1
|
Wang J, Qi C, Liu L, Zhao L, Cui W, Tian Y, Liu B, Li J. Taurine Protects Primary Neonatal Cardiomyocytes Against Apoptosis Induced by Hydrogen Peroxide. Int Heart J 2018; 59:190-196. [DOI: 10.1536/ihj.16-372] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Junnan Wang
- Department of Angiocardiology, the Second Hospital of Jilin University
| | - Chao Qi
- Department of Angiocardiology, the Second Hospital of Jilin University
| | - Lulu Liu
- Department of Angiocardiology, the Second Hospital of Jilin University
| | - Lijing Zhao
- Department of Pharmacology, College of Basic Medical Science, Jilin University
| | - Wenzhang Cui
- Department of Angiocardiology, Jilin Province People's Hospital
| | - Yuantong Tian
- Department of Angiocardiology, the Second Hospital of Jilin University
| | - Bin Liu
- Department of Angiocardiology, the Second Hospital of Jilin University
| | - Jing Li
- Department of Pharmacology, College of Basic Medical Science, Jilin University
| |
Collapse
|
2
|
Xu K, Chen G, Qiu Y, Yuan Z, Li H, Yuan X, Sun J, Xu J, Liang X, Yin P. miR-503-5p confers drug resistance by targeting PUMA in colorectal carcinoma. Oncotarget 2017; 8:21719-21732. [PMID: 28423513 PMCID: PMC5400618 DOI: 10.18632/oncotarget.15559] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/22/2017] [Indexed: 01/04/2023] Open
Abstract
The development of multidrug-resistance (MDR) is a major contributor to death in colorectal carcinoma (CRC). Here, we investigated the possible role of microRNA (miR)-503-5p in drug resistant CRC cells. Unbiased microRNA array screening revealed that miR-503-5p is up-regulated in two oxaliplatin (OXA)-resistant CRC cell lines. Overexpression of miR-503-5p conferred resistance to OXA-induced apoptosis and inhibition of tumor growth in vitro and in vivo through down-regulation of PUMA expression. miR-503-5p knockdown sensitized chemoresistant CRC cells to OXA. Our studies indicated that p53 suppresses miR-503-5p expression and that deletion of p53 upregulates miR-503-5p expression. Inhibition of miR-503-5p in p53 null cells increased their sensitivity to OXA treatment. Importantly, analysis of patient samples showed that expression of miR-503-5p negatively correlates with PUMA in CRC. These results indicate that a p53/miR-503-5p/PUMA signaling axis regulates the CRC response to chemotherapy, and suggest that miR-503-5p plays an important role in the development of MDR in CRC by modulating PUMA expression.
Collapse
Affiliation(s)
- Ke Xu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Guo Chen
- Department of Radiation Oncology, School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, USA
| | - Yanyan Qiu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China.,Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| | - Zeting Yuan
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Hongchang Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| | - Xia Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Jian Sun
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Jianhua Xu
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China.,Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| |
Collapse
|
3
|
Guo B, Li L, Guo J, Liu A, Wu J, Wang H, Shi J, Pang D, Cao Q. M2 tumor-associated macrophages produce interleukin-17 to suppress oxaliplatin-induced apoptosis in hepatocellular carcinoma. Oncotarget 2017; 8:44465-44476. [PMID: 28591705 PMCID: PMC5546494 DOI: 10.18632/oncotarget.17973] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 05/07/2017] [Indexed: 11/25/2022] Open
Abstract
M2 macrophages are a major component of the tumor microenvironment and are important promoters of tumor occurrence and progression. In this study, we detected large numbers of M2 macrophages in hepatocellular carcinoma tissues using immunohistochemistry and immunofluorescence. Moreover, upon oxaliplatin treatment, the M2 macrophages overexpressed interleukin-17, an important inflammatory cytokine, and thus inhibited oxaliplatin-induced apoptosis. By knocking down the interleukin-17 receptor and lysosome-associated membrane protein 2A (a key protein in chaperone-mediated autophagy) in hepatocellular carcinoma cells, we found that interleukin-17 stimulated chaperone-mediated autophagy, which further suppressed apoptosis upon oxaliplatin treatment. Chaperone-mediated autophagy induced tolerance to oxaliplatin treatment by reducing cyclin D1 expression; thus, cyclin D1 overexpression stimulated oxaliplatin-induced apoptosis. In addition, cyclin D1 expression was inhibited by interleukin-17, but increased when the interleukin-17 receptor was knocked down. Thus M2 macrophages in the hepatocellular carcinoma microenvironment generate large amounts of interleukin-17, which suppress oxaliplatin-induced tumor cell apoptosis by activating chaperone-mediated autophagy and in turn reducing cyclin D1 expression. These findings may facilitate the development of novel therapeutic strategies for chemorefractory liver cancer.
