1
|
Lin Z, Lin J. Circ_0004585 Facilitates Tumorigenesis of Colorectal Cancer Via Modulating the miR-338-3p/ZFX Axis and Activating the MEK/ERK Pathway. Cell Mol Bioeng 2023; 16:159-171. [PMID: 37096071 PMCID: PMC10121944 DOI: 10.1007/s12195-022-00756-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 12/06/2022] [Indexed: 01/22/2023] Open
Abstract
Background Colorectal cancer (CRC) is a common malignant tumor in the digestive tract. Circular RNAs (circRNAs) have been identified as crucial regulators of tumorigenesis. However, the role and potential mechanism of circ_0004585 in CRC are poorly understood. Methods The expression of circ_0004585, microRNA-338-3p (miR-338-3p), and zinc finger protein X-linked (ZFX) was detected by quantitative real-time PCR and Western blot. Cell proliferation, cell cycle arrest, apoptosis, and angiogenesis were evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), 5-Ethynyl-2'-deoxyuridine (EdU), flow cytometry and tube formation assays. Western blot assay was applied to detect the expression of epithelial-mesenchymal transition (EMT)-related proteins and MEK/ERK signaling pathway-related proteins. A xenograft model was used to analyze tumor growth in vivo. The targeted relationship between miR-338-3p and circ_0004585/ZFX was verified by a dual-luciferase reporter assay. Results Circ_0004585 and ZFX were up-regulated, while miR-338-3p was down-regulated in CRC tissues and cells. Silencing of circ_0004585 inhibited proliferation, angiogenesis, and EMT and triggered apoptosis in CRC cells. Consistently, circ_0004585 depletion blocked tumor growth in vivo. Circ_0004585 contributed to CRC cell development via sequestering miR-338-3p. Also, miR-338-3p hindered the malignant progression of CRC cells by targeting ZFX. Circ_0004585 activated MEK/ERK pathway via regulating ZFX. Conclusion Circ_0004585 facilitated CRC progression through modulating miR-338-3p/ZFX/MEK/ERK pathway, which might provide a potential therapeutic target for CRC. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00756-6.
Collapse
Affiliation(s)
- Zenghai Lin
- Department of General Surgery, Guangdong Province, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou City, 515041 Guangdong China
| | - Jianwei Lin
- Department of General Surgery, Guangdong Province, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou City, 515041 Guangdong China
| |
Collapse
|
2
|
Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma. Biomolecules 2023; 13:biom13020398. [PMID: 36830767 PMCID: PMC9953423 DOI: 10.3390/biom13020398] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a cellular process that has been linked to the promotion of aggressive cellular features in many cancer types. It is characterized by the loss of the epithelial cell phenotype and a shift to a more mesenchymal phenotype and is accompanied by an associated change in cell markers. EMT is highly complex and regulated via multiple signaling pathways. While the importance of EMT is classically described for carcinomas-cancers of epithelial origin-it has also been clearly demonstrated in non-epithelial cancers, including osteosarcoma (OS), a primary bone cancer predominantly affecting children and young adults. Recent studies examining EMT in OS have highlighted regulatory roles for multiple proteins, non-coding nucleic acids, and components of the tumor micro-environment. This review serves to summarize these experimental findings, identify key families of regulatory molecules, and identify potential therapeutic targets specific to the EMT process in OS.
