1
|
Li Z, Shu X, Liu X, Li Q, Hu Y, Jia B, Song M. Cellular and Molecular Mechanisms of Chemoresistance for Gastric Cancer. Int J Gen Med 2024; 17:3779-3788. [PMID: 39224691 PMCID: PMC11368108 DOI: 10.2147/ijgm.s473749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors in the digestive tract, and chemotherapy plays an irreplaceable role in the comprehensive treatment of GC. However, chemoresistance makes it difficult for patients with GC to benefit steadily from chemotherapy in the long term, which ultimately leads to tumor recurrence, metastasis, and patient death. Elucidating the detailed mechanism of chemoresistance in GC and identifying specific therapeutic targets will help to solve the difficult problem of chemoresistance and improve the prognosis of patients with GC. This review summarizes and clarifies the cellular and molecular mechanisms underlying chemoresistance for GC.
Collapse
Affiliation(s)
- Zonglin Li
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Xingming Shu
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Xin Liu
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qiuyun Li
- Grade 2023, Clinical Medicine College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yifu Hu
- Grade 2023, Clinical Medicine College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Bingbing Jia
- Grade 2023, Clinical Medicine College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Min Song
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
2
|
Zhang C, Pan G, Qin JJ. Role of F-box proteins in human upper gastrointestinal tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189035. [PMID: 38049014 DOI: 10.1016/j.bbcan.2023.189035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Protein ubiquitination and degradation is an essential physiological process in almost all organisms. As the key participants in this process, the E3 ubiquitin ligases have been widely studied and recognized. F-box proteins, a crucial component of E3 ubiquitin ligases that regulates diverse biological functions, including cell differentiation, proliferation, migration, and apoptosis by facilitating the degradation of substrate proteins. Currently, there is an increasing focus on studying the role of F-box proteins in cancer. In this review, we present a comprehensive overview of the significant contributions of F-box proteins to the development of upper gastrointestinal tumors, highlighting their dual roles as both carcinogens and tumor suppressors. We delve into the molecular mechanisms underlying the involvement of F-box proteins in upper gastrointestinal tumors, exploring their interactions with specific substrates and their cross-talks with other key signaling pathways. Furthermore, we discuss the implications of F-box proteins in radiotherapy resistance in the upper gastrointestinal tract, emphasizing their potential as clinical therapeutic and prognostic targets. Overall, this review provides an up-to-date understanding of the intricate involvement of F-box proteins in human upper gastrointestinal tumors, offering valuable insights for the identification of prognostic markers and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Che Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guangzhao Pan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jiang-Jiang Qin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
3
|
Yang Y, Zhong F, Jiang J, Li M, Yao F, Liu J, Cheng Y, Xu S, Chen S, Zhang H, Xu Y, Huang B. Bioinformatic analysis of the expression profile and identification of RhoGDI2 as a biomarker in imatinib-resistant K562 cells. Hematology 2023; 28:2244856. [PMID: 37594290 DOI: 10.1080/16078454.2023.2244856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/26/2023] [Indexed: 08/19/2023] Open
Abstract
OBJECTIVES Chronic myeloid leukemia (CML) is an aggressive malignancy originating from hematopoietic stem cells. Imatinib (IM), the first-generation tyrosine kinase inhibitor, has greatly improved theliving quality of CML patients. However, owing to the recurrence and treatment failure coming from tyrosine kinase inhibitor (TKIs) resistance, some CML patients still bear poor prognosis. Therefore, we aimed to seek potential signaling pathways and specific biomarkers for imatinib resistance. METHODS We performed mRNA and miRNA expression profiling in imatinib-sensitive K562 cells (IS-K562) and imatinib-resistant K562 cells (IR-K562). Differentially expressed genes (DEGs) were identified and pathway enrichment analyses were performed to explore the potential mechanism. The protein-protein interaction (PPI) network and miRNA-mRNA regulatory network were constructed to explore potential relationships among these genes. RT-qPCR, western blot and CCK8 were used for further experiments. RESULTS A total of 623 DEGs and 61 differentially expressed miRNAs were identified. GO revealed that DEGs were mainly involved in cell adhesion, cell migration, differentiation, and inflammatory response. KEGG revealed that DEGs were typically enriched in the Rap1 signaling pathway, focal adhesion, proteoglycans and transcriptional misregulation in cancer, signaling pathways regulating pluripotency of stem cells and some immune-related pathways. The protein-protein interaction (PPI) network and miRNA-mRNA regulatory network revealed a web of diverse connections among genes. Finally, we proved that RHoGDI2 played a critical role in imatinib resistance. CONCLUSION The dynamic interplay between genes and signaling pathways is associated with TKIs resistance and RHoGDI2 is identified as a biomarker in IR-K562.
Collapse
Affiliation(s)
- Yulin Yang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- School of Public Health, Nanchang University, Nanchang, People's Republic of China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Junyao Jiang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Meiyong Li
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Ying Cheng
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- School of Public Health, Nanchang University, Nanchang, People's Republic of China
| | - Shuai Xu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- School of Public Health, Nanchang University, Nanchang, People's Republic of China
| | - Song Chen
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- School of Public Health, Nanchang University, Nanchang, People's Republic of China
| | - Haibin Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yanmei Xu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
4
|
Hui T, Yiling J, Guangqun C, Ran L, Hui L, Lan Y, Jie H, Su Q. Diallyl disulfide downregulating RhoGDI2 induces differentiation and inhibit invasion via the Rac1/Pak1/LIMK1 pathway in human leukemia HL-60 cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:1063-1077. [PMID: 36793247 DOI: 10.1002/tox.23748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Leukemia is a type of disease in which hematopoietic stem cells proliferate clonally at the genetic level. We discovered previously by high-resolution mass spectrometry that diallyl disulfide (DADS), which is one of the effective ingredients of garlic, reduces the performance of RhoGDI2 from APL HL-60 cells. Although RhoGDI2 is oversubscribed in several cancer categories, the effect of RhoGDI2 in HL-60 cells has remained unexplained. We aimed to investigate the influence of RhoGDI2 on DADS-induced differentiation of HL-60 cells to elucidate the association among the effect of inhibition or over-expression of RhoGDI2 with HL-60 cell polarization, migration and invasion, which is important for establishing a novel generation of inducers to elicit leukemia cell polarization. Co-transfection with RhoGDI2-targeted miRNAs apparently decreases the malignant biological behavior of cells and upregulates cytopenias in DADS-treated HL-60 cell lines, which increases CD11b and decreases CD33 and mRNA levels of Rac1, PAK1 and LIMK1. Meanwhile, we generated HL-60 cell lines with high-expressing RhoGDI2. The proliferation, migration and invasion capacity of such cells were significantly increased by the treated with DADS, while the reduction capacity of the cells was decreased. There was a reduction in CD11b and an increase in CD33 production, as well as an increase in the mRNA levels of Rac1, PAK1 and LIMK1. It also confirmed that inhibition of RhoGDI2 attenuates the EMT cascade via the Rac1/Pak1/LIMK1 pathway, thereby inhibiting the malignant biological behavior of HL-60 cells. Thus, we considered that inhibition of RhoGDI2 expression might be a new therapeutic direction for the treatment of human promyelocytic leukemia. The anti-cancer property of DADS against HL-60 leukemia cells might be regulated by RhoGDI2 through the Rac1-Pak1-LIMK1 pathway, which provides new evidence for DADS as a clinical anti-cancer medicine.