Collapse
Affiliation(s)
- Bin Guo
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Leilei Li
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Jiapei Guo
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Aidong Liu
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Jinghua Wu
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Haixin Wang
- Hospital of Traditional Chinese Medicine of Tangshan City, Tangshan, Hebei, China
| | - Jun Shi
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Dequan Pang
- North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Qing Cao
- Hebei Medical University Second Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
4
|
Xiao H, Qi R, Li T, Awuah SG, Zheng Y, Wei W, Kang X, Song H, Wang Y, Yu Y, Bird MA, Jing X, Yaffe MB, Birrer MJ, Ghoroghchian PP. Maximizing Synergistic Activity When Combining RNAi and Platinum-Based Anticancer Agents. J Am Chem Soc 2017; 139:3033-3044. [PMID: 28166401 DOI: 10.1021/jacs.6b12108] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RNAi approaches have been widely combined with platinum-based anticancer agents to elucidate cellular responses and to target gene products that mediate acquired resistance. Recent work has demonstrated that platination of siRNA prior to transfection may negatively influence RNAi efficiency based on the position and sequence of its guanosine nucleosides. Here, we used detailed spectroscopic characterization to demonstrate rapid formation of Pt-guanosine adducts within 30 min after coincubation of oxaliplatin [OxaPt(II)] or cisplatin [CisPt(II)] with either guanosine monophosphate or B-cell lymphoma 2 (BCL-2) siRNA. After 3 h of exposure to these platinum(II) agents, >50% of BCL-2 siRNA transcripts were platinated and unable to effectively suppress mRNA levels. Platinum(IV) analogues [OxaPt(IV) or CisPt(IV)] did not form Pt-siRNA adducts but did display decreased in vitro uptake and reduced potency. To overcome these challenges, we utilized biodegradable methoxyl-poly(ethylene glycol)-block-poly(ε-caprolactone)-block-poly(l-lysine) (mPEG-b-PCL-b-PLL) to generate self-assembled micelles that covalently conjugated OxaPt(IV) and/or electrostatically complexed siRNA. We then compared multiple strategies by which to combine BCL-2 siRNA with either OxaPt(II) or OxaPt(IV). Overall, we determined that the concentrations of siRNA (nM) and platinum(II)-based anticancer agents (μM) that are typically used for in vitro experiments led to rapid Pt-siRNA adduct formation and ineffective RNAi. Coincorporation of BCL-2 siRNA and platinum(IV) analogues in a single micelle enabled maximal suppression of BCL-2 mRNA levels (to <10% of baseline), augmented the intracellular levels of platinum (by ∼4×) and the numbers of resultant Pt-DNA adducts (by >5×), increased the cellular fractions that underwent apoptosis (by ∼4×), and enhanced the in vitro antiproliferative activity of the corresponding platinum(II) agent (by 10-100×, depending on the cancer cell line). When combining RNAi and platinum-based anticancer agents, this generalizable strategy may be adopted to maximize synergy during screening or for therapeutic delivery.