Collapse
|
3
|
Zhang J, Wang Z, Liang Z, Jin C, Shi Y, Fan M, Hu X, Wan Y. NFIC1 inhibits the migration and invasion of MDA-MB-231 cells through S100A2-mediated inactivation of MEK/ERK pathway. Arch Biochem Biophys 2023; 734:109497. [PMID: 36574914 DOI: 10.1016/j.abb.2022.109497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
NFIC is a potent transcriptional factor involved in many physiological and pathological processes, including tumorigenesis. However, the role of NFIC1, the longest isoform of NFIC, in the progression of triple negative breast cancer (TNBC) remains elusive. Our study demonstrates that overexpression of NFIC1 inhibits the migration and invasion of TNBC MDA-MB-231 cells. NFIC1 regulates the expression of S100A2, and knockdown of S100A2 reverses the inhibitive effects of NFIC1 on the migration and invasion of MDA-MB-231 cells. Furthermore, knockdown of S100A2 activates the MEK/ERK signaling transduction pathway that is inhibited by NFIC1 overexperssion. Treatment with MEK/ERK pathway inhibitor, U0126, abolishes the effects of S100A2 knockdown. In addition, overexpression of NFIC1 in MDA-MB-231 cells increases the expression of epithelial markers and decreases the expression of mesenchymal markers, and these effects could also be reversed by knockdown of S100A2. Collectively, these results demonstrate that NFIC1 inhibits the Epithelial-mesenchymal transition (EMT) of MDA-MB-231 cells by regulating S100A2 expression, which suppress the activation of MEK/ERK pathway. Therefore, our study confirms the role of NFIC1 as a tumor repressor in TNBC, and reveals the molecular mechanism through which NFIC1 inhibits the migration and invasion of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Jing Zhang
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China; School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Zhaoying Wang
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China; School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Zehua Liang
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China
| | - Chanjuan Jin
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China
| | - Yueru Shi
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China
| | - Mingyue Fan
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China; School of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Xin Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China
| | - Youzhong Wan
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China.
| |
Collapse
|
4
|
He H, Zhang S, Yang H, Xu P, Kutschick I, Pfeffer S, Britzen-Laurent N, Grützmann R, Fu D, Pilarsky C. Identification of Genes Associated with Liver Metastasis in Pancreatic Cancer Reveals PCSK6 as a Crucial Mediator. Cancers (Basel) 2022; 15:cancers15010241. [PMID: 36612240 PMCID: PMC9818395 DOI: 10.3390/cancers15010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Liver metastasis occurs frequently in patients with pancreatic cancer. We analyzed the molecular profiling in liver metastatic lesions aiming to uncover novel genes responsible for tumor progression. Bioinformatics analysis was applied to identify genes directing liver metastasis. CRISPR/Cas9 technology was used to knock out the candidate gene. Proliferation assays, colony formation assays, cell cycle analysis, migration assays, wound healing assays, Immunofluorescence analysis, and the tumor xenograft model of intrasplenic injection were adopted to evaluate the effects of PCSK6 inactivation on cell growth, migration and liver metastasis. GSEA and Western blot were used to investigate the corresponding signaling pathway. PCSK6 was one of the obtained liver-metastasis-related genes in pancreatic cancer. PCSK6 inactivation inhibited cell growth and cell migration, due to G0/G1 cell cycle arrest and the remodeling of cell-cell junctions or the cell skeleton, respectively. PCSK6 inactivation led to fewer counts and lower outgrowth rates of liver metastatic niches in vivo. The Raf-MEK1/2-ERK1/2 axis was repressed by PCSK6 inactivation. Accordingly, we found PCSK6 inactivation could inhibit cell growth, cell migration, and liver metastasis, and explored the role of the Raf-MEK1/2-ERK1/2 axis in PCSK6 inactivation. PCSK6-targeted therapy might represent a novel approach for combatting liver metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Hang He
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Shuman Zhang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Hai Yang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Pengyan Xu
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Isabella Kutschick
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Susanne Pfeffer
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Nathalie Britzen-Laurent
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Correspondence:
| |
Collapse
|
5
|
Marles H, Biddle A. Cancer stem cell plasticity and its implications in the development of new clinical approaches for oral squamous cell carcinoma. Biochem Pharmacol 2022; 204:115212. [PMID: 35985402 DOI: 10.1016/j.bcp.2022.115212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
Oral squamous cell carcinoma (SCC) represents a major worldwide disease burden, with high rates of recurrence and metastatic spread following