Collapse
Affiliation(s)
- Tan Hui
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Jiang Yiling
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, First Affiliated Hospital, University of South China, Hengyang, China
| | - Chen Guangqun
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, Loudi Central Hospital, Loudi, China
| | - Liu Ran
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Pathology, The First Hospital of Changsha, Changsha, China
| | - Ling Hui
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Yi Lan
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - He Jie
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Qi Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
5
|
Yi B, Hu Y, Zhu D, Yao J, Zhou J, Zhang Y, He Z, Zhang L, Zhang Z, Yang J, Tang Y, Huang Y, Li D, Liu Q. RhoGDI2 induced malignant phenotypes of pancreatic cancer cells via regulating Snail expression. Genes Genomics 2022; 44:561-569. [PMID: 35147897 DOI: 10.1007/s13258-022-01217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 01/16/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Rho GDP dissociation inhibitor 2 (RhoGDI2) has been shown to contribute to the aggressive phenotypes of human cancers, such as tumor metastasis and chemoresistance. OBJECTIVE This study aimed to assess the effects of RhoGDI2 on tumor progression and chemoresistance in pancreatic cancer cells. METHODS The expression of RhoGDI2 in pancreatic cancer cells was detected by Western blot analysis. Gain-of-function and loss-of-function approaches were done to examine the malignant phenotypes of the RhoGDI2-expressing or RhoGDI2-depleting cells. The correlation between RhoGDI2 and Snail was also analyzed. RESULTS Differential expression of RhoGDI2 protein in pancreatic cancer cell lines was identified. Gain-of-function and loss-of-function experiments showed that RhoGDI2 induced the malignant phenotypes of pancreatic cancer cells, including proliferation, migration, invasion, and gemcitabine (GEM) chemoresistance. The upregulation of RhoGDI2 stimulated the expression of Snail, resulting in the altered expression of epithelial marker E-cadherin and mesenchymal marker Vimentin, which were characteristics of the tumorigenic activity of epithelial-mesenchymal transition. The expression of RhoGDI2 and Snail was upregulated in clinical tumor samples, and higher expression of RhoGDI2 or Snail was significantly associated with poor patient survival in pancreatic ductal adenocarcinoma (PDAC). CONCLUSION The findings indicated that RhoGDI2 promoted GEM resistance and tumor progression in pancreatic cancer and that RhoGDI2 might be a potential therapeutic target in patients with PDAC.
Collapse
Affiliation(s)
- Bin Yi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - You Hu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Dongming Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Jun Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Yi Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Zhilong He
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Lifeng Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Zixiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Jian Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Yuchen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Yujie Huang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China
| | - Dechun Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, People's Republic of China.
| | - Qiuhua Liu
- Department of General Surgery, The First People's Hospital of Zhangjiagang City, No. 68 Jiyang Western Road, Suzhou, People's Republic of China.
| |
Collapse
|
6
|
Wang H, Lu Y, Wang M, Wu Y, Wang X, Li Y. Roles of E3 ubiquitin ligases in gastric cancer carcinogenesis and their effects on cisplatin resistance. J Mol Med (Berl) 2021; 99:193-212. [PMID: 33392633 DOI: 10.1007/s00109-020-02015-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Although gastric cancer (GC) is one of the most common cancers with high incidence and mortality rates, its pathogenesis is still not elucidated. GC carcinogenesis is complicated and involved in the activation of oncoproteins and inactivation of tumor suppressors. The ubiquitin-proteasome system (UPS) is crucial for protein degradation and regulation of physiological and pathological processes. E3 ubiquitin ligases are pivotal enzymes in UPS, containing various subfamily proteins. Previous studies report that some E3 ligases, including SKP2, CUL1, and MDM2, act as oncoproteins in GC carcinogenesis. On the other hand, FBXW7, FBXL5, FBXO31, RNF43, and RNF180 exert as tumor suppressors in GC carcinogenesis. Moreover, E3 ligases modulate cell growth, cell apoptosis, and cell cycle; thus, it is complicated to confer cisplatin resistance/sensitivity in GC cells. The intrinsic and acquired cisplatin resistance limits its clinical application against GC. In this review, we explore oncogenic and tumor suppressive roles of E3 ligases in GC carcinogenesis and focus on the effects of E3 ligases on cisplatin resistance in GC cells, which will provide novel therapeutic targets for GC therapy, especially for cisplatin-resistant patients.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Mingliang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Youliang Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
7
|
Establishment of Acquired Cisplatin Resistance in Ovarian Cancer Cell Lines Characterized by Enriched Metastatic Properties with Increased Twist Expression. Int J Mol Sci 2020; 21:ijms21207613. [PMID: 33076245 PMCID: PMC7589258 DOI: 10.3390/ijms21207613] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal of the gynecologic cancers, and platinum-based treatment is a part of the standard first-line chemotherapy regimen. However, rapid development of acquired cisplatin resistance remains the main cause of treatment failure, and the underlying mechanism of resistance in OC treatment remains poorly understood. Faced with this problem, our aim in this study was to generate cisplatin-resistant (CisR) OC cell models in vitro and investigate the role of epithelial–mesenchymal transition (EMT) transcription factor Twist on acquired cisplatin resistance in OC cell models. To achieve this aim, OC cell lines OV-90 and SKOV-3 were exposed to cisplatin using pulse dosing and stepwise dose escalation methods for a duration of eight months, and a total of four CisR sublines were generated, two for each cell line. The acquired cisplatin resistance was confirmed by determination of 50% inhibitory concentration (IC50) and clonogenic survival assay. Furthermore, the CisR cells were studied to assess their respective characteristics of metastasis, EMT phenotype, DNA repair and endoplasmic reticulum stress-mediated cell death. We found the IC50 of CisR cells to cisplatin was 3–5 times higher than parental cells. The expression of Twist and metastatic ability of CisR cells were significantly greater than those of sensitive cells. The CisR cells displayed an EMT phenotype with decreased epithelial cell marker E-cadherin and increased mesenchymal proteins N-cadherin and vimentin. We observed that CisR cells showed significantly higher expression of DNA repair proteins, X-ray repair cross-complementing protein 1 (XRCC1) and poly (ADP-ribose) polymerases 1 (PARP1), with significantly reduced endoplasmic reticulum (ER) stress-mediated cell death. Moreover, Twist knockdown reduced metastatic ability of CisR cells by suppressing EMT, DNA repair and inducing ER stress-induced cell death. In conclusion, we highlighted the utilization of an acquired cisplatin resistance model to identify the potential role of Twist as a therapeutic target to reverse acquired cisplatin resistance in OC.
Collapse
|
8
|
Zeng Y, Ren M, Li Y, Liu Y, Chen C, Su J, Su B, Xia H, Liu F, Jiang H, Ling H, Zeng X, Su Q. Knockdown of RhoGDI2 represses human gastric cancer cell proliferation, invasion and drug resistance via the Rac1/Pak1/LIMK1 pathway. Cancer Lett 2020; 492:136-146. [PMID: 32860848 DOI: 10.1016/j.canlet.2020.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/03/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is the fifth most common primary malignancy in humans. Rho GDP dissociation inhibitor 2 (RhoGDI2) is overexpressed in multiple cancer types, but the role of RhoGDI2 in GC has not been elucidated. This study aims to determine the level of RhoGDI2 in GC and to confirm the effect of its inhibition or overexpression on GC cell migration, invasion and chemosensitivity. RhoGDI2 level is significantly enhanced in human GC tissue samples in comparison with normal gastric epithelium and corresponding para-cancerous samples. The expression of RhoGDI2 is correlated with clinicopathological parameters and prognosis. Transfection in combination with miRNA targeting of RhoGDI2 in GC cell lines remarkably downregulates GC cell migration and invasion and reduces the mRNA levels of Rac1, Pak1 and LIMK1. The inhibition of RhoGDI2 downregulates GC cell migration and invasion by attenuating the EMT cascade via the Rac1/Pak1/LIMK1 pathway. Knockdown of RhoGDI2 is a potential therapeutic strategy for GC.
Collapse
Affiliation(s)
- Ying Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China; School of Nursing, University of South China, China
| | - Mei Ren
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China
| | - Yukun Li
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China
| | - Yanli Liu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China
| | - Cong Chen
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China
| | - Jian Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China; Department of Pathology, Second Affiliated Hospital, University of South China, China
| | - Bo Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China; Key Laboratory for Pharmacoproteomics of Hunan Provincial University, Institute of Pharmacy and Pharmacology, University of South China, China
| | - Hong Xia
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China
| | - Fang Liu
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China
| | - Hao Jiang
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China
| | - Hui Ling
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China
| | - Xi Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China; Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China.
| | - Qi Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, China; Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, China.
| |
Collapse
|
9
|
Zheng CW, Zeng RJ, Xu LY, Li EM. Rho GTPases: Promising candidates for overcoming chemotherapeutic resistance. Cancer Lett 2020; 475:65-78. [PMID: 31981606 DOI: 10.1016/j.canlet.2020.01.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
Despite therapeutic advances, resistance to chemotherapy remains a major challenge to patients with malignancies. Rho GTPases are essential for the development and progression of various diseases including cancer, and a vast number of studies have linked Rho GTPases to chemoresistance. Therefore, understanding the underlying mechanisms can expound the effects of Rho GTPases towards chemotherapeutic agents, and targeting Rho GTPases is a promising strategy to downregulate the chemo-protective pathways and overcome chemoresistance. Importantly, exceptions in certain biological conditions and interactions among the members of Rho GTPases should be noted. In this review, we focus on the role of Rho GTPases, particularly Rac1, in regulating chemoresistance and provide an overview of their related mechanisms and available inhibitors, which may offer novel options for future targeted cancer therapy.
