1
|
Lou X, Zhou Q, Yin Y, Zhou C, Shen Y. Correction: Inhibition of the Met Receptor Tyrosine Kinase Signaling Enhances the Chemosensitivity of Glioma Cell Lines to CDDP Through Activation of p38 MAPK Pathway. Mol Cancer Ther 2025; 24:152. [PMID: 39692519 DOI: 10.1158/1535-7163.mct-24-0974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 12/19/2024]
|
2
|
Zhan Y, Chen Q, Song Y, Wei X, Zhao T, Chen B, Li C, Zhang W, Jiang Y, Tan Y, Du B, Xiao J, Wang K. Berbamine Hydrochloride inhibits lysosomal acidification by activating Nox2 to potentiate chemotherapy-induced apoptosis via the ROS-MAPK pathway in human lung carcinoma cells. Cell Biol Toxicol 2023; 39:1297-1317. [PMID: 36070022 DOI: 10.1007/s10565-022-09756-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/26/2022] [Indexed: 01/18/2023]
Abstract
Autophagy is typically activated in cancer cells as a rescue strategy in response to cellular stress (e.g., chemotherapy). Herein, we found that Berbamine Hydrochloride (Ber) can act as an effective inhibitor of the late stage of autophagic flux, thereby potentiating the killing effect of chemotherapy agents. Lung carcinoma cells exposed to Ber exhibited increased autophagosomes, marked by LC3-II upregulation. The increased level of p62 after Ber treatment indicated that the autophagic flux was blocked at the late stage. The lysosome staining assay and cathepsin maturation detection indicated impaired lysosomal acidification. We found that Nox2 exhibited intensified co-localization with lysosomes in Ber-treated cells. Nox2 is a key enzyme for superoxide anion production capable of transferring electrons into the lysosomal lumen, thereby neutralizing the inner protons; this might explain the aberrant acidification. This hypothesis is further supported by the observed reversal of lysosomal cathepsin maturation by Nox2 inhibitors. Finally, Ber combined with cisplatin exhibited a synergistic killing effect on lung carcinoma cells. Further data suggested that lung carcinoma cells co-treated with Ber and cisplatin accumulated excessive reactive oxygen species (ROS), which typically activated MAPK-mediated mitochondria-dependent apoptosis. The enhanced anti-cancer effect of Ber combined with cisplatin was also confirmed in an in vivo xenograft mouse model. These findings indicate that Ber might be a promising adjuvant for enhancing the cancer cell killing effect of chemotherapy via the inhibition of autophagy. In this process, Nox2 might be a significant mediator of Ber-induced aberrant lysosomal acidification.
Collapse
Affiliation(s)
- Yujuan Zhan
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qiugu Chen
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yue Song
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xianli Wei
- Department of Medical Instruments, Guangdong Food and Drug Vocational College, Guangzhou, 510520, People's Republic of China
| | - Tingxiu Zhao
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Chengxi Li
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wenbo Zhang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yanjun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Yuhui Tan
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Biaoyan Du
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jianyong Xiao
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Kun Wang
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Huang YK, Chang KC, Li CY, Lieu AS, Lin CL. AKR1B1 Represses Glioma Cell Proliferation through p38 MAPK-Mediated Bcl-2/BAX/Caspase-3 Apoptotic Signaling Pathways. Curr Issues Mol Biol 2023; 45:3391-3405. [PMID: 37185746 PMCID: PMC10136867 DOI: 10.3390/cimb45040222] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to investigate the regulatory role of Aldo-keto reductase family 1 member B1 (AKR1B1) in glioma cell proliferation through p38 MAPK activation to control Bcl-2/BAX/caspase-3 apoptosis signaling. AKR1B1 expression was quantified in normal human astrocytes, glioblastoma multiforme (GBM) cell lines, and normal tissues by using quantitative real-time polymerase chain reaction. The effects of AKR1B1 overexpression or knockdown and those of AKR1B1-induced p38 MAPK phosphorylation and a p38 MAPK inhibitor (SB203580) on glioma cell proliferation were determined using an MTT assay and Western blot, respectively. Furthermore, the AKR1B1 effect on BAX and Bcl-2 expression was examined in real-time by Western blot. A luminescence detection reagent was also utilized to identify the effect of AKR1B1 on caspase-3/7 activity. The early and late stages of AKR1B1-induced apoptosis were assessed by performing Annexin V-FITC/PI double-staining assays. AKR1B1 expression was significantly downregulated in glioma tissues and GBM cell lines (T98G and 8401). Glioma cell proliferation was inhibited by AKR1B1 overexpression but was slightly increased by AKR1B1 knockdown. Additionally, AKR1B1-induced p38 MAPK phosphorylation and SB203580 reversed AKR1B1's inhibitory effect on glioma cell proliferation. AKR1B1 overexpression also inhibited Bcl-2 expression but increased BAX expression, whereas treatment with SB203580 reversed this phenomenon. Furthermore, AKR1B1 induced caspase-3/7 activity. The induction of early and late apoptosis by AKR1B1 was confirmed using an Annexin V-FITC/PI double-staining assay. In conclusion, AKR1B1 regulated glioma cell proliferation through the involvement of p38 MAPK-induced BAX/Bcl-2/caspase-3 apoptosis signaling. Therefore, AKR1B1 may serve as a new therapeutic target for glioma therapy development.
Collapse
Affiliation(s)
- Yu-Kai Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ann-Shung Lieu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chih-Lung Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
4
|
Li X, Yang DH, Ranganathan P. Editorial: Chemo-Resistance in Gastrointestinal Cancers. Front Oncol 2022; 12:821212. [PMID: 35155256 PMCID: PMC8834535 DOI: 10.3389/fonc.2022.821212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Xia Li
- Research Center for Biomedical Information Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Xia Li, ;
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | | |
Collapse
|
5
|
Variation of PPARG Expression in Chemotherapy-Sensitive Patients of Hypopharyngeal Squamous Cell Carcinoma. PPAR Res 2021; 2021:5525091. [PMID: 34054937 PMCID: PMC8149230 DOI: 10.1155/2021/5525091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/30/2021] [Indexed: 11/18/2022] Open
Abstract
Our previous study showed that the upregulation of peroxisome proliferator-activated receptor gamma (PPARG) could promote chemosensitivity of hypopharyngeal squamous cell carcinoma (HSCC) in chemotherapeutic treatments. Here, we acquired two more independent expression data of PPARG to validate the expression levels of PPARG in chemotherapy-sensitive patients (CSP) and its individualized variations compared to chemotherapy-non-sensitive patients (CNSP). Our results showed that overall PPARG expression was mildly downregulated (log fold change = −0.55; p value = 0.42; overexpression in three CSPs and reduced expression in four CSPs), which was not consistent with previous results (log fold change = 0.50; p = 0.22; overexpression in nine CSPs and reduced expression in three CSPs). Both studies indicated that PPARG expression variation was significantly associated with the Tumor-Node-Metastasis (TNM) stage (p = 7.45e − 7 and 6.50e − 4, for the first and second studies, respectively), which was used as one of the predictors of chemosensitivity. The new dataset analysis revealed 51 genes with significant gene expression changes in CSPs (LFC > 1 or <-1; p value < 0.01), and two of them (TMEM45A and RBP1) demonstrated strong coexpression with PPARG (Pearson correlation coefficient > 0.6 or <-0.6). There were 21 significant genes in the data from the first study, with no significant association with PPARG and no overlap with the 51 genes revealed in this study. Our results support the connection between PPARG and chemosensitivity in HSCC tumor cells. However, significant PPARG variation exists in CSPs, which may be influenced by multiple factors, including the TNM stage.
Collapse
|
6
|
Chen L, Jiang K, Chen H, Tang Y, Zhou X, Tan Y, Yuan Y, Xiao Q, Ding K. Deguelin induces apoptosis in colorectal cancer cells by activating the p38 MAPK pathway. Cancer Manag Res 2018; 11:95-105. [PMID: 30588113 PMCID: PMC6305136 DOI: 10.2147/cmar.s169476] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objectives Deguelin, a rotenoid extracted from Mundulea sericea (Leguminosae), exhibits antitumor effects on several types of human cancers. Due to the limited studies of deguelin on colorectal cancer (CRC), the present study was designed to investigate the antitumor effect of deguelin and to explore the underlying mechanism in CRC. Materials and methods Cell viability was assessed by the cell counting kit-8 (CCK-8) assay, and cell apoptosis was determined by the annexin v-propidium iodide staining using flow cytometry and Western blot in CRC cell lines after incubation with deguelin. The antitumor effect of deguelin was further evaluated in tumor xenograft models. Moreover, SB203580, a specific inhibitor of p38 MAPK, was used to confirm the involvement of p38 MAPK pathway in deguelin-induced apoptosis. Results Deguelin significantly inhibited cell proliferation and induced apoptosis in CRC cell lines (SW620 and RKO) in a time-dependent and dose-dependent manner. Western blot analysis also showed that the expression of proapoptotic proteins (cleaved caspase 3 and cleaved PARP) was upregulated, while that of antiapoptotic proteins (Bcl-2 and survivin) was downregulated after deguelin treatment in CRC cell lines. Moreover, oral administration of deguelin significantly suppressed tumor growth and induced apoptosis in subcutaneous xenograft mouse models without obvious toxicity. Additionally, Western blot revealed that deguelin-induced apoptosis might be regulated by the p38 MAPK pathway and inhibition of p38 MAPK could attenuate deguelin-induced proliferative inhibition and apoptosis in CRC cells. Conclusion Collectively, these results demonstrated that deguelin inhibited CRC cell growth by inducing apoptosis via activation of p38 MAPK pathway.
Collapse
Affiliation(s)
- Liubo Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| | - Kai Jiang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| | - Haiyan Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| | - Xinyi Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| | - Yinuo Tan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian Xiao
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| | - Kefeng Ding
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ; .,Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China, ;
| |
Collapse
|
7
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017. [PMID: 29156754 DOI: 10.18632/oncotarget.20482] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People's Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
8
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017. [PMID: 29156754 DOI: 10.18632/oncotarget.20482]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People's Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
9
|
Liu J, Zhang L, Mao P, Jiang G, Liu L, Wang J, Yang W, Owusu L, Li W. Functional characterization of a novel transcript of ERCC1 in chemotherapy resistance of ovarian cancer. Oncotarget 2017; 8:85759-85771. [PMID: 29156754 PMCID: PMC5689644 DOI: 10.18632/oncotarget.20482] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/25/2017] [Indexed: 01/13/2023] Open
Abstract
Approximately 15-20% of ovarian cancer patients receiving platinum-based chemotherapy are primary platinum-resistant. Identification of these patients and transfer to other more effective therapy could reduce the morbidity of ovarian cancer. ERCC1 is a DNA repair gene which can complex with XPF to repair cisplatin-induced DNA damage and cause chemotherapy resistance. In this study, we found a novel ERCC1 transcript initiated upstream of the normal transcription initiation site. The expression of this larger ERCC1 transcript dramatically increased following cisplatin treatment in ovarian cancer cells and was regulated by the MAPK pathway. This phenomenon conferred enhanced cisplatin resistance on ovarian cancer cells, and was confirmed with chemosensitive and chemoresistant patients' samples. Our data suggested that larger ERCC1 transcript levels correlated with the outcome of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ping Mao
- Department of General Surgery, The People’s Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jing Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Wei Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Sheng 450000, China
| | - Lawrence Owusu
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
10
|
Avan A, Narayan R, Giovannetti E, Peters GJ. Role of Akt signaling in resistance to DNA-targeted therapy. World J Clin Oncol 2016; 7:352-369. [PMID: 27777878 PMCID: PMC5056327 DOI: 10.5306/wjco.v7.i5.352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/06/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
The Akt signal transduction pathway controls most hallmarks of cancer. Activation of the Akt cascade promotes a malignant phenotype and is also widely implicated in drug resistance. Therefore, the modulation of Akt activity is regarded as an attractive strategy to enhance the efficacy of cancer therapy and irradiation. This pathway consists of phosphatidylinositol 3 kinase (PI3K), mammalian target of rapamycin, and the transforming serine-threonine kinase Akt protein isoforms, also known as protein kinase B. DNA-targeted agents, such as platinum agents, taxanes, and antimetabolites, as well as radiation have had a significant impact on cancer treatment by affecting DNA replication, which is aberrantly activated in malignancies. However, the caveat is that they may also trigger the activation of repairing mechanisms, such as upstream and downstream cascade of Akt survival pathway. Thus, each target can theoretically be inhibited in view of improving the potency of conventional treatment. Akt inhibitors, e.g., MK-2206 and perifosine, or PI3K modulators, e.g., LY294002 and Wortmannin, have shown some promising results in favor of sensitizing the cancer cells to the therapy in vitro and in vivo, which have provided the rationale for incorporation of these novel agents into multimodality treatment of different malignancies. Nevertheless, despite the acceptable safety profile of some of these agents in the clinical studies, with regard to the efficacy, the results are still too preliminary. Hence, we need to wait for the upcoming data from the ongoing trials before utilizing them into the standard care of cancer patients.
Collapse
|
11
|
Hong WG, Cho JH, Hwang SG, Lee E, Lee J, Kim JI, Um HD, Park JK. Chemosensitizing effect of podophyllotoxin acetate on topoisomerase inhibitors leads to synergistic enhancement of lung cancer cell apoptosis. Int J Oncol 2016; 48:2265-76. [PMID: 27035096 PMCID: PMC4863929 DOI: 10.3892/ijo.2016.3471] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022] Open
Abstract
Podophyllotoxin acetate (PA) acts as a radiosensitizer against non-small cell lung cancer (NSCLC) in vitro and in vivo. In this study, we examined its potential role as a chemosensitizer in conjunction with the topoisomerase inhibitors etoposide (Eto) and camptothecin (Cpt). The effects of combinations of PA and Eto/Cpt were examined with CompuSyn software in two NSCLC cell lines, A549 and NCI-H1299. Combination index (CI) values indicated synergistic effects of PA and the topoisomerase inhibitors. The intracellular mechanism underlying synergism was further determined using propidium iodide uptake, immunoblotting and electrophoretic mobility shift assay (EMSA). Combination of PA with Eto/Cpt promoted disruption of the dynamics of actin filaments, leading to subsequent enhancement of apoptotic cell death via induction of caspase-3, -8, and -9, accompanied by increased phosphorylation of p38. Conversely, suppression of p38 phosphorylation blocked the apoptotic effect of the drug combinations. Notably, CREB-1, a transcription factor, was constitutively activated in both cell types, and synergistically inhibited upon combination treatment. Our results collectively indicate that PA functions as a chemosensitizer by enhancing apoptosis through activation of the p38/caspase axis and suppression of CREB-1.
Collapse
Affiliation(s)
- Wan Gi Hong
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Jeong Hyun Cho
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Sang-Gu Hwang
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Eunah Lee
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si, Geonggi-do, Republic of Korea
| | - Jaeseok Lee
- Department of Biological Science, College of Biological Science, Konkuk University, Seoul, Republic of Korea
| | - Jong-Il Kim
- Department of Food and Microbial Technology, College of Natural Sciences, Seoul Women's University, Seoul, Republic of Korea
| | - Hong-Duck Um
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| | - Jong Kuk Park
- Department of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-Gu, Seoul, Republic of Korea
| |
Collapse
|
12
|
Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, He C, Pan H. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett 2013; 344:174-9. [PMID: 24333738 DOI: 10.1016/j.canlet.2013.11.019] [Citation(s) in RCA: 745] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/09/2013] [Accepted: 11/19/2013] [Indexed: 01/01/2023]
Abstract
The Mitogen Activated Protein Kinase (MAPK) signaling plays a critical role in the outcome and the sensitivity to anticancer therapies. Activated MAPK can transmit extracellular signals to regulate cell growth, proliferation, differentiation, migration, apoptosis and so on. Apoptosis as well as macroautophagy (hereafter referred to as autophagy) can be induced by extracellular stimuli such the treatment of chemotherapeutic agents, resulting in different cell response to these drugs. However, the molecular mechanisms mediating these two cellular processes remain largely unknown. Recently, several studies provide new insights into p38 and JNK MAPK pathways function in the control of the balance of autophagy and apoptosis in response to genotoxic stress. Our increased understanding of the role of p38 and JNK MAPK pathways in regulating the balance of autophagy and apoptosis will hopefully provide prospective strategies for cancer therapy.
Collapse
Affiliation(s)
- Xinbing Sui
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China; Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Na Kong
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China; Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Li Ye
- Department of General Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China; Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Qin Zhang
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China.
| | - Chao He
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China; Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China; Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
13
|
Yang K, Liu Y, Liu Z, Liu J, Liu X, Chen X, Li C, Zeng Y. p38γ overexpression in gliomas and its role in proliferation and apoptosis. Sci Rep 2013; 3:2089. [PMID: 23807566 PMCID: PMC3695572 DOI: 10.1038/srep02089] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/06/2013] [Indexed: 12/27/2022] Open
Abstract
The objective of this study was to confirm the biological role of p38γ in human gliomas. The expression profiles of p38γ and hTERT in human glioma samples were detected by Western Blot and immunohistochemistry. RNA interference was performed in U251 cells by p38γ silencing. Cell proliferation and apoptosis were assayed by CCK-8 and flow cytometric analysis, and then RNA and protein expression levels were measured by real-time RT-PCR and Western Blot, respectively. Telomerase activity assays and Caspase-3,-9 activation assays were also conducted. The results showed p38γ had a positive correlation with the glioma's malignancy grade and that the treatment of U251 cells with p38γ-siRNA inhibited proliferation and induced apoptosis. Correspondingly, hTERT expression and telomerase activity were down regulated and Caspase-3 and -9 activities were elevated. In conclusion, p38γ may serve as an oncogenic factor promoting the growth and progression of gliomas and may become a useful therapeutic target.
Collapse
Affiliation(s)
- Kui Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen HJ, Jiang YL, Lin CM, Tsai SC, Peng SF, Fushiya S, Hour MJ, Yang JS. Dual inhibition of EGFR and c-Met kinase activation by MJ-56 reduces metastasis of HT29 human colorectal cancer cells. Int J Oncol 2013; 43:141-50. [PMID: 23677180 DOI: 10.3892/ijo.2013.1941] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 04/09/2013] [Indexed: 11/05/2022] Open
Abstract
Quinazolinone derivatives are known to possess anticancer activities on cell metastasis and cell death in different human cancer cell lines. Here, we studied the anti-metastasis activity and the underlying mechanisms of the novel quinazoline derivative MJ-56 (6-pyrrolidinyl-2-(3-bromostyryl)quinazolin-4-one). MJ-56 inhibited cell migration and invasion of HT29 human colorectal cancer cells by wound-healing and Matrigel-coated transwell assays in a concentration-dependent manner. MJ-56-treated cells resulted in the reduced expression of matrix metalloproteinase (MMP)-2, -7, -9 and -10 and the reduced enzymatic activities of MMP-2 and MMP-9. In contrast, MJ-56-treated cells enhanced the expression of the tissue inhibitors of metalloproteinases (TIMPs) TIMP-1 and TIMP-2. Further analyses showed that MJ-56 attenuated the activities of epidermal growth factor receptor (EGFR), c-Met and the downstream ERK-mediated MAPK and PI3K/AKT/mTOR signaling pathways, which led to decreased protein synthesis by dephosphorylating the translation initiation factors eIF-4B, eIF-4E, eIF-4G and S6 ribosomal protein. In addition, MJ-56 interfered with the NF-κB signaling via impairing PI3K/AKT activation and subsequently reduced the NF-κB-mediated transcription of MMPs. Taken together, the reduced expression of phosphor-EGFR and c-MET is chiefly responsible for all events of blocking metastasis. Our results suggest a potential role of MJ-56 on therapy of colorectal cancer metastasis.
Collapse
Affiliation(s)
- Hui-Jye Chen
- Graduate Institute of Molecular Systems Biomedicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Guo YF, Wang XB, Tian XY, Li Y, Li B, Huang Q, Zhang M, Li Z. Tumor-derived hepatocyte growth factor is associated with poor prognosis of patients with glioma and influences the chemosensitivity of glioma cell line to cisplatin in vitro. World J Surg Oncol 2012; 10:128. [PMID: 22741575 PMCID: PMC3447698 DOI: 10.1186/1477-7819-10-128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/29/2012] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND We examined the association of tumor-derived hepatocyte growth factor (HGF) with the clinicopathological features of gliomas and investigated the effect of HGF inhibition on the biological behavior of tumor cells in vitro in order to determine whether HGF is a valuable prognostic predictor for glioma patients. METHODS Seventy-six cases of glioma were collected. The tumor-derived HGF expression, cell proliferation index (PI) and intratumoral microvessels were evaluated by immunohistochemistry. Correlation between immunostaining and clinicopathological parameters, as well as the follow-up data of patients, was analyzed statistically. U87MG glioma cells were transfected with short interference (si)-RNA for HGF, and the cell viability, migratory ability and chemosensitivity to cisplatin were evaluated in vitro. RESULTS Both high HGF expression in tumor cells (59.2%, 45/76) and high PI were significantly associated with high-grade glioma and increased microvessels in tumors (P < 0.05). However, only histological grading (P = 0.004) and high-expression of HGF (P = 0.008) emerged as independent prognostic factors for the overall survival of glioma patients. The tumor-derived HGF mRNA and protein expressions were significantly decreased in vitro after transfection of HGF siRNA. HGF siRNA inhibited the cell growth and reduced cell migratory ability. Moreover, HGF siRNA transfection enhanced the chemosensitivity of U87MG glioma cells to cisplatin. CONCLUSION This study indicated that there was significant correlation among tumor cell-derived HGF, cell proliferation and microvessel proliferation in gliomas. HGF might influence tumor progression by modulating the cell growth, migration and chemoresistance to drugs. Increased expression of HGF may be a valuable predictor for prognostic evaluation of glioma patients.
Collapse
Affiliation(s)
- You-feng Guo
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Xiao-bing Wang
- Department of Pathology, Guangdong General Hospital, 106 Zhongshan Road II, Guangzhou 510080, China
| | - Xiao-ying Tian
- School of Chinese Medicine, Hong Kong Baptist University, 7, Baptist University Road, Kowloon Tong, Hong Kong, China
| | - Yang Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Bin Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Quan Huang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, 58, Zhongshan Road II, Guangzhou 510080, China
| | - Meng Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Zhi Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| |
Collapse
|
16
|
Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol 2012; 14:958-78. [PMID: 22573310 DOI: 10.1093/neuonc/nos116] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microglia, which contribute substantially to the tumor mass of glioblastoma, have been shown to play an important role in glioma growth and invasion. While a large number of experimental studies on functional attributes of microglia in glioma provide evidence for their tumor-supporting roles, there also exist hints in support of their anti-tumor properties. Microglial activities during glioma progression seem multifaceted. They have been attributed to the receptors expressed on the microglia surface, to glioma-derived molecules that have an effect on microglia, and to the molecules released by microglia in response to their environment under glioma control, which can have autocrine effects. In this paper, the microglia and glioma literature is reviewed. We provide a synopsis of the molecular profile of microglia under the influence of glioma in order to help establish a rational basis for their potential therapeutic use. The ability of microglia precursors to cross the blood-brain barrier makes them an attractive target for the development of novel cell-based treatments of malignant glioma.
Collapse
Affiliation(s)
- Wei Li
- Brain Tumor Research Laboratories, The Brain and Mind Research Institute, University of Sydney, 94 Mallett St, Camperdown, Sydney, NSW 2050, Australia
| | | |
Collapse
|
17
|
Cantelmo AR, Cammarota R, Noonan DM, Focaccetti C, Comoglio PM, Prat M, Albini A. Cell delivery of Met docking site peptides inhibit angiogenesis and vascular tumor growth. Oncogene 2010; 29:5286-98. [PMID: 20603611 PMCID: PMC3007100 DOI: 10.1038/onc.2010.267] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hepatocyte growth factor (HGF) and its receptor Met are responsible for a wide variety of cellular responses, both physiologically during embryo development and tissue homeostasis, and pathologically, particularly during tumor growth and dissemination. In cancer, Met can act as an oncogene on tumor cells, as well as a pro-angiogenic factor activating endothelial cells and inducing new vessel formation. Molecules interfering with Met activity could be valuable therapeutic agents. Here we have investigated the antiangiogenic properties of a synthetic peptide mimicking the docking site of the Met carboxyl-terminal tail, which was delivered into the cells by fusion with the internalization sequences from Antennapedia or HIV-Tat. We showed that these peptides inhibit ligand-dependent endothelial cell proliferation, motility, invasiveness and morphogenesis in vitro to an even greater extent and with much less toxicity than the Met inhibitor PHA-665752, which correlated with interference of HGF-dependent downstream signaling. In vivo, the peptides inhibited HGF-induced angiogenesis in the matrigel sponge assay and impaired xenograft tumor growth and vascularization in Kaposi's sarcoma. These data show that interference with the Met receptor intracellular sequence impairs HGF-induced angiogenesis, suggesting the use of antidocking site compounds as a therapeutic strategy to counteract angiogenesis in cancer as well as in other diseases.
Collapse
Affiliation(s)
- A R Cantelmo
- Oncology Research, Science and Technology Pole, Casa di Cura MultiMedica-IRCCS, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|