1
|
miR-874-3p mitigates cisplatin resistance through modulating NF-κB/inhibitor of apoptosis protein signaling pathway in epithelial ovarian cancer cells. Mol Cell Biochem 2021; 477:307-317. [PMID: 34716858 DOI: 10.1007/s11010-021-04271-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
The resistance to cisplatin, the most common platinum chemotherapy drug, may confine the efficacy of treatment in epithelial ovarian cancer patients. Aberrant expression of inhibitor of apoptosis proteins set the stage for resistance to cisplatin in EOC; besides, chemosensitivity in EOC can be chalked up to dysregulation of specific miRNAs. Herein, we investigated whether there is a potential correlation between miR-874-3p and the X-chromosome-linked inhibitor of apoptosis, a member of the IAP protein family in cisplatin-resistant EOC cells. The lower expression of miR-874-3p was found in SKOV3-DDP cells; it was also in association with cisplatin-resistance in EOC cells. XIAP was found to contribute to developing platinum resistance and is an authentic target for miR-874-3p in SKOV3-DDP cells. Consistently, restoration of miR-874-3p expression reversed cisplatin resistance in such cells by modulating XIAP and NF-κB/Survivin signaling pathway. Besides, siRNA knock down of XIAP in SKOV3-DDP cells had an anti-migratory effect like those with miR-874 overexpression. Importantly, the enforced expression of XIAP rescued SKOV3-DDP cells from the cytotoxic effects of miR-874-3p. Finally, miR-874-3p sensitized EOC cells to cisplatin-induced apoptosis, at least in part, through targeting XIAP. The cytotoxic effects of miR-874-3p can be attributed to the targeting XIAP in cisplatin-resistant EOC cells. We believe that the combination of cisplatin with miR-874-3p may make a potential strategy to reverse cisplatin resistance.
Collapse
|
2
|
Chen M, Yang S, Wu Y, Zhao Z, Zhai X, Dong D. High temperature requirement A1 in cancer: biomarker and therapeutic target. Cancer Cell Int 2021; 21:513. [PMID: 34563186 PMCID: PMC8466973 DOI: 10.1186/s12935-021-02203-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
As the life expectancy of the population increases worldwide, cancer is becoming a substantial public health problem. Considering its recurrence and mortality rates, most cancer cases are difficult to cure. In recent decades, a large number of studies have been carried out on different cancer types; unfortunately, tumor incidence and mortality have not been effectively improved. At present, early diagnostic biomarkers and accurate therapeutic strategies for cancer are lacking. High temperature requirement A1 (HtrA1) is a trypsin-fold serine protease that is also a chymotrypsin-like protease family member originally discovered in bacteria and later discovered in mammalian systems. HtrA1 gene expression is decreased in diverse cancers, and it may play a role as a tumor suppressor for promoting the death of tumor cells. This work aimed to examine the role of HtrA1 as a cell type-specific diagnostic biomarker or as an internal and external regulatory factor of diverse cancers. The findings of this study will facilitate the development of HtrA1 as a therapeutic target.
Collapse
Affiliation(s)
- Mingming Chen
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China.,Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China
| | - Yu Wu
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China.,Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Zirui Zhao
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China.,Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China.
| | - Deshi Dong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, 222, Zhongshan Road, Xigang District, 116011, Dalian, China.
| |
Collapse
|
3
|
Montazeri V, Ghahremani MH, Montazeri H, Hasanzad M, Safavi DM, Ayati M, Chehrazi M, Arefi Moghaddam B, Ostad SN. A Preliminary Study of NER and MMR Pathways Involved in Chemotherapy Response in Bladder Transitional Cell Carcinoma: Impact on progression-free survival. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:355-365. [PMID: 32922493 PMCID: PMC7462481 DOI: 10.22037/ijpr.2020.112646.13878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
One of the main genotoxic drugs used in bladder cancer chemotherapy is cisplatin. While it is applied in most types of cancers, resistance to cisplatin is wildly common. In order to overcome drug resistance, it is necessary to determine a predictive marker. This study was conducted to provide basic data for selecting and designing a gene profile for further cohort and RCT studies in the future to improve response to treatment in bladder cancer. The expression levels of ERCC1, MLH1, MSH2, and CTR1 mRNA were determined in the tumor tissue using real-time q-PCR. Progression-free survival (PFS) was analyzed in term of the level of genes expression. The results revealed that the level of ERCC1 mRNA expression was higher in the recurrence (R) group compared to the no recurrence (NR) group. Moreover, the PFS time was increased in the patients with an ERCC1 expression level of below 1.57. The level of MLH1 and MSH2 mRNA expression was lower in the R group compared to the NR group; therefore, PFS time was increased in the patients with MLH1 and MSH2 gene expression levels above the cutoff point. While the level of CTR1 mRNA expression was higher in the R group versus the NR group, the PFS time was longer in the patients with CTR1 expression levels of below 1.265 compared to the patients with high levels of CTR1 expression. It can be concluded that the level of ERCC1, MLH1, MSH2, and CTR1 mRNA expression may be associated with PFS time as possible therapeutic targets for decreasing cisplatin resistance.