Collapse
Affiliation(s)
- Chun-Wen Zheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China
| | - Rui-Jie Zeng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China
| | - Li-Yan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China.
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China; The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
10
|
Wong KM, Song J, Saini V, Wong YH. Small Molecules as Drugs to Upregulate Metastasis Suppressors in Cancer Cells. Curr Med Chem 2019; 26:5876-5899. [PMID: 29788870 DOI: 10.2174/0929867325666180522090842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/20/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022]
Abstract
It is well-recognized that the majority of cancer-related deaths is attributed to metastasis, which can arise from virtually any type of tumor. Metastasis is a complex multistep process wherein cancer cells must break away from the primary tumor, intravasate into the circulatory or lymphatic systems, extravasate, proliferate and eventually colonize secondary sites. Since these molecular processes involve the coordinated actions of numerous proteins, targeted disruptions of key players along these pathways represent possible therapeutic interventions to impede metastasis formation and reduce cancer mortality. A diverse group of proteins with demonstrated ability to inhibit metastatic colonization have been identified and they are collectively known as metastasis suppressors. Given that the metastasis suppressors are often downregulated in tumors, drug-induced re-expression or upregulation of these proteins represents a promising approach to limit metastasis. Indeed, over 40 compounds are known to exhibit efficacy in upregulating the expression of metastasis suppressors via transcriptional or post-transcriptional mechanisms, and the most promising ones are being evaluated for their translational potentials. These small molecules range from natural products to drugs in clinical use and they apparently target different molecular pathways, reflecting the diverse nature of the metastasis suppressors. In this review, we provide an overview of the different classes of compounds known to possess the ability to upregulate one or more metastasis suppressors, with an emphasis on their mechanisms of action and therapeutic potentials.
Collapse
Affiliation(s)
- Ka Ming Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Jiaxing Song
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Vasu Saini
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yung H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
11
|
Zhang J, Wei Y, Min J, Wang Y, Yin L, Cao G, Shen H. Knockdown of RAP2A gene expression suppresses cisplatin resistance in gastric cancer cells. Oncol Lett 2019; 19:350-358. [PMID: 31897147 PMCID: PMC6923840 DOI: 10.3892/ol.2019.11086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Cisplatin (DDP) resistance is closely associated with the failure of chemotherapy to manage various different types of human cancer. The GTPase protein Ras-related protein Rap-2a (RAP2A) regulates cancer cell proliferation, migration and invasion; however, little is currently known regarding its role in cancer cell resistance to chemotherapy. The present study investigated the potential roles of the RAP2A gene in gastric cancer cell resistance to DDP treatment. The DDP half maximal inhibitory concentration (IC50) values for the proliferation inhibition of MGC803 and MGC803/DDP gastric cancer cells were determined by treating the cells with a DDP concentration gradient and measuring their survival rates using the Cell Counting Kit-8 (CCK-8) assay; cell viability was also assessed using the CCK-8 assay. Cell migration and invasion were assessed using Transwell Matrigel assays, and apoptosis and DNA damage were evaluated using flow cytometry and Hoechst staining. RAP2A expression was knocked down by siRNA transfection, and RAP2A protein levels were examined using western blotting. The DDP IC50 values for DDP-resistant MGC803/DDP cells were greater than those for MGC803 cells. Furthermore, MGC803/DDP cells exhibited increased levels of viability, migration and invasion, and decreased levels of apoptosis and DNA damage during DDP treatment. Knockdown of RAP2A expression significantly promoted MGC803/DDP cell apoptosis and DNA damage, and decreased the viability and invasion capabilities of these cells following treatment with DDP. The results of the present study revealed that RAP2A expression promotes DDP resistance in gastric cancer cells by increasing their viability, migration and invasion capacities, and by suppressing apoptosis and DNA damage.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Yunhai Wei
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Jie Min
- Intensive Care Unit, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Yan Wang
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Lei Yin
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Guoliang Cao
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Hua Shen
- Department of Gastrointestinal Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
12
|
Cho HJ, Kim JT, Baek KE, Kim BY, Lee HG. Regulation of Rho GTPases by RhoGDIs in Human Cancers. Cells 2019; 8:cells8091037. [PMID: 31492019 PMCID: PMC6769525 DOI: 10.3390/cells8091037] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Rho GDP dissociation inhibitors (RhoGDIs) play important roles in various cellular processes, including cell migration, adhesion, and proliferation, by regulating the functions of the Rho GTPase family. Dissociation of Rho GTPases from RhoGDIs is necessary for their spatiotemporal activation and is dynamically regulated by several mechanisms, such as phosphorylation, sumoylation, and protein interaction. The expression of RhoGDIs has changed in many human cancers and become associated with the malignant phenotype, including migration, invasion, metastasis, and resistance to anticancer agents. Here, we review how RhoGDIs control the function of Rho GTPases by regulating their spatiotemporal activity and describe the regulatory mechanisms of the dissociation of Rho GTPases from RhoGDIs. We also discuss the role of RhoGDIs in cancer progression and their potential uses for therapeutic intervention.
Collapse
Affiliation(s)
- Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Jong-Tae Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Kyoung Eun Baek
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Bo-Yeon Kim
- Anticancer Cancer Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
- Department of Biomolecular Science, University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
13
|
Gong J, Zhou Y, Liu D, Huo J. F-box proteins involved in cancer-associated drug resistance. Oncol Lett 2018; 15:8891-8900. [PMID: 29805625 PMCID: PMC5958692 DOI: 10.3892/ol.2018.8500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
The ubiquitin proteasome system (UPS) regulated human biological processes through the appropriate and efficient proteolysis of cellular proteins. F-box proteins are the vital components of SKP1-CUL1-FBP (SCF)-type E3 ubiquitin ligases that determine substrate specificity. As F-box proteins have the ability to control the degradation of several crucial protein targets associated with drug resistance, the dysregulation of these proteins may lead to induction of chemoresistance in cancer cells. Chemotherapy is one of the most conventional therapeutic approaches of treatment of patients with cancer. However, its exclusive application in clinical settings is restricted due to the development of chemoresistance, which typically results treatment failure. Therefore, overcoming drug resistance is considered as one of the most critical issues that researchers and clinician associated with oncology face. The present review serves to provide a comprehensive overview of F-box proteins and their possible targets as well as their correlation with the chemoresistance and chemosensitization of cancer cells. The article also presents an integrated representation of the complex regulatory mechanisms responsible for chemoresistance, which may lay the foundation to explore sensible candidate drugs for therapeutic intervention.
Collapse
Affiliation(s)
- Jian Gong
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yuqian Zhou
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jirong Huo
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
14
|
Yu Y, Jin H, Xu J, Gu J, Li X, Xie Q, Huang H, Li J, Tian Z, Jiang G, Chen C, He F, Wu XR, Huang C. XIAP overexpression promotes bladder cancer invasion in vitro and lung metastasis in vivo via enhancing nucleolin-mediated Rho-GDIβ mRNA stability. Int J Cancer 2018; 142:2040-2055. [PMID: 29250796 PMCID: PMC5867227 DOI: 10.1002/ijc.31223] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/02/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022]
Abstract
Our recent studies demonstrate that X-linked inhibitor of apoptosis protein (XIAP) is essential for regulating colorectal cancer invasion. Here, we discovered that RhoGDIβ was a key XIAP downstream effector mediating bladder cancer (BC) invasion in vitro and in vivo. We found that both XIAP and RhoGDIβ expressions were consistently elevated in BCs of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-treated mice in comparison to bladder tissues from vehicle-treated mice and human BCs in comparison to the paired adjacent normal bladder tissues. Knockdown of XIAP attenuated RhoGDIβ expression and reduced cancer cell invasion, whereas RhoGDIβ expression was attenuated in BBN-treated urothelium of RING-deletion knockin mice. Mechanistically, XIAP stabilized RhoGDIβ mRNA by its positively regulating nucleolin mRNA stability via Erks-dependent manner. Moreover, ectopic expression of GFP-RhoGDIβ in T24T(shXIAP) cells restored its lung metastasis in nude mice. Our results demonstrate that XIAP-regulated Erks/nucleolin/RhoGDIβ axis promoted BC invasion and lung metastasis.