Collapse
Affiliation(s)
- Haihua Xiao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Ruogu Qi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Ting Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Samuel G Awuah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States.,Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Yaorong Zheng
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States.,Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Wei Wei
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School , Boston, Massachusetts 02114, United States
| | - Xiang Kang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Haiqin Song
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Yongheng Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Yingjie Yu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Molly A Bird
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Michael J Birrer
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School , Boston, Massachusetts 02114, United States
| | - P Peter Ghoroghchian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States.,Dana-Farber Cancer Institute , Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Kotelevets L, Chastre E, Desmaële D, Couvreur P. Nanotechnologies for the treatment of colon cancer: From old drugs to new hope. Int J Pharm 2016; 514:24-40. [DOI: 10.1016/j.ijpharm.2016.06.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/03/2016] [Accepted: 06/04/2016] [Indexed: 12/15/2022]
|
6
|
Shan J, Xuan Y, Zhang Q, Zhu C, Liu Z, Zhang S. Ursolic acid synergistically enhances the therapeutic effects of oxaliplatin in colorectal cancer. Protein Cell 2016; 7:571-85. [PMID: 27472952 PMCID: PMC4980335 DOI: 10.1007/s13238-016-0295-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Oxaliplatin is a key drug in chemotherapy of colorectal cancer (CRC). However, its efficacy is unsatisfied due to drug resistance of cancer cells. In this study, we tested whether a natural agent, ursolic acid, was able to enhance the efficacy of oxaliplatin for CRC. Four CRC cell lines including SW480, SW620, LoVo, and RKO were used as in vitro models, and a SW620 xenograft mouse model was used in further in vivo study. We found that ursolic acid inhibited proliferation and induced apoptosis of all four cells and enhanced the cytotoxicity of oxaliplatin. This effect was associated with down-regulation of Bcl-xL, Bcl-2, survivin, activation of caspase-3, 8, 9, and inhibition of KRAS expression and BRAF, MEK1/2, ERK1/2, p-38, JNK, AKT, IKKα, IκBα, and p65 phosphorylation of the MAPK, PI3K/AKT, and NF-κB signaling pathways. The two agents also showed synergistic effects against tumor growth in vivo. In addition, ursolic acid restored liver function and body weight of the mice treated with oxaliplatin. Thus, we concluded that ursolic acid could enhance the therapeutic effects of oxaliplatin against CRC both in vitro and in vivo, which offers an effective strategy to minimize the burden of oxaliplatin-induced adverse events and provides the groundwork for a new clinical strategy to treat CRC.
Collapse
Affiliation(s)
- Jianzhen Shan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yanyan Xuan
- Cancer Institute of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Chunpeng Zhu
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhen Liu
- Cancer Institute of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Suzhan Zhang
- Cancer Institute of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
7
|
Chow MJ, Babak MV, Wong DYQ, Pastorin G, Gaiddon C, Ang WH. Structural Determinants of p53-Independence in Anticancer Ruthenium-Arene Schiff-Base Complexes. Mol Pharm 2016; 13:2543-54. [PMID: 27174050 DOI: 10.1021/acs.molpharmaceut.6b00348] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
p53 is a key tumor suppressor gene involved in key cellular processes and implicated in cancer therapy. However, it is inactivated in more than 50% of all cancers due to mutation or overexpression of its negative regulators. This leads to drug resistance and poor chemotherapeutic outcome as most clinical drugs act via a p53-dependent mechanism of action. An attractive strategy to circumvent this resistance would be to identify new anticancer drugs that act via p53-independent mode of action. In the present study, we identified 9 Ru (II)-Arene Schiff-base (RAS) complexes able to induce p53-independent cytotoxicity and discuss structural features that are required for their p53-independent activity. Increasing hydrophobicity led to an increase in cellular accumulation in cells with a corresponding increase in efficacy. We further showed that all nine complexes demonstrated p53-independent activity. This was despite significant differences in their physicochemical properties, suggesting that the iminoquinoline ligand, a common structural feature for all the complexes, is required for the p53-independent activity.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| | - Maria V Babak