Collapse
Affiliation(s)
- Vahideh Montazeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - D Majid Safavi
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Ayati
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Chehrazi
- Department of Biostatistics and Epidemiology, School of Public Health, Babol University of Medical Sciences, Babol, Iran
| | | | - Seyed Nasser Ostad
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Wu G, Mai X, Liu F, Lin M, Dong X, Xu Q, Hao C, Zhang L, Yu R, Jiang T. Synthesis of novel 10,11-methylenedioxy-camptothecin glycoside derivatives and investigation of their anti-tumor effects in vivo. RSC Adv 2019; 9:11142-11150. [PMID: 35520228 PMCID: PMC9063016 DOI: 10.1039/c9ra00315k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/21/2019] [Indexed: 11/21/2022] Open
Abstract
10,11-Methylenedioxy-camptothecin (FL118) is a novel camptothecin analogue that possesses exceptional antitumor efficacy in human tumor xenograft models. The aim of the current study was to develop novel 20-substituted FL118 derivatives coupled with glycosyl-succinic acid esters with improved antitumor efficacy. These FL118 glycoside derivatives were designed, synthesized and their cytotoxicity evaluated in three tumor cell lines (A-549, MDA-MB-231 and RM-1). All of the derivatives showed superior in vitro cytotoxic activity and were more potent than irinotecan in A549 and MDA-MB-231 cells. In mouse prostate cancer cells RM-1, 10,11-methylenedioxy-camptothecin rhamnoside 11b displayed significant activities with IC50 of 48.27 nM. Western blot analysis demonstrated that 11b inhibited survivin expression and induced cancer cells apoptosis. Further cell cycle analyses clearly showed 11b induced G2/M phase cell cycle arrest. Molecule docking studies suggested that the binding mode of 11b was different from that of the crystal complex of ligand topotecan in Top1/DNA. Importantly, 11b showed high in vivo antitumor efficacy in the RM-1 mouse model with transplantation of prostate cancer (TGI = 44.9%) at dose of 9 mg kg−1 without apparent toxicity. In a RM-1 xenograft model, 11b had superior in vivo antitumor efficacy (TGI = 44.9%) at a dose of 9 mg kg−1.![]()
Collapse
|
5
|
Li Y, Zhang S, Wang Y, Peng J, Fang F, Yang X. MLH1 enhances the sensitivity of human endometrial carcinoma cells to cisplatin by activating the MLH1/c-Abl apoptosis signaling pathway. BMC Cancer 2018; 18:1294. [PMID: 30594176 PMCID: PMC6311060 DOI: 10.1186/s12885-018-5218-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 12/13/2018] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND MLH1 plays a critical role in maintaining the fidelity of DNA replication, and defects in human MLH1 have been reported. However, the role of MLH1 in endometrial carcinoma has not been fully investigated. Therefore, we aimed to study the role of MLH1 in the sensitivity of human endometrial carcinoma cells to cisplatin. METHODS In this study, we detected the expression of MLH1 in Ishikawa and RL95-2 cells. MLH1-siRNA and ADV-MLH1 were adopted for the silencing and overexpression of MLH1, respectively. Real-time polymerase chain reaction, Western blotting, cell proliferation assays, and cell cycle and apoptotic analyses by flow cytometry were employed to explore the underlying mechanism. A mouse xenograft model was used to investigate the effect of MLH1 on tumor growth after treatment with cisplatin. RESULTS Over-expression of MLH1 in Ishikawa cells dramatically increased the sensitivity of cells to cisplatin and enhanced cell apoptosis. By contrast, knockdown of MLH1 yielded the opposite effects in vitro. Mechanistically, cisplatin induced the MLH1/c-Abl apoptosis signaling pathway in ADV-MLH1-infected endometrial carcinoma cells, and these effects involved c-Abl, caspase-9, caspase-3 and PARP. Altogether, our results indicate that ADV-MLH1 might attenuate Ishikawa cell growth in vivo, resulting in increased cisplatin sensitivity. CONCLUSIONS MLH1 may render endometrial carcinoma cells more sensitive to cisplatin by activating the MLH1/c-Abl apoptosis signaling pathway. In addition, an applicable adenovirus vector (ADV-MLH1) for MLH1 overexpression in endometrial carcinoma was generated. Thus, ADV-MLH1 might be a novel potential therapeutic target for endometrial carcinoma.