Collapse
Affiliation(s)
- Yonghui Yu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Honglei Jin
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Jiheng Xu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Jiayan Gu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Xin Li
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Qipeng Xie
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Zhongxian Tian
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Guosong Jiang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Caiyi Chen
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Feng He
- Departments of Urology and Pathology, New York University School of Medicine, Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY 10016, USA
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine, Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY 10016, USA
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| |
Collapse
|
15
|
Jang HJ, Suh PG, Lee YJ, Shin KJ, Cocco L, Chae YC. PLCγ1: Potential arbitrator of cancer progression. Adv Biol Regul 2018; 67:179-189. [PMID: 29174396 DOI: 10.1016/j.jbior.2017.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 06/07/2023]
Abstract
Phospholipase C (PLC) is an essential mediator of cellular signaling. PLC regulates multiple cellular processes by generating bioactive molecules such as inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). These products propagate and regulate cellular signaling via calcium (Ca2+) mobilization and activation of protein kinase C (PKC), other kinases, and ion channels. PLCγ1, one of the primary subtypes of PLC, is directly activated by membrane receptors, including receptor tyrosine kinases (RTKs), and adhesion receptors such as integrin. PLCγ1 mediates signaling through direct interactions with other signaling molecules via SH domains, as well as its lipase activity. PLCγ1 is frequently enriched and mutated in various cancers, and is involved in the processes of tumorigenesis, including proliferation, migration, and invasion. Although many studies have suggested that PLCγ functions in cell mobility rather than proliferation in cancer, questions remain as to whether PLCγ regulates mitogenesis and whether PLCγ promotes or inhibits proliferation. Moreover, how PLCγ regulates cancer-associated cellular processes and the interplay among other proteins involved in cancer progression have yet to be fully elucidated. In this review, we discuss the current understanding of the role of PLCγ1 in cancer mobility and proliferation.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yu Jin Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Kyeong Jin Shin
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, Institute of Human Anatomy, University of Bologna, Bologna, Italy
| | - Young Chan Chae
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea.
| |
Collapse
|
16
|
Liu B, Pan CF, Ma T, Wang J, Yao GL, Wei K, Chen YJ. Long non‑coding RNA AK001796 contributes to cisplatin resistance of non‑small cell lung cancer. Mol Med Rep 2017; 16:4107-4112. [PMID: 29067469 DOI: 10.3892/mmr.2017.7081] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 03/30/2017] [Indexed: 11/06/2022] Open
Abstract
Cisplatin (DDP)‑based chemotherapy is the most widely used therapy for non‑small cell lung cancer (NSCLC). However, the existence of chemoresistance has become a major limitation in its efficacy. Long non‑coding RNAs (lncRNAs) have been shown to be involved in chemotherapy drug resistance. The aim of the present study was to investigate the biological role of lncRNA AK001796 in cisplatin‑resistant NSCLC A549/DDP cells. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) analysis was performed to monitor the differences in the expression of AK001796 in cisplatin-resistant (A549/DDP) cells and parental A549 cells. Cellular sensitivity to cisplatin and cell viability were examined using an MTT assay. Cell apoptosis and cell cycle distribution were measured using flow cytometry. The expression levels of cell cycle proteins cyclin C (CCNC), baculoviral IAP repeat containing 5 (BIRC5), cyclin‑dependent kinase 1 (CDK1) and G2 and S phase‑expressed 1 (GTSE1) were assessed using RT‑qPCR and western blot analyses. It was found that the expression of AK001796 was increased in A549/DDP cells, compared with that in A549 cells. The knockdown of AK001796 by small interfering RNA reduced cellular cisplatin resistance and cell viability, and resulted in cell‑cycle arrest, with a marked increase in the proportion of A549/DDP cells in the G0/G1 phase. By contrast, the knockdown of AK001796 increased the number of apoptotic cancer cells during cisplatin treatment. It was also shown that the knockdown of AK001796 positively induced the expression of cell apoptosis‑associated factors, CCNC and BIRC5, and suppressed the expression of cell cycle‑associated factors, CDK1 and GTSE5. Taken together, these findings indicated that lncRNA AK001796 increased the resistance of NSCLC cells to cisplatin through regulating cell apoptosis and cell proliferation, and thus provides an attractive therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Bin Liu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Chun-Feng Pan
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Teng Ma
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Jun Wang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Guo-Liang Yao
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Ke Wei
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| | - Yi-Jiang Chen
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Guangzhou 210029, P.R. China
| |
Collapse
|
17
|
The Zuo Jin Wan Formula Induces Mitochondrial Apoptosis of Cisplatin-Resistant Gastric Cancer Cells via Cofilin-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:8203789. [PMID: 27872653 PMCID: PMC5107242 DOI: 10.1155/2016/8203789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 11/18/2022]
Abstract
Despite the status of cisplatin (DDP) as a classical chemotherapeutic agent in the treatment of cancer, the development of multidrug resistance often leads to a failure of DDP therapy. Here we found that phosphorylated cofilin-1 (p-cofilin-1) was overexpressed in the DDP-resistant human gastric cancer cell lines SGC7901/DDP and BGC823/DDP, relative to the respective parent cell lines (SGC7901 and BGC823), and that DDP induced the dephosphorylation of p-cofilin-1 in both parent lines but not in the DDP-resistant lines. However, we noted that the traditional Chinese medicine formula Zuo Jin Wan (ZJW) could induce the dephosphorylation of p-cofilin-1 and promote cofilin-1 translocation from the cytoplasm into the mitochondria in both SGC7901/DDP and BGC823/DDP cells. This mitochondrial translocation of cofilin-1 was found to induce the conversion of filamentous actin to globular-actin, activate mitochondrial damage and calcium overloading, and induce the mitochondrial apoptosis pathway. We further observed that these effects of ZJW on DDP-resistant human gastric cancer cell lines could be reversed via transfection with cofilin-1-specific siRNA, or treatment with a PP1 and PP2A inhibitor. These results suggest that ZJW is an effective drug therapy for patients with DDP-resistant gastric cancer.
Collapse
|
18
|
Shi WJ, Gao JB. Molecular mechanisms of chemoresistance in gastric cancer. World J Gastrointest Oncol 2016; 8:673-681. [PMID: 27672425 PMCID: PMC5027022 DOI: 10.4251/wjgo.v8.i9.673] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/07/2016] [Accepted: 06/29/2016] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer is the fourth most common cancer and the second leading cause of cancer deaths worldwide. Chemotherapy is one of the major treatments for gastric cancer, but drug resistance limits the effectiveness of chemotherapy, which results in treatment failure. Resistance to chemotherapy can be present intrinsically before the administration of chemotherapy or it can develop during chemotherapy. The mechanisms of chemotherapy resistance in gastric cancer are complex and multifactorial. A variety of factors have been demonstrated to be involved in chemoresistance, including the reduced intracellular concentrations of drugs, alterations in drug targets, the dysregulation of cell survival and death signaling pathways, and interactions between cancer cells and the tumor microenvironment. This review focuses on the molecular mechanisms of chemoresistance in gastric cancer and on recent studies that have sought to overcome the underlying mechanisms of chemoresistance.
Collapse
|
19
|
Du MD, He KY, Qin G, Chen J, Li JY. Adriamycin resistance-associated prohibitin gene inhibits proliferation of human osteosarcoma MG63 cells by interacting with oncogenes and tumor suppressor genes. Oncol Lett 2016; 12:1994-2000. [PMID: 27602127 DOI: 10.3892/ol.2016.4862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/16/2016] [Indexed: 12/19/2022] Open
Abstract
The resistance of cancer cells to chemotherapeutic agents is a major obstacle for successful chemotherapy, and the mechanism of chemoresistance remains unclear. The present study developed an adriamycin-resistant human osteosarcoma MG-63 sub-line (MG-63/ADR), and identified differentially expressed proteins that may be associated with adriamycin resistance. Two dimensional gel electrophoresis, matrix-assisted laser desorption ionization time-of-flight mass spectrometry analysis and a protein identification assay were performed. Western blot analysis was used to examine the prohibitin (PHB) levels in the MG-63/ADR cells. Quantitative polymerase chain reaction was utilized to detect adriamycin resistant-associated genes. Laser-scanning confocal microscope was employed to examine the colocalization of PHB with v-myc avian myelocytomatosis viral oncogene homolog (c-myc), FBJ murine osteosarcoma viral oncogene homolog (c-fos), tumor protein p53 and retinoblastoma 1 (Rb). In addition, the full length of the open reading frame of human PHB was subcloned into a lentiviral vector pLVX-puro. The proliferative rate of MG-63 cells was also investigated. The overall protein expression in MG-63/ADR cells was clearly suppressed. Three notable protein regions, representing high mobility group box 1, Ras homolog gene family, member A, and PHB, were identified to be significantly altered in MG-63/ADR cells when compared with its parental cells. Therefore, PHB modulated the chemoresistance of MG-63/ADR cells by interacting with multiple oncogenes or tumor suppressor genes (c-myc, c-fos, p53 and Rb). In addition, overexpression of PHB decreases the proliferative rate of MG-63 cells. In conclusion, PHB is an adriamycin resistance-associated gene, which may inhibit the proliferation of human osteosarcoma MG-63 cells by interacting with the oncogenes or tumor suppressor genes, c-myc, c-fos, p53 and Rb.
Collapse
Affiliation(s)
- Min-Dong Du
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, Guangxi 530023, P.R. China
| | - Kai-Yi He
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, Guangxi 530023, P.R. China
| | - Gang Qin
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, Guangxi 530023, P.R. China
| | - Jin Chen
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, Guangxi 530023, P.R. China
| | - Jin-Yi Li
- Department of Osteoarthrosis, The First Affiliated Hospital of Guangxi Traditional Chinese Medical University, Nanning, Guangxi 530023, P.R. China
| |
Collapse
|
20
|
Jin R, Xia Y, Chen Q, Li W, Chen D, Ye H, Zhao C, Du X, Shi D, Wu J, Liang G. Da0324, an inhibitor of nuclear factor-κB activation, demonstrates selective antitumor activity on human gastric cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:979-95. [PMID: 27042000 PMCID: PMC4780725 DOI: 10.2147/dddt.s90081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The transcription factor nuclear factor-κB (NF-κB) is constitutively activated in a variety of human cancers, including gastric cancer. NF-κB inhibitors that selectively kill cancer cells are urgently needed for cancer treatment. Curcumin is a potent inhibitor of NF-κB activation. Unfortunately, the therapeutic potential of curcumin is limited by its relatively low potency and poor cellular bioavailability. In this study, we presented a novel NF-κB inhibitor named Da0324, a synthetic asymmetric mono-carbonyl analog of curcumin. The purpose of this study is to research the expression of NF-κB in gastric cancer and the antitumor activity and mechanism of Da0324 on human gastric cancer cells. Methods The expressions between gastric cancer tissues/cells and normal gastric tissues/cells of NF-κB were evaluated by Western blot. The inhibition viability of compounds on human gastric cancer cell lines SGC-7901, BGC-823, MGC-803, and normal gastric mucosa epithelial cell line GES-1 was assessed with the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Absorption spectrum method and high-performance liquid chromatography method detected the stability of the compound in vitro. The compound-induced changes of inducible NF-κB activation in the SGC-7901 and BGC-823 cells were examined by Western blot analysis and immunofluorescence methods. The antitumor activity of compound was performed by clonogenic assay, matrigel invasion assay, flow cytometric analysis, Western blot analysis, and Hoechst 33258 staining assay. Results High levels of p65 were found in gastric cancer tissues and cells. Da0324 displayed higher growth inhibition against several types of gastric cancer cell lines and showed relatively low toxicity to GES-1. Moreover, Da0324 was more stable than curcumin in vitro. Western blot analysis and immunofluorescence methods showed that Da0324 blocked NF-κB activation. In addition, Da0324 significantly inhibited tumor proliferation and invasion, arrested the cell cycle, and induced apoptosis in vitro. Conclusion The asymmetric mono-carbonyl analog of curcumin Da0324 exhibited significantly improved antigastric cancer activity. Da0324 may be a promising NF-κB inhibitor for the selective targeting of cancer cells. However, further studies are needed in animals to validate these findings for the therapeutic use of Da0324.
Collapse
Affiliation(s)
- Rong Jin
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Department of Epidemiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiuxiang Chen
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wulan Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; College of Information Science and Computer Engineering, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Dahui Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hui Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China; School of Basic Medical Sciences, Wenzhou Medical College, Wenzhou, People's Republic of China
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiaojing Du
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Dengjian Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jianzhang Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
21
|
Huang D, Duan H, Huang H, Tong X, Han Y, Ru G, Qu L, Shou C, Zhao Z. Cisplatin resistance in gastric cancer cells is associated with HER2 upregulation-induced epithelial-mesenchymal transition. Sci Rep 2016; 6:20502. [PMID: 26846307 PMCID: PMC4742832 DOI: 10.1038/srep20502] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
Cisplatin remains to be primary chemotherapeutic drug for gastric cancer patients, especially for advanced stage ones. However, primary or acquired resistance often occurs with the mechanisms being not well understood, which results in relapse of the cancer and poor survival. Herein, we found that HER2 upregulation was associated with cisplatin resistance. We observed that cisplatin-resistant gastric cancer cells underwent a morphological change similar to epithelial-mesenchymal transition (EMT) which is mediated by HER2 overexpression. When specific monoclonal antibody Herceptin, small molecular targeted drug CP724714, or small interfering RNA against HER2 was applied, the EMT-like phenotypic change was dramatically reversed. More importantly, the IC50 and Resistance Index of resistant gastric cancer cells to cisplatin were also decreased by any of these treatments.We demonstrated that expression and amplification of HER2 positively correlated with expression of EMT-related transcription factor Snail in gastric cancer tissues. Furthermore, for the first time, we found that HER2/Snail double positive gastric cancer patients had poorer survival than single positive or double negative counterparts, which provided experimental evidence for the necessity of HER2/Snail double testing in gastric cancer. In conclusion, this study provides some clues of the association of cisplatin resistance with HER2 upregulation-induced EMT in gastric cancer cells.
Collapse
Affiliation(s)
- Dongsheng Huang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Hongying Duan
- Clinical Research Institute, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China.,Department of Pathology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Hao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital &Institute, Beijing 100142, China
| | - Xiangmin Tong
- Clinical Research Institute, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Yong Han
- Department of Pathology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Like Qu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital &Institute, Beijing 100142, China
| | - Chengchao Shou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital &Institute, Beijing 100142, China
| | - Zhongsheng Zhao
- Department of Pathology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| |
Collapse
|
22
|
Wang WJ, Wang Y, Hou PP, Li FW, Zhou B, Chen HZ, Bian XL, Cai QX, Xing YZ, He JP, Zhang H, Huang PQ, Lin T, Wu Q. Induction of Autophagic Death in Cancer Cells by Agonizing TR3 and Attenuating Akt2 Activity. CHEMISTRY & BIOLOGY 2015; 22:1040-1051. [PMID: 26235054 DOI: 10.1016/j.chembiol.2015.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 05/27/2015] [Accepted: 06/15/2015] [Indexed: 11/16/2022]
Abstract
Apoptotic resistance is becoming a significant obstacle for cancer therapy as the majority of treatment takes the route of apoptotic induction. It is of great importance to develop an alternative strategy to induce cancer cell death. We previously reported that autophagic cell death mediated by nuclear receptor TR3 and driven by a chemical agonist, 1-(3,4,5-trihydroxyphenyl)nonan-1-one (THPN), is highly effective in the therapy of melanoma but not any other cancer types. Here, we discovered that the insensitivity of cancer cells to THPN originated from a high cellular Akt2 activity. Akt2 phosphorylation interferes with TR3 export to cytoplasm and targeting to mitochondria, which lead to the autophagic induction. Therefore, the TR3-mediated autophagy could be effectively induced in the otherwise insensitive cells by downregulating Akt2 activity. Highly effective antineoplastic compounds are developed through optimizing the structure of THPN. This study implicates a general strategy for cancer therapy by the induction of autophagic cell death.
Collapse
Affiliation(s)
- Wei-jia Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Yuan Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Pei-pei Hou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Feng-wei Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Bo Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Hang-zi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Xue-li Bian
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Qi-xu Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Yong-zhen Xing
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Jian-ping He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Hongkui Zhang
- Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, Fujian Province, P.R. China
| | - Pei-qiang Huang
- Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, Fujian Province, P.R. China
| | - Tianwei Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China.
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China.
| |
Collapse
|
23
|
Zheng Z, Liu B, Wu X. RhoGDI2 up-regulates P-glycoprotein expression via Rac1 in gastric cancer cells. Cancer Cell Int 2015; 15:41. [PMID: 25901126 PMCID: PMC4404694 DOI: 10.1186/s12935-015-0190-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/28/2015] [Indexed: 11/21/2022] Open
Abstract
Multidrug resistance (MDR) is a major clinical obstacle in treatment of gastric cancer. Previously, using 2D electrophoresis-mass spectrometry, we identified RhoGDI2 as a contributor to 5-FU resistance in colon cancer cells, and also confer gastric cancer cells resistance to 5-FU. Here, we reported RhoGDI2 also induced MDR in gastric cancer cell line (MKN-45). To explore the underlining mechanism, we detected the mRNA, protein expression, activity of P-glycoprotein (P-gp) in MKN-45 stably transfected with RhoGDI2 expressing or control vector. All the mRNA, protein level, activity were increased by 130%, 230%, 35% respectively after ectopic expression of RhoGDI2. RhoGDI2 was correlated with P-gp expression in gastric cancer tissues as detected by immunohistochemistry. To further study how RhoGDI2 up-regulates P-gp expression, we tested the activity of Rac1 in MKN-45/RhoGDI2 and MKN-45/GFP. Ectopic expression of RhoGDI2 increased Rac1 activity (P < 0.05). For more important, silencing of Rac1 expression by siRNA decreased P-gp expression to undetectable level. Overall, these findings suggest that RhoGDI2 up-regulates P-gp expression via Rac1 to induce MDR.
Collapse
Affiliation(s)
- Zhong Zheng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032 People's Republic of China
| | - Bingya Liu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, and Gastroenterology, Ruijin Hospital, Jiaotong University School of Medicine, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai, 200032 People's Republic of China
| |
Collapse
|
24
|
Cho HJ, Park SM, Kim IK, Nam IK, Baek KE, Im MJ, Yoo JM, Park SH, Ryu KJ, Han HT, Kim HJ, Hong SC, Kim KD, Pak Y, Kim JW, Lee CW, Yoo J. RhoGDI2 promotes epithelial-mesenchymal transition via induction of Snail in gastric cancer cells. Oncotarget 2015; 5:1554-64. [PMID: 24721928 PMCID: PMC4039231 DOI: 10.18632/oncotarget.1733] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) expression correlates with tumor growth, metastasis, and chemoresistance in gastric cancer. Here, we show that RhoGDI2 functions in the epithelial-mesenchymal transition (EMT), which is responsible for invasiveness during tumor progression. This tumorigenic activity is associated with repression of E-cadherin by RhoGDI2 via upregulation of Snail. Overexpression of RhoGDI2 induced phenotypic changes consistent with EMT in gastric cancer cells, including abnormal epithelial cell morphology, fibroblast-like properties, and reduced intercellular adhesion. RhoGDI2 overexpression also resulted in decreased expression of the epithelial markers E-cadherin and β-catenin and increased expression of the mesenchymal markers vimentin and fibronectin. Importantly, RhoGDI2 overexpression also stimulated the expression of Snail, a repressor of E-cadherin and inducer of EMT, but not other family members such as Slug or Twist. RNA interference-mediated knockdown of Snail expression suppressed RhoGDI2-induced EMT and invasion, confirming that the effect was Snail-specific. These results indicate that RhoGDI2 plays a critical role in tumor progression in gastric cancer through induction of EMT. Targeting RhoGDI2 may thus be a useful strategy to inhibit gastric cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Hee Jun Cho
- Division of Applied Life Science (BK21 plus), Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
14-3-3σ attenuates RhoGDI2-induced cisplatin resistance through activation of Erk and p38 in gastric cancer cells. Oncotarget 2014; 4:2045-56. [PMID: 24185104 PMCID: PMC3875768 DOI: 10.18632/oncotarget.1334] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) promotes tumor growth and malignant progression and enhances chemoresistance of gastric cancer. Recently, we noted an inverse correlation between RhoGDI2 and 14-3-3σ expression, which suggests that 14-3-3σ is a target of gastric cancer metastasis and the chemoresistance-promoting effect of RhoGDI2. Herein, we evaluated whether 14-3-3σ is regulated by RhoGDI2 and is functionally important for the RhoGDI2-induced cisplatin resistance of gastric cancer cells. We used highly metastatic and cisplatin-resistant RhoGDI2-overexpressing SNU-484 cells and observed decreased 14-3-3σ mRNA and protein expression. Depletion of 14-3-3σ in SNU-484 control cells enhanced cisplatin resistance, whereas restoration of 14-3-3σ in RhoGDI2-overexpressing SNU-484 cells impaired cisplatin resistance in vitro and in vivo. We also found that the phosphorylation levels of Erk and p38 kinases significantly decreased in RhoGDI2-overexpressing SNU-484 cells and recovered after 14-3-3σ expression, and that decreased activities of these kinases were critical for RhoGDI2-induced cisplatin resistance. In conclusion, 14-3-3σ is a RhoGDI2-regulated gene that appears to be important for suppressing the chemoresistance of gastric cancer cells.
Collapse
|
26
|
Silencing of HIF-1α enhances the radiation sensitivity of human glioma growth in vitro and in vivo. Neuropharmacology 2014; 89:168-74. [PMID: 24859610 DOI: 10.1016/j.neuropharm.2014.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 11/20/2022]
Abstract
Gliomas are the leading cause of cancer-related mortality worldwide, and the incidence is increasing. Because gliomas often become resistant to radiation treatment, it is urgent to develop novel therapeutic methods that are more effective and less toxic than current therapies so as to enhance patient survival and quality of life. Effective enhancement of radiation therapy for gliomas in vivo and in vitro was observed upon silencing of hypoxia-inducible factor 1α (HIF-1α) with RNA interference; this enhancement was related to changes in the cell cycle and apoptosis that were accompanied by modulation of Cdc2, cyclin B1, and Bcl-2 expression. Our data suggest that HIF-1α silencing combined with radiation therapy will increase the therapeutic efficacy of glioma treatment via regulation of cell cycle and apoptosis-related signaling pathways.
Collapse
|
27
|
Xu W, Hu X, Chen Z, Zheng X, Zhang C, Wang G, Chen Y, Zhou X, Tang X, Luo L, Xu X, Pan W. Normal fibroblasts induce E-cadherin loss and increase lymph node metastasis in gastric cancer. PLoS One 2014; 9:e97306. [PMID: 24845259 PMCID: PMC4028202 DOI: 10.1371/journal.pone.0097306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/16/2014] [Indexed: 12/15/2022] Open
Abstract
Background A tumor is considered a heterogeneous complex in a three-dimensional environment that is flush with pathophysiological and biomechanical signals. Cell-stroma interactions guide the development and generation of tumors. Here, we evaluate the contributions of normal fibroblasts to gastric cancer. Methodology/Principal Findings By coculturing normal fibroblasts in monolayers of BGC-823 gastric cancer cells, tumor cells sporadically developed short, spindle-like morphological characteristics and demonstrated enhanced proliferation and invasive potential. Furthermore, the transformed tumor cells demonstrated decreased tumor formation and increased lymphomatic and intestinal metastatic potential. Non-transformed BGC-823 cells, in contrast, demonstrated primary tumor formation and delayed intestinal and lymph node invasion. We also observed E-cadherin loss and the upregulation of vimentin expression in the transformed tumor cells, which suggested that the increase in metastasis was induced by epithelial-to-mesenchymal transition. Conclusion Collectively, our data indicated that normal fibroblasts sufficiently induce epithelial-to-mesenchymal transition in cancer cells, thereby leading to metastasis.
Collapse
Affiliation(s)
- Wen Xu
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xinlei Hu
- Department of Orthopedics, Second Affiliated Hospital (Binjiang Branch) of Zhejiang University, School of Medicine, Hangzhou, China
| | - Zhongting Chen
- Department of Pharmacy, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaoping Zheng
- Department of Pathology, Qujiang People’s Hospital, Quzhou, China
| | - Chenjing Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Gang Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yu Chen
- Zhejiang Academy of Traditional Chinese Medicine, Experimental Animal Research Center, Hangzhou, China
| | - Xinglu Zhou
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaoxiao Tang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Laisheng Luo
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiang Xu
- Department of Pharmacy, Second Affiliated Hospital (Binjiang Branch) of Zhejiang University, School of Medicine, Hangzhou, China
| | - Wensheng Pan
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
- Department of Gastroenterology, Second Affiliated Hospital (Binjiang Branch) of Zhejiang University, School of Medicine, Hangzhou, China
- * E-mail:
| |
Collapse
|
28
|
Ni S, Zhao J, Fu Z, Liu H. Lentivirus vector-mediated Rho guanine nucleotide dissociation inhibitor 2 induces beta-2 adrenergic receptor desensitization in β2AR desensitization mice model. J Thorac Dis 2014; 6:118-25. [PMID: 24605225 DOI: 10.3978/j.issn.2072-1439.2013.12.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/28/2013] [Indexed: 11/14/2022]
Abstract
BACKGROUND It is well-known that chronic administration of β2AR agonists can induce β2AR desensitization. Our previous study showed that Rho guanine nucleotide dissociation inhibitor 2 (RhoGDI2) overexpression induced beta-2 adrenergic receptor (β2AR) desensitization in airway smooth muscle cells. The purpose of this study was to further study the function of RhoGDI2 in β2AR desensitization by β2AR desensitization mouse model. METHODS Studies were performed using a β2AR desensitization mice model induced by salbutamol. The mice were randomly divided into five groups (n=45): RhoGDI2 overexpression group (n=10); RhoGDI2 siRNA group (n=10); empty viral vector group (n=10); experimental control group (n=10); blank control group-without any drug treatment (n=5). The first four groups were used the same methods and the same dose to establish β2AR desensitization mice model by salbutamol. The first three groups that salbutamol-treated were used for intratracheal delivery of lentiviral vectors. Airway hyperreactivity was measured through a whole-body plethysmograph system. RhoGDI2, β2AR, GRK2 mRNA and protein expression levels were then detected by RT-PCR and western blot analyses in fresh lung tissues. As well as the activity of GRK was assessed by light-dependent phosphorylation of rhodopsin. RESULTS We successfully constructed β2AR desensitization mouse model. As expected, airway responsiveness after inhaling acetylcholine chloride (Ach) was markedly increased in the RhoGDI2 overexpression group compared to experimental control group and blank control group when concentrations of Ach was 45 mg/mL (all P<0.05), while, it was markedly decreased in the RhoGDI2 siRNA group compared to experimental control group (P<0.05). RhoGDI2, GRK2 expressions and GRK enzymatic activity were significantly increased in RhoGDI2 overexpression group compared to experimental control group and blank control group (all P<0.05). RhoGDI2, GRK2 expressions and GRK enzymatic activity were significantly decreased in RhoGDI2 siRNA group compared to experimental control group and blank control group (all P<0.05). Conversely, β2AR expression were significantly lower in RhoGDI2 overexpression group compared to experimental control group and blank control group (all P<0.05), exhibiting an inverse correlation with RhoGDI2 expression. CONCLUSIONS To sum up, our present studies found that RhoGDI2 might induce β2AR desensitization and GRK2 might take part in RhoGDI2-mediated β2AR desensitization.
Collapse
Affiliation(s)
- Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jing Zhao
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhenxue Fu
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Hua Liu
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
29
|
Cho HJ, Kim IK, Park SM, Baek KE, Nam IK, Park SH, Ryu KJ, Choi J, Ryu J, Hong SC, Jeong SH, Lee YJ, Ko GH, Kim J, Won Lee C, Soo Kang S, Yoo J. VEGF-C mediates RhoGDI2-induced gastric cancer cell metastasis and cisplatin resistance. Int J Cancer 2014; 135:1553-63. [PMID: 24585459 DOI: 10.1002/ijc.28801] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 02/10/2014] [Indexed: 12/28/2022]
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) expression is correlated with tumor growth, metastasis and chemoresistance in gastric cancer. However, the mechanisms by which RhoGDI2 promotes tumor cell survival and metastasis remain unclear. In this study, we clearly demonstrate that RhoGDI2 upregulates VEGF-C expression and RhoGDI2 expression is positively correlated with VEGF-C expression in human gastric tumor tissues as well as parental gastric cancer cell lines. VEGF-C depletion suppressed RhoGDI2-induced gastric cancer metastasis and sensitized RhoGDI2-overexpressing cells to cisplatin-induced apoptosis in vitro and in vivo. Secreted VEGF-C enhanced gastric cancer cell invasion and conferred cisplatin resistance to RhoGDI2-overexpressing cells. We also show that RhoGDI2 positively regulates Rac1 activity in gastric cancer cells. Inhibition of Rac1 expression suppressed RhoGDI2-induced VEGF-C expression, and this inhibition was associated with decreased invasiveness and increased sensitivity to cisplatin in RhoGDI2-overexpressing cells. Our results indicate that RhoGDI2 might be a potential therapeutic target for simultaneously reducing metastasis risk and enhancing chemotherapy efficacy in gastric cancer.
Collapse
|
30
|
Abstract
RhoGDI2 is a guanine nucleotide dissociation inhibitor (GDI) specific for the Rho family of small GTPases that plays dual opposite roles in tumor progression, being both a promoter in tissues such as breast and a metastasis suppressor in tissues such as the bladder. Despite a clear role for this protein in modulating the invasive and metastatic process, the mechanisms through which RhoGDI2 executes these functions remain unclear. This review will highlight the current state of our knowledge regarding how RhoGDI2 functions in metastasis with a focus on bladder cancer and will also seek to highlight other potential underappreciated avenues through which this protein may affect cancer cell behavior.
Collapse
Affiliation(s)
- Erin M Griner
- Center for Cell Signaling and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
31
|
Zheng Z, He XY, Li JF, Yu BQ, Chen XH, Ji J, Zhang JN, Gu QL, Zhu ZG, Liu BY. RhoGDI2 confers resistance to 5-fluorouracil in human gastric cancer cells. Oncol Lett 2012; 5:255-260. [PMID: 23255931 DOI: 10.3892/ol.2012.949] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/08/2012] [Indexed: 11/06/2022] Open
Abstract
Resistance to 5-fluorouracil (5-FU) in patients with gastric cancer is a serious therapeutic problem and major efforts are underway to understand the underlying mechanisms. We have previously identified RhoGDI2 as a contributor to 5-FU resistance in colon cancer cells using 2D electrophoresis and mass spectrometry and the current study aimed to further investigate this role. The expression of RhoGDI2 in seven gastric cancer cell lines was positively correlated with resistance to 5-FU. Lower 5-FU sensitivity of isolated tumor cells from patients with gastric cancer was also associated with higher RhoGDI2 expression. Ectopic expression of RhoGDI2 in gastric cancer cells increased the resistance to 5-FU and reverted low dose 5-FU-induced G2/M phase arrest without affecting the population of sub-G1 cells. Overall, these findings suggest that RhoGDI2 is associated with 5-FU resistance and is a potential therapeutic target for enhancing chemotherapy efficacy in gastric cancer.
Collapse
Affiliation(s)
- Zhong Zheng
- Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Xu HY, Chen ZW, Pan YM, Fan L, Guan J, Lu YY. Transfection of PDCD5 effect on the biological behavior of tumor cells and sensitized gastric cancer cells to cisplatin-induced apoptosis. Dig Dis Sci 2012; 57:1847-56. [PMID: 22359193 DOI: 10.1007/s10620-012-2090-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 02/06/2012] [Indexed: 12/09/2022]
Abstract
BACKGROUND Programmed cell death 5 (PDCD5) expression is reduced in various human tumor cells, and the protein concentration and nuclear translocation of PDCD5 is also observed during tumor cell apoptosis. AIMS The purpose of this study was to investigate the differential expression of PDCD5 in six gastric cell lines, and to explore the changes of biological behavior mechanism underlying enhanced apoptosis-inducing effects of cisplatin by PDCD5 over-expression on gastric cancer BGC823 cells. METHODS RT-PCR and real-time PCR were used to determine PDCD5 expression. BGC823/PDCD5 cells were assessed the cellular proliferating ability by MTT assay, soft agar cloning experiments and tumorigenicity in nude mice experiments in vivo. The effects of cisplatin in combination with PDCD5 on the proliferation and apoptosis were measured by MTT, Annexin-V-FITC/PI dual labeling and cell cycle analysis, respectively. Immunofluorescence was used to detect co-localization of p53 and PDCD5 protein to explore the mechanism underlying the synergistic therapeutic effect of PDCD5 with cisplatin (5 μg/ml for 24 h). RESULTS PDCD5 had the highest expression level in the GES1 cell among other cell lines. The growths of BGC823 cells transfected with PDCD5 for six (6th) or 17 (17th) days were both slower than that of BGC823 and BGC823/Neo (P < 0.01). The stable transfection of PDCD5 demonstrated G2/M cell cycle arrest, increased apoptosis and nuclear translocation of PDCD5 and p53 after cisplatin treatment. CONCLUSIONS Stable transfection of the PDCD5 gene can inhibit the growth of the BGC823 cell line and notably improve apoptosis-inducing effects of cisplatin, indicating a novel strategy for better chemotherapeutic effects on gastric cancer.
Collapse
Affiliation(s)
- Hui-Yu Xu
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang, People's Republic of China
| | | | | | | | | | | |
Collapse
|
33
|
Yuan X, Zhang B, Chen N, Chen XY, Liu LL, Zheng QS, Wang ZP. Isoliquiritigenin treatment induces apoptosis by increasing intracellular ROS levels in HeLa cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2012; 14:789-798. [PMID: 22694287 DOI: 10.1080/10286020.2012.694873] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study focuses on the relationship between the apoptosis induced by isoliquiritigenin (ISL) and the production of reactive oxygen species (ROS). Cell viability was evaluated using sulforhodamine B assay. The apoptotic rate was determined via flow cytometry. Intracellular ROS level was assessed using the 2,7-dichlorofluorescein probe assay. Poly-ADP-ribose polymerase (PARP) protein expression was examined using Western blot analysis. The results showed that ISL treatment inhibited cell proliferation by inducing apoptosis. The increased apoptotic rate and ROS production induced by ISL were inhibited by the co-treatment of ISL and free radical scavenger N-acetyl-cysteine (NAC), catalase (CAT), and 4,5-dihydroxyl-1,3-benzededisulfonic acid (Tiron). On the contrary, the increased apoptotic rate and the ROS production were compensated by the co-treatment of ISL and l-buthionine-(S,R)-sulfoximine (BSO). ISL treatment increased the degradation of PARP, which was counteracted by antioxidants (NAC or CAT), whereas the combination treatment of ISL and pro-oxidant (BSO) enhanced the PARP degradation induced by ISL. Our findings suggested that ISL treatment induced apoptosis by increasing intracellular ROS levels in HeLa cells.
Collapse
Affiliation(s)
- Xuan Yuan
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730000, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Takeba Y, Matsumoto N, Watanabe M, Takenoshita-Nakaya S, Ohta Y, Kumai T, Takagi M, Koizumi S, Asakura T, Otsubo T. The Rho kinase inhibitor fasudil is involved in p53-mediated apoptosis in human hepatocellular carcinoma cells. Cancer Chemother Pharmacol 2012; 69:1545-55. [DOI: 10.1007/s00280-012-1862-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 03/12/2012] [Indexed: 01/10/2023]
|
35
|
Cho HJ, Baek KE, Kim IK, Park SM, Choi YL, Nam IK, Park SH, Im MJ, Yoo JM, Ryu KJ, Oh YT, Hong SC, Kwon OH, Kim JW, Lee CW, Yoo J. Proteomics-based strategy to delineate the molecular mechanisms of RhoGDI2-induced metastasis and drug resistance in gastric cancer. J Proteome Res 2012; 11:2355-64. [PMID: 22364609 DOI: 10.1021/pr2011186] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) was initially identified as a regulator of the Rho family of GTPases. Our recent works suggest that RhoGDI2 promotes tumor growth and malignant progression, as well as enhances chemoresistance in gastric cancer. Here, we delineate the mechanism by which RhoGDI2 promotes gastric cancer cell invasion and chemoresistance using two-dimensional gel electrophoresis (2-DE) on proteins derived from a RhoGDI2-overexpressing SNU-484 human gastric cancer cell line and control cells. Differentially expressed proteins were identified using matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF-MS). In total, 47 differential protein spots were identified; 33 were upregulated, and 14 were downregulated by RhoGDI2 overexpression. Upregulation of SAE1, Cathepsin D, Cofilin1, CIAPIN1, and PAK2 proteins was validated by Western blot analysis. Loss-of-function analysis using small interference RNA (siRNA) directed against candidate genes reveals the need for CIAPIN1 and PAK2 in RhoGDI2-induced cancer cell invasion and Cathepsin D and PAK2 in RhoGDI2-mediated chemoresistance in gastric cancer cells. These data extend our understanding of the genes that act downstream of RhoGDI2 during the progression of gastric cancer and the acquisition of chemoresistance.
Collapse
Affiliation(s)
- Hee Jun Cho
- Department of Microbiology/Research Institute of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
PLCγ is required for RhoGDI2-mediated cisplatin resistance in gastric cancer. Biochem Biophys Res Commun 2011; 414:575-80. [PMID: 21986528 DOI: 10.1016/j.bbrc.2011.09.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/23/2023]
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) is a regulator of the Rho family GTPases. Recent work from our laboratory suggests that RhoGDI2 expression potentially enhances resistance to cisplatin as well as promotes tumor growth and malignant progression in gastric cancer. In this study, we demonstrate that phospholipase C-gamma (PLCγ) is required for RhoGDI2-mediated cisplatin resistance and cancer cell invasion in gastric cancer. The levels of phosphorylated PLCγ are markedly enhanced in RhoGDI2-overexpressing SNU-484 cells and, by contrast, repressed in RhoGDI2-depleted MKN-28 cells. Depletion of PLCγ expression or inhibition of its activity not only significantly increases cisplatin-induced apoptosis but also suppresses the invasive ability of RhoGDI2-overexpressing SNU-484 cells. Taken together, our results suggest that PLCγ plays a key role in RhoGDI2-mediated cisplatin resistance and cell invasion in gastric cancer cells.
Collapse
|
37
|
Rufini A, Melino G. Cell death pathology: the war against cancer. Biochem Biophys Res Commun 2011; 414:445-50. [PMID: 21971555 DOI: 10.1016/j.bbrc.2011.09.110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 09/21/2011] [Indexed: 12/25/2022]
Abstract
Programmed cell death was a fundamental discovery, awarded with the Nobel price in 2002 to Sulston, Brenner and Horvitz. Since then it has been clear that alteration of apoptotic pathways is a common feature of tumors, enabling cancer cells to survive chemotherapeutic interventions. Thus, apoptosis is an attractive target in cancer therapy, with the aim to revert the cancer-related alterations of the cell death machinery. Here, we overview the fundamental apoptotic pathways and summarize the attempts to target apoptosis to restore cell death in cancer cells with a special focus on the p53-family and autophagy.
Collapse
|