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Daniel Yuan Qiang Wong
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Giorgia Pastorin
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore.,Department of Pharmacy, National University of Singapore , 18 Science Drive 4, 117543 Singapore
| | - Christian Gaiddon
- U1113 INSERM, 3 Avenue Molière, Strasbourg 67200, France.,Oncology Section, FMTS, Université de Strasbourg , F-67081 Strasbourg, France
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| |
Collapse
|
8
|
Kachalaki S, Ebrahimi M, Mohamed Khosroshahi L, Mohammadinejad S, Baradaran B. Cancer chemoresistance; biochemical and molecular aspects: a brief overview. Eur J Pharm Sci 2016; 89:20-30. [PMID: 27094906 DOI: 10.1016/j.ejps.2016.03.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/02/2016] [Accepted: 03/31/2016] [Indexed: 01/27/2023]
Abstract
The effectiveness of chemotherapy is one of the main challenges in cancer treatment and resistance to classic drugs and traditional treatment processes is an obstacle to this goal. Drug resistance that may be inherent or adventitious can cause poor treatment outcome and tumor relapse. In most cases, resistance to a drug can lead to resistance to many other drugs structure and function of which is not necessarily similar to the first drug. This phenomenon is the main mechanism behind failure of many of metastatic cancers. There are various molecular mechanisms involved in multidrug resistance, including change in the activity of membrane transporters (such as ABC transporters), increase of drug metabolism, change of the target enzyme (such as mutations that change thymidylate synthase and topoisomerases), promotion of DNA damage repair, and escape from drug induced apoptosis. Clinical and laboratory investigations on biomarkers involved in the response to chemotherapy have characterized the key factors behind the failure of treatments. Knowing the molecular factors involved in drug resistance may help us to develop new strategies for more promising chemotherapy and reduce the rate of relapse. In this brief review, molecular mechanisms and tumor microenvironment leading to decreased drug sensitivity, and strategies of reversing drug resistance are described.
Collapse
Affiliation(s)
- Saeed Kachalaki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mina Ebrahimi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Sina Mohammadinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Vasilevskaya IA, Selvakumaran M, Hierro LC, Goldstein SR, Winkler JD, O'Dwyer PJ. Inhibition of JNK Sensitizes Hypoxic Colon Cancer Cells to DNA-Damaging Agents. Clin Cancer Res 2015; 21:4143-52. [PMID: 26023085 DOI: 10.1158/1078-0432.ccr-15-0352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/12/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE We showed previously that in HT29 colon cancer cells, modulation of hypoxia-induced stress signaling affects oxaliplatin cytotoxicity. To further study the significance of hypoxia-induced signaling through JNK, we set out to investigate how modulation of kinase activities influences cellular responses of hypoxic colon cancer cells to cytotoxic drugs. EXPERIMENTAL DESIGN In a panel of cell lines, we investigated effects of pharmacologic and molecular inhibition of JNK on sensitivity to oxaliplatin, SN-38, and 5-FU. Combination studies for the drugs and JNK inhibitor CC-401 were carried out in vitro and in vivo. RESULTS Hypoxia-induced JNK activation was associated with resistance to oxaliplatin. CC-401 in combination with chemotherapy demonstrates synergism in colon cancer cell lines, although synergy is not always hypoxia specific. A more detailed analysis focused on HT29 and SW620 (responsive), and HCT116 (nonresponsive) lines. In HT29 and SW620 cells, CC-401 treatment results in greater DNA damage in the sensitive cells. In vivo, potentiation of bevacizumab, oxaliplatin, and the combination by JNK inhibition was confirmed in HT29-derived mouse xenografts, in which tumor growth delay was greater in the presence of CC-401. Finally, stable introduction of a dominant negative JNK1, but not JNK2, construct into HT29 cells rendered them more sensitive to oxaliplatin under hypoxia, suggesting differing input of JNK isoforms in cellular responses to chemotherapy. CONCLUSIONS These findings demonstrate that signaling through JNK is a determinant of response to therapy in colon cancer models, and support the testing of JNK inhibition to sensitize colon tumors in the clinic.
Collapse
Affiliation(s)
| | - Muthu Selvakumaran
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lucia Cabal Hierro
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara R Goldstein
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey D Winkler
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter J O'Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Sillars-Hardebol AH, Carvalho B, Beliën JAM, de Wit M, Delis-van Diemen PM, Tijssen M, van de Wiel MA, Pontén F, Fijneman RJA, Meijer GA. BCL2L1has a functional role in colorectal cancer and its protein expression is associated with chromosome 20q gain. J Pathol 2012; 226:442-450. [DOI: 10.1002/path.2983] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
11
|
Meng RD, Shelton CC, Li YM, Qin LX, Paty PB, Schwartz GK. gamma-Secretase inhibitors abrogate oxaliplatin-induced activation of the Notch-1 signaling pathway in colon cancer cells resulting in enhanced chemosensitivity. Cancer Res 2009; 69:573-82. [PMID: 19147571 PMCID: PMC3242515 DOI: 10.1158/0008-5472.can-08-2088] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Because Notch signaling is implicated in colon cancer tumorigenesis and protects cells from apoptosis by inducing prosurvival targets, it was hypothesized that inhibition of Notch signaling with gamma-secretase inhibitors (GSI) may enhance the chemosensitivity of colon cancer cells. We first show that the Notch-1 receptor, as well as its downstream target Hes-1, is up-regulated with colon cancer progression, similar to other genes involved in chemoresistance. We then report that chemotherapy induces Notch-1, as oxaliplatin, 5-fluorouracil (5-FU), or SN-38 (the active metabolite of irinotecan) induced Notch-1 intracellular domain (NICD) protein and activated Hes-1. Induction of NICD by oxaliplatin was caused by an increase in the activity and expression of gamma-secretase complex, as suppression of the protein subunit nicastrin with small interfering RNA (siRNA) prevented NICD induction after oxaliplatin. Subsequent inhibition of Notch-1 signaling with a sulfonamide GSI (GSI34) prevented the induction of NICD by chemotherapy and blunted Hes-1 activation. Blocking the activation of Notch signaling with GSI34 sensitized cells to chemotherapy and was synergistic with oxaliplatin, 5-FU, and SN-38. This chemosensitization was mediated by Notch-1, as inhibition of Notch-1 with siRNA enhanced chemosensitivity whereas overexpression of NICD increased chemoresistance. Down-regulation of Notch signaling also prevented the induction of prosurvival pathways, most notably phosphoinositide kinase-3/Akt, after oxaliplatin. In summary, colon cancer cells may up-regulate Notch-1 as a protective mechanism in response to chemotherapy. Therefore, combining GSIs with chemotherapy may represent a novel approach for treating metastatic colon cancers by mitigating the development of chemoresistance.
Collapse
Affiliation(s)
- Raymond D. Meng
- Laboratory of New Drug Development, Division of Solid Tumor Oncology, Department of Medicine, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| | - Christopher C. Shelton
- Molecular Pharmacology and Chemistry Program, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| | - Philip B. Paty
- Colorectal Service, Department of Surgery, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| | - Gary K. Schwartz
- Laboratory of New Drug Development, Division of Solid Tumor Oncology, Department of Medicine, New York, New York
- Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York
| |
Collapse
|
12
|
HEFFETER P, JUNGWIRTH U, JAKUPEC M, HARTINGER C, GALANSKI M, ELBLING L, MICKSCHE M, KEPPLER B, BERGER W. Resistance against novel anticancer metal compounds: Differences and similarities. Drug Resist Updat 2008; 11:1-16. [DOI: 10.1016/j.drup.2008.02.002] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 02/14/2008] [Accepted: 02/15/2008] [Indexed: 11/26/2022]
|
13
|
Xie YE, Tang EJ, Zhang DR, Ren BX. Down-regulation of Bcl-X L by RNA interference suppresses cell growth and induces apoptosis in human esophageal cancer cells. World J Gastroenterol 2006; 12:7472-7. [PMID: 17167836 PMCID: PMC4087593 DOI: 10.3748/wjg.v12.i46.7472] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the inhibitory effect of the vector-generated small interfering RNAs (siRNAs) on the expression of the Bcl-XL gene in established human esophageal cancer cells, and to investigate the effect of the Bcl-XL siRNAs on cell growth and apoptosis in esophageal cancer cells.
METHODS: Three siRNA-expressing vectors targeting different sites of the Bcl-XL gene were constructed from pTZ-U6+1 vector. Cultured esophageal cancer cells were transfected with the siRNA-expressing vector (or the control vector) using lipofectamine 2000. Bcl-XL gene expression was determined with semiquantitative RT-PCR assay and Western blotting. Among the three siRNA-expressing vectors, the most highly functional vector and its effect on cell growth and apoptosis in esophageal cancer cells was further analyzed.
RESULTS: Of the three siRNA-expressing vectors, siRNA-expressing vector No.1 was the most potent one which suppressed Bcl-XL mRNA production to 32.5% of that in the untreated esophageal cancer cells. Western blotting analysis showed that siRNA-expressing vector No.1 markedly down-regulated the expression of Bcl-XL in human esophageal cancer cells. Treatment of esophageal cancer cells with siRNA-expressing vector No.1 resulted in inhibition of cell growth and induction of apoptosis.
CONCLUSION: Down-regulation of Bcl-XL by vector-generated small interfering RNAs can suppress cell growth and induce apoptosis in human esophageal cancer cells.
Collapse
Affiliation(s)
- Yong-En Xie
- The Institute of Immunology and Molecular Biology, North Sichuan Medical College, No.234, Fujiang Road, Nanchong 637007, Sichuan Province, China.
| | | | | | | |
Collapse
|
14
|
Hata T, Yamamoto H, Ngan CY, Koi M, Takagi A, Damdinsuren B, Yasui M, Fujie Y, Matsuzaki T, Hemmi H, Xu X, Kitani K, Seki Y, Takemasa I, Ikeda M, Sekimoto M, Matsuura N, Monden M. Role of p21waf1/cip1 in effects of oxaliplatin in colorectal cancer cells. Mol Cancer Ther 2006; 4:1585-94. [PMID: 16227409 DOI: 10.1158/1535-7163.mct-05-0011] [Citation(s) in RCA: 34] [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
Clinical studies have shown that oxaliplatin, a novel platinum derivative, is a potent chemotherapeutic agent for colorectal cancer when combined with 5-fluorouracil and leucovorin. Although the toxic activity is based on covalent adducts between platinum and DNA, its actual biological behavior is mostly unknown. In an effort to explore the mechanism of tumor susceptibility to oxaliplatin, we examined the cytotoxic effects of oxaliplatin in colorectal cancer cell lines in reference to p53 gene status. Although p53 gene status did not clearly predict sensitivity to oxaliplatin, p53 wild-type cells including HCT116 were sensitive but HCT116 p53-/- were found to be resistant to oxaliplatin. Oxaliplatin caused strong p21waf1/cip1 induction and G0-G1 arrest in p53 wild-type cells, whereas cisplatin did not induce G0-G1 arrest. Assays using p53 wild but p21waf1/cip1 null HCT116 cells revealed that oxaliplatin did not show G0-G1 arrest and reduced growth-inhibitory effects, suggesting that p21waf1/cip1 may be a key element in oxaliplatin-treated p53 wild-type cells. Although HCT116 is DNA mismatch repair-deficient, a mismatch repair-proficient HCT116+ch3 cell line displayed similar responses with regard to p21waf1/cip1-mediated growth inhibition and G0-G1 arrest. In p53 mutant cells, on the other hand, oxaliplatin caused an abrupt transition from G1 to S phase and eventually resulted in G2-M arrest. This abrupt entry into S phase was associated with loss of the p21waf1/cip1 protein via proteasome-mediated degradation. These findings suggest that p21waf1/cip1 plays a role in oxaliplatin-mediated cell cycle and growth control in p53-dependent and -independent pathways.
Collapse
Affiliation(s)
- Taishi Hata
- Department of Surgery and Clinical Oncology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|