expression of P-glycoprotein and rendered cells resistant to doxorubicin. Collectively, these findings uncovered a novel function for the longevity gene sirt1 as a potential target for diagnosis and/or treatment of cancer resistance to chemotherapy. They also describe a proof of principle that signaling pathways implicated in longevity may share similarities with those leading to development of drug resistance in cancer.
Collapse
Affiliation(s)
- Fei Chu
- Department of Pediatrics, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60614, USA
| | | | | | | | | |
Collapse
|
42
|
Chen KG, Wang YC, Schaner ME, Francisco B, Durán GE, Juric D, Huff LM, Padilla-Nash H, Ried T, Fojo T, Sikic BI. Genetic and Epigenetic Modeling of the Origins of Multidrug-Resistant Cells in a Human Sarcoma Cell Line. Cancer Res 2005; 65:9388-97. [PMID: 16230402 DOI: 10.1158/0008-5472.can-04-4133] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The origin of drug-resistant cells in human cancers has been a fundamental problem of cancer pharmacology. Two major contrasting hypotheses (genetics versus epigenetics) have been proposed to elucidate the mechanisms of acquired drug resistance. In this study, we answer these fundamental questions through investigation of the genetic and epigenetic pathways that control the origin of ABCB1 (MDR1) gene activation with acquired multidrug resistance in drug-sensitive human sarcoma (MES-SA cells). The genetic and epigenetic bases of this selected activation involve the initiation of transcription at a site 112 kb upstream of the ABCB1 proximal promoter (P1) in the drug-resistant cells. This activation was associated with a chromatin-remodeling process characterized by an increase in acetylated histone H3 within a 968-bp region 5' of the ABCB1 upstream promoter. These alterations provide both genetic and epigenetic susceptibility for ABCB1 expression in drug-resistant cells. Complete activation of the ABCB1 gene through the coding region was proposed by interactions of selected trans-alterations or epigenetic changes on the ABCB1 proximal promoter, which occurred during initial drug exposure. Thus, our data provide evidence for a major genomic alteration that changes the chromatin structure of the ABCB1 upstream promoter via acetylation of histone H3 initiating ABCB1 activation, further elucidating the genetic and epigenetic bases that determine chemotherapeutic response in drug-resistant derivatives of MES-SA cells.
Collapse
Affiliation(s)
- Kevin G Chen
- Division of Oncology, Department of Medicine and Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kuwano M, Oda Y, Izumi H, Yang SJ, Uchiumi T, Iwamoto Y, Toi M, Fujii T, Yamana H, Kinoshita H, Kamura T, Tsuneyoshi M, Yasumoto K, Kohno K. The role of nuclear Y-box binding protein 1 as a global marker in drug resistance. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.1485.3.11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Gene expression can be regulated by nuclear factors at the transcriptional level. Many such factors regulate MDR1 gene expression, but what are the sequence elements and transcription factors that control the basal and inducible expression of this gene? The general principles through which transcription factors participate in drug resistance are now beginning to be understood. Here, we review the factors involved in the transcriptional regulation of the MDR1 gene. In particular, we focus on the transcription factor Y-box binding protein 1 and discuss the possible links between Y-box binding protein 1 expression and drug resistance in cancer, which are mediated by the transmembrane P-glycoprotein or non–P-glycoprotein.
Collapse
Affiliation(s)
- Michihiko Kuwano
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
| | | | | | - Song-Ju Yang
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
| | | | - Yukihide Iwamoto
- 8Orthopedic Surgery, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Masakazu Toi
- 9Breast Oncology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Teruhiko Fujii
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
- 2Surgery and
| | - Hideaki Yamana
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
- 2Surgery and
| | - Hisafumi Kinoshita
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
- 2Surgery and
| | - Toshiharu Kamura
- 1Research Center for Innovative Cancer Therapy of the 21st Century COE Program for Medical Science, and Departments of
- 3Gynecology Obstetrics, Kurume University, Fukuoka, Japan; Departments of
| | | | - Kosei Yasumoto
- 5Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan; Departments of
| | | |
Collapse
|