Collapse
Affiliation(s)
- Yue Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, No. 107, Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China.,Department of Obstetrics and Gynecology, Weihai Municipal Hospital, No. 70, Heping Road, WeiHai, Shandong, 264200, People's Republic of China
| | - Shihong Zhang
- Department of Obstetrics and Gynecology, Weihai Municipal Hospital, No. 70, Heping Road, WeiHai, Shandong, 264200, People's Republic of China
| | - Yuanjian Wang
- Huaxi Clinical Medical College of Sichuan University, Jiang'an campus, Chengdu City, Sichuan, 610207, People's Republic of China
| | - Jin Peng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, No. 107, Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Fang Fang
- Department of Obstetrics and Gynecology, Weihai Municipal Hospital, No. 70, Heping Road, WeiHai, Shandong, 264200, People's Republic of China
| | - Xingsheng Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, No. 107, Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
6
|
A Whole-genome CRISPR Screen Identifies a Role of MSH2 in Cisplatin-mediated Cell Death in Muscle-invasive Bladder Cancer. Eur Urol 2018; 75:242-250. [PMID: 30414698 DOI: 10.1016/j.eururo.2018.10.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/15/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The response to first-line, platinum-based treatment of muscle-invasive bladder cancer has not improved in 3 decades. OBJECTIVE To identify genes that influence cisplatin resistance in bladder cancer. DESIGN, SETTING, AND PARTICIPANTS We performed a whole-genome CRISPR screen in a bladder cancer cell line to identify genes that mediate resistance to cisplatin. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Targeted validation was performed in two bladder cancer cell lines. The top gene candidate was validated in a publicly available bladder cancer dataset. RESULTS AND LIMITATIONS From the CRISPR screen, we identified MSH2 as the most significantly enriched gene and mismatch repair as the most significantly enriched pathway that promoted resistance to cisplatin. Bladder cancer cells with knockdown of MSH2 showed a reduction in cisplatin-mediated apoptosis. MSH2 loss did not impact the sensitivity to other chemotherapies, including the cisplatin analog oxaliplatin. Bladder tumors with low MSH2 protein levels, quantified using reverse-phase protein array, showed poorer survival when treated with cisplatin- or carboplatin-based therapy; these results require future validation using immunohistochemistry. Additionally, results are retrospective from patients with primarily high-grade tumors; thus, validation in a controlled clinical trial is needed. CONCLUSIONS We generated in vitro evidence that bladder cancer cell lines depleted of MSH2 are more resistant to cisplatin. We additionally found an association between low MSH2 in bladder tumors and poorer patient survival when treated with platinum-based chemotherapy. If successfully validated prospectively, MSH2 protein level could assist in the selection of patients for chemotherapy. PATIENT SUMMARY We report the first evidence that MSH2 protein level may contribute to chemotherapy resistance observed in muscle-invasive bladder cancer. MSH2 has potential as a biomarker predictive of response to platinum-based therapy.
Collapse
|
7
|
Güçlü H, Doganlar ZB, Gürlü VP, Özal A, Dogan A, Turhan MA, Doganlar O. Effects of cisplatin-5-fluorouracil combination therapy on oxidative stress, DNA damage, mitochondrial apoptosis, and death receptor signalling in retinal pigment epithelium cells. Cutan Ocul Toxicol 2018; 37:291-304. [PMID: 29606027 DOI: 10.1080/15569527.2018.1456548] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
AIM Combination therapies of cisplatin with 5-FU (PF) are an effective solution and have been widely used for the treatment of various categories of cancer including anal, gastrointestinal, and oral cancer, as well as head and neck tumors. The effects of combined PF treatment on vital intracellular signalling pathways in nontargeted cells remain unclear. The aim of this study is to explain the possible mechanisms by which combined PF treatment results in retinal toxicity and to investigate the effects of PF on important vital signalling pathways in ARPE 19 retinal pigmented epithelial cells. MATERIALS AND METHODS We analysed the cellular and molecular effects of PF on cell viability, oxidative stress, gene repair response, and induction of apoptosis in ARPE 19 cells using molecular probe fluorescent staining, cell cytometer, RAPD, qRT-PCR, and western blot assays. RESULTS We determined that PF causes excessive generation of reactive oxygen species (ROS) and prevents ROS scavenging by suppressing antioxidant systems. We found induction of DNA damage, particularly mismatch and double strand break repair, in ARPE 19 cells treated with PF. In this study, PF also induced both the intrinsic apoptosis pathway and death receptor signalling in ARPE 19 cells. CONCLUSIONS Our data proved that PF causes cytotoxicity and genotoxicity, at both the cellular and molecular levels, in ARPE 19 cells following particularly prolonged treatment (48 h). Additionally, our results suggest key molecular signals for prevention strategies that can be developed to reduce the severe side effects of PF chemotherapy.
Collapse
Affiliation(s)
- Hande Güçlü
- a Department of Ophthalmology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Zeynep Banu Doganlar
- b Department of Medical Biology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Vuslat Pelitli Gürlü
- a Department of Ophthalmology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Altan Özal
- a Department of Ophthalmology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Ayten Dogan
- b Department of Medical Biology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Meryem Aysenur Turhan
- b Department of Medical Biology, Faculty of Medicine , Trakya University , Edirne , Turkey
| | - Oguzhan Doganlar
- b Department of Medical Biology, Faculty of Medicine , Trakya University , Edirne , Turkey
| |
Collapse
|
8
|
Zhao C, Li S, Zhao M, Zhu H, Zhu X. Prognostic values of DNA mismatch repair genes in ovarian cancer patients treated with platinum-based chemotherapy. Arch Gynecol Obstet 2018; 297:153-159. [PMID: 29063235 PMCID: PMC5762798 DOI: 10.1007/s00404-017-4563-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 10/11/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE DNA mismatch repair (MMR) is a highly conserved biological pathway that plays a key role in maintaining genomic stability. MMR has been reported as a prognostic marker in certain cancers; however, the results are controversial. Therefore, identification of the prognostic value of MMR genes in ovarian cancer based on a large sample size is pivotal. METHODS In the current study, we systemically investigated the prognostic roles of seven MMR genes, MSH2, MSH3, MSH6, MLH1, MLH3, PMS1 and PMS2, in ovarian cancer patients treated with platinum-based chemotherapy through "The Kaplan-Meier plotter" (KM plotter) database, which contains gene expression data and survival information of ovarian cancer patients. RESULTS Among seven MMR genes, high mRNA levels of MSH6, MLH1 and PMS2 were significantly associated with a better overall survival for all ovarian cancer patients treated with platinum-based chemotherapy, especially in late-stage and poor-differentiated ovarian cancer patients. Increased MSH6 and PMS2 mRNA expression was correlated with a favorable overall survival in serous ovarian cancer patients. CONCLUSIONS Our results indicate that sufficient MMR system is associated with an improved survival in ovarian cancer treated with platinum-based chemotherapy. MMR gene may be a potential prognosis predictor in ovarian cancer.
Collapse
Affiliation(s)
- Chuchu Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Saisai Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Menghuang Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China
| | - Haiyan Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
9
|
Fukuhara S, Chang I, Mitsui Y, Chiyomaru T, Yamamura S, Majid S, Saini S, Hirata H, Deng G, Gill A, Wong DK, Shiina H, Nonomura N, Dahiya R, Tanaka Y. DNA mismatch repair gene MLH1 induces apoptosis in prostate cancer cells. Oncotarget 2015; 5:11297-307. [PMID: 25526032 PMCID: PMC4294331 DOI: 10.18632/oncotarget.2315] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/05/2014] [Indexed: 01/13/2023] Open
Abstract
Mismatch repair (MMR) enzymes have been shown to be deficient in prostate cancer (PCa). MMR can influence the regulation of tumor development in various cancers but their role on PCa has not been investigated. The aim of the present study was to determine the functional effects of the mutL-homolog 1 (MLH1) gene on growth of PCa cells. The DU145 cell line has been established as MLH1-deficient and thus, this cell line was utilized to determine effects of MLH1 by gene expression. Lack of MLH1 protein expression was confirmed by Western blotting in DU145 cells whereas levels were high in normal PWR-1E and RWPE-1 prostatic cells. MLH1-expressing stable transfectant DU145 cells were then created to characterize the effects this MMR gene has on various growth properties. Expression of MLH1 resulted in decreased cell proliferation, migration and invasion properties. Lack of cell growth in vivo also indicated a tumor suppressive effect by MLH1. Interestingly, MLH1 caused an increase in apoptosis along with phosphorylated c-Abl, and treatment with MLH1 siRNAs countered this effect. Furthermore, inhibition of c-Abl with STI571 also abrogated the effect on apoptosis caused by MLH1. These results demonstrate MLH1 protects against PCa development by inducing c-Abl-mediated apoptosis.
Collapse
Affiliation(s)
- Shinichiro Fukuhara
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America. Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Inik Chang
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Oral Biology,Yonsei University College of Dentistry, Seoul, Korea
| | - Yozo Mitsui
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America. Department of Urology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Takeshi Chiyomaru
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America. Department of Urology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Soichiro Yamamura
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Shahana Majid
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Sharanjot Saini
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Hiroshi Hirata
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Guoren Deng
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Ankurpreet Gill
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America
| | - Darryn K Wong
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America
| | - Hiroaki Shiina
- Department of Urology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Rajvir Dahiya
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| | - Yuichiro Tanaka
- Department of Surgery/Urology, Veterans Affairs Medical Center, San Francisco, California, United States of America. Department of Urology, University of California, San Francisco, California, United States of America
| |
Collapse
|
10
|
Zhang ST, Zuo C, Li WN, Fu XQ, Xing S, Zhang XP. Identification of key genes associated with the effect of estrogen on ovarian cancer using microarray analysis. Arch Gynecol Obstet 2015; 293:421-7. [PMID: 26264810 DOI: 10.1007/s00404-015-3833-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/27/2015] [Indexed: 01/15/2023]
Abstract
PURPOSE To identify key genes related to the effect of estrogen on ovarian cancer. METHODS Microarray data (GSE22600) were downloaded from Gene Expression Omnibus. Eight estrogen and seven placebo treatment samples were obtained using a 2 × 2 factorial designs, which contained 2 cell lines (PEO4 and 2008) and 2 treatments (estrogen and placebo). Differentially expressed genes were identified by Bayesian methods, and the genes with P < 0.05 and |log2FC (fold change)| ≥0.5 were chosen as cut-off criterion. Differentially co-expressed genes (DCGs) and differentially regulated genes (DRGs) were, respectively, identified by DCe function and DRsort function in DCGL package. Topological structure analysis was performed on the important transcriptional factors (TFs) and genes in transcriptional regulatory network using tYNA. Functional enrichment analysis was, respectively, performed for DEGs and the important genes using Gene Ontology and KEGG databases. RESULTS In total, 465 DEGs were identified. Functional enrichment analysis of DEGs indicated that ACVR2B, LTBP1, BMP7 and MYC involved in TGF-beta signaling pathway. The 2285 DCG pairs and 357 DRGs were identified. Topological structure analysis showed that 52 important TFs and 65 important genes were identified. Functional enrichment analysis of the important genes showed that TP53 and MLH1 participated in DNA damage response and the genes (ACVR2B, LTBP1, BMP7 and MYC) involved in TGF-beta signaling pathway. CONCLUSION TP53, MLH1, ACVR2B, LTBP1 and BMP7 might participate in the pathogenesis of ovarian cancer.
Collapse
Affiliation(s)
- Shi-tao Zhang
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, Jilin University, Changchun, 130012, China
| | - Chao Zuo
- Department of Anesthesiology, The Fifth Affiliated Hospital of Zunyi Medical College, Zhu Hai, 519100, China
| | - Wan-nan Li
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, Jilin University, Changchun, 130012, China
| | - Xue-qi Fu
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, Jilin University, Changchun, 130012, China
| | - Shu Xing
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, Jilin University, Changchun, 130012, China
| | - Xiao-ping Zhang
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China.
| |
Collapse
|
11
|
Zhao J, Ling X, Cao S, Liu X, Wan S, Jiang T, Li F. Antitumor activity of FL118, a survivin, Mcl-1, XIAP, and cIAP2 selective inhibitor, is highly dependent on its primary structure and steric configuration. Mol Pharm 2014; 11:457-67. [PMID: 24329001 DOI: 10.1021/mp4004282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We recently reported the identification and characterization of a novel small chemical molecule designated FL118. FL118 selectively inhibits multiple cancer survival and proliferation-associated antiapoptotic proteins (survivin, Mcl-1, XIAP, cIAP2) and eliminates small and large human tumor xenografts in animal models (Ling et al., PLoS One 2012, 7, e45571). Here, we report a follow-up study on the structure-activity relationship (SAR) of the hydroxyl group in the lactone ring of FL118. We found that the superior antitumor efficacy of FL118 heavily depends on its steric configuration through comparing the antitumor activity of FL118 with FL113 (the racemic mixture of FL118). Consistently, FL118 proved much more effective in inhibiting the expression of survivin, Mcl-1, and cIAP2, both in vitro and in vivo, compared to FL113. Additionally, Tet-on controlled induction of survivin or forced expression of Mcl-1 protects cancer cells from FL118-mediated growth inhibition and cell death. To further explore the SAR, we synthesized seven position 20-esterifiable FL118 and FL113 derivatives. Studies on these seven new compounds revealed that keeping a free hydroxyl group of FL118 is also important for high antitumor efficacy. Together, these studies confirm the superior anticancer activity of FL118 and narrow the window for further SAR studies to generate novel analogues based on FL118 core structure on its other potential chemical positions.
Collapse
Affiliation(s)
- Jiuyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China , 5 Yushan Road, Qingdao, Shandong 266003 China
| | | | | | | | | | | | | |
Collapse
|
12
|
de Camargo EA, da Silva GN, Gobette CP, de Castro Marcondes JP, Salvadori DMF. No Relationship between the Amount of DNA Damage and the Level of hMLH1 and RASSF1A Gene Expression in Bladder Cancer Cells Treated with Cisplatin and Gemcitabine. Asian Pac J Cancer Prev 2013; 14:5941-8. [DOI: 10.7314/apjcp.2013.14.10.5941] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
13
|
MLH1 as a direct target of MiR-155 and a potential predictor of favorable prognosis in pancreatic cancer. J Gastrointest Surg 2013; 17:1399-405. [PMID: 23715647 DOI: 10.1007/s11605-013-2230-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 05/07/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND The regulation of Mut L homologue 1 (MLH1) expression by microRNA (miR)-155 and its prognostic significance in pancreatic cancer (PC) remain to be elucidated. This study aimed to address the issues. METHODS MiR-155 mimics and inhibitor were transfected to PC cell lines, Panc-1 and Capan-1. Expression of MLH1 was subsequently evaluated. Then, luciferase activity was detected after miR-155 mimics and pRL-TK plasmids containing wild-type and mutant 3'UTRs of MLH1 mRNA were co-transfected. Finally, immunohistochemical staining for MLH1 was performed in PC samples. RESULTS Transfection of miR-155 mimics and inhibitor led to reversely altered protein expressions of miR-155 and MLH1, whereas the corresponding mRNA expressions were similar. A significant decrease in luciferase activity in the cells transfected with the wild-type pRL-TK plasmid was shown in contrast to those transfected with the mutant one. In addition, MLH1 was less expressed in tumor than in para-tumor tissues of PC. Extensive MLH1 expression was significantly associated with favorable differentiation and less lymph node metastasis. MLH1 expression was found to be a prognosticator in univariate analysis, and being of marginally significant impact in multivariate test. CONCLUSIONS MLH1 might serve as a direct target of miR-155 and a potential prognosis predictor in PC.
Collapse
|
14
|
Martinez-Becerra P, Vaquero J, Romero MR, Lozano E, Anadon C, Macias RIR, Serrano MA, Grañé-Boladeras N, Muñoz-Bellvis L, Alvarez L, Sangro B, Pastor-Anglada M, Marin JJG. No correlation between the expression of FXR and genes involved in multidrug resistance phenotype of primary liver tumors. Mol Pharm 2012; 9:1693-704. [PMID: 22524153 DOI: 10.1021/mp300028a] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Farnesoid X receptor (FXR) has been recently reported to enhance chemoresistance through bile acid-independent mechanisms. Thus, FXR transfection plus activation with GW4064 resulted in reduced sensitivity to cisplatin-induced toxicity. This is interesting because primary tumors of the liver, an organ where FXR is expressed, exhibit marked refractoriness to pharmacological treatment. Here we have determined whether FXR is upregulated in hepatocellular carcinoma (HCC), cholangiocarcinoma (CGC) and hepatoblastoma (HPB) and whether this is related with the expression of genes involved in mechanisms of chemoresistance. Using RT-QPCR and Taqman low density arrays we have analyzed biopsies from healthy livers or surgically removed tumors from naive patients and cell lines derived from HCC (SK-HEP-1, Alexander and Huh7), CGC (TFK1) and HPB (HepG2), before and after exposure to cisplatin at IC50 for 72 h. In liver tumors FXR expression was not enhanced but significantly decreased (healthy liver > HCC > HPB ≈ CGC). Except for CGC, this was not accompanied by changes in the proportions of FXR isoforms. Changes in 36 genes involved in drug uptake/efflux and metabolism, expression/function of molecular targets, and survival/apoptosis balance were found. Changes affecting SLC22A1, CYP2A1 and BIRC5 were shared by HCC, CGC and HPB. Similarity in gene expression profiles between cell lines and parent tumors was found. Pharmacological challenge with cisplatin induced changes that increased this resemblance. This was not dependent upon FXR expression. Thus, although FXR may play a role in inducing chemoresistance under certain circumstances, its upregulation does not seem to be involved in the multidrug resistance phenotype characteristic of HCC, CGC and HPB.
Collapse
Affiliation(s)
- P Martinez-Becerra
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Suppression of Hepatoma Carcinoma Cell Bel-7404 by Small Molecule XIAP Inhibitor Embelin Through Blocking NF-κB Signaling Pathway*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2011.00300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
He X, Khurana A, Maguire JL, Chien J, Shridhar V. HtrA1 sensitizes ovarian cancer cells to cisplatin-induced cytotoxicity by targeting XIAP for degradation. Int J Cancer 2012; 130:1029-35. [PMID: 21387310 PMCID: PMC3206182 DOI: 10.1002/ijc.26044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 02/22/2011] [Indexed: 01/13/2023]
Abstract
HtrA1, a member of serine protease family, has been previously found to be involved in resistance to chemotherapy in ovarian cancer although the underlying mechanism is not clear. Using mixture-based oriented peptide library approach, previously we identified X-linked inhibitor of apoptosis protein (XIAP), a member of the inhibitor of apoptosis proteins family, as a potential substrate of HtrA1. The aim of our work is to investigate the link between HtrA1 and XIAP proteins and their relationships with chemoresistance in ovarian cancer. Our results showed that recombinant XIAP was degraded by purified wild-type HtrA1 but not mutant HtrA1 in vitro. Consistent with the in vitro data, coimmunoprecipitation assays showed that HtrA1 and XIAP formed a protein complex in vivo. Ectopic expression of HtrA1 led to decreased level of XIAP in OV167 and OV202 ovarian cancer cells, while knockdown of HtrA1 resulted in increased level of XIAP in SKOV3 ovarian cancer cells. Furthermore, overexpression of HtrA1 in OV202 cells promoted cell sensitivity to cisplatin-induced apoptosis that could be reversed by increased expression of XIAP. The cleavage of XIAP induced by HtrA1 was enhanced by cisplatin treatment. Taken together, our experiments have identified XIAP as a novel substrate of HtrA1 and the degradation of XIAP by HtrA1 contributes to cell response to chemotherapy, suggesting that restoring the expression of HtrA1 may be a promising treatment strategy for ovarian cancer.
Collapse
Affiliation(s)
- Xiaoping He
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
17
|
Loughery JEP, Dunne PD, O'Neill KM, Meehan RR, McDaid JR, Walsh CP. DNMT1 deficiency triggers mismatch repair defects in human cells through depletion of repair protein levels in a process involving the DNA damage response. Hum Mol Genet 2011; 20:3241-55. [PMID: 21636528 DOI: 10.1093/hmg/ddr236] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2025] Open
Abstract
DNA methyltransferase 1 (DNMT1) maintains methylation at CpG dinucleotides, important for transcriptional silencing at many loci. It is also implicated in stabilizing repeat sequences: DNMT1 deficiency causes microsatellite instability in mouse embryonic stem cells, but it is unclear how this occurs, how repeats lacking CpG become unstable and whether the effect is confined to stem cells. To address these questions, we transfected hTERT-immortalized normal human fibroblasts (hTERT-1604) with a short hairpin RNA construct targeting DNMT1 and isolated stable integrants with different levels of protein. DNMT1 expression levels agreed well with methylation levels at imprinted genes. Knockdown cells showed two key characteristics of mismatch repair (MMR) deficiency, namely resistance to the drug 6-thioguanine and up to 10-fold elevated mutation rates at a CA(17) microsatellite reporter, but had limited viability. The likely cause of MMR defects is a matching drop in steady-state protein levels for key repair components in DNMT1 knockdown cells, affecting both the MutLα and MutSα complexes. This indirect effect on MMR proteins was also seen using a different targeting method in HT29 colon cancer cells and did not involve transcriptional silencing of the respective genes. Decreased levels of MMR components follow activation of the DNA damage response and blocking this response, and in particular poly(ADP-ribose) polymerase (PARP) overactivation, rescues cell viability in DNMT1-depleted cells. These results offer an explanation for how and why unmethylated microsatellite repeats can be destabilized in cells with decreased DNMT1 levels and uncover a novel and important role for PARP in this process.
Collapse
Affiliation(s)
- Jayne E P Loughery
- Transcriptional Regulation and Epigenetics Group, Centre for Molecular Biosciences, University of Ulster, Coleraine BT52 1SA, UK
| | | | | | | | | | | |
Collapse
|
18
|
Muenyi CS, States VA, Masters JH, Fan TW, Helm CW, States JC. Sodium arsenite and hyperthermia modulate cisplatin-DNA damage responses and enhance platinum accumulation in murine metastatic ovarian cancer xenograft after hyperthermic intraperitoneal chemotherapy (HIPEC). J Ovarian Res 2011; 4:9. [PMID: 21696631 PMCID: PMC3143084 DOI: 10.1186/1757-2215-4-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 06/22/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is the leading cause of gynecologic cancer death in the USA. Recurrence rates are high after front-line therapy and most patients eventually die from platinum (Pt) - resistant disease. Cisplatin resistance is associated with increased nucleotide excision repair (NER), decreased mismatch repair (MMR) and decreased platinum uptake. The objective of this study is to investigate how a novel combination of sodium arsenite (NaAsO2) and hyperthermia (43°C) affect mechanisms of cisplatin resistance in ovarian cancer. METHODS We established a murine model of metastatic EOC by intraperitoneal injection of A2780/CP70 human ovarian cancer cells into nude mice. We developed a murine hyperthermic intraperitoneal chemotherapy model to treat the mice. Mice with peritoneal metastasis were perfused for 1 h with 3 mg/kg cisplatin ± 26 mg/kg NaAsO2 at 37 or 43°C. Tumors and tissues were collected at 0 and 24 h after treatment. RESULTS Western blot analysis of p53 and key NER proteins (ERCC1, XPC and XPA) and MMR protein (MSH2) suggested that cisplatin induced p53, XPC and XPA and suppressed MSH2 consistent with resistant phenotype. Hyperthermia suppressed cisplatin-induced XPC and prevented the induction of XPA by cisplatin, but it had no effect on Pt uptake or retention in tumors. NaAsO2 prevented XPC induction by cisplatin; it maintained higher levels of MSH2 in tumors and enhanced initial accumulation of Pt in tumors. Combined NaAsO2 and hyperthermia decreased cisplatin-induced XPC 24 h after perfusion, maintained higher levels of MSH2 in tumors and significantly increased initial accumulation of Pt in tumors. ERCC1 levels were generally low except for NaAsO2 co-treatment with cisplatin. Systemic Pt and arsenic accumulation for all treatment conditions were in the order: kidney > liver = spleen > heart > brain and liver > kidney = spleen > heart > brain respectively. Metal levels generally decreased in systemic tissues within 24 h after treatment. CONCLUSION NaAsO2 and/or hyperthermia have the potential to sensitize tumors to cisplatin by inhibiting NER, maintaining functional MMR and enhancing tumor platinum uptake.
Collapse
Affiliation(s)
- Clarisse S Muenyi
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
| | - Vanessa A States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
| | - Joshua H Masters
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
| | - Teresa W Fan
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40292, USA
- Center for Genetics & Molecular Medicine, University of Louisville, Louisville, KY 40292, USA
- Center for Environmental Genomics & Integrative Biology, University of Louisville, Louisville, KY 40292, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| | - C William Helm
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, St. Louis University School of Medicine, St Louis, MO 63117, USA
| | - J Christopher States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA
- Center for Genetics & Molecular Medicine, University of Louisville, Louisville, KY 40292, USA
- Center for Environmental Genomics & Integrative Biology, University of Louisville, Louisville, KY 40292, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
19
|
Basu A, Krishnamurthy S. Cellular responses to Cisplatin-induced DNA damage. J Nucleic Acids 2010; 2010:201367. [PMID: 20811617 PMCID: PMC2929606 DOI: 10.4061/2010/201367] [Citation(s) in RCA: 335] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 06/28/2010] [Indexed: 12/29/2022] Open
Abstract
Cisplatin is one of the most effective anticancer agents widely used in the treatment of solid tumors. It is generally considered as a cytotoxic drug which kills cancer cells by damaging DNA and inhibiting DNA synthesis. How cells respond to cisplatin-induced DNA damage plays a critical role in deciding cisplatin sensitivity. Cisplatin-induced DNA damage activates various signaling pathways to prevent or promote cell death. This paper summarizes our current understandings regarding the mechanisms by which cisplatin induces cell death and the bases of cisplatin resistance. We have discussed various steps, including the entry of cisplatin inside cells, DNA repair, drug detoxification, DNA damage response, and regulation of cisplatin-induced apoptosis by protein kinases. An understanding of how various signaling pathways regulate cisplatin-induced cell death should aid in the development of more effective therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Soumya Krishnamurthy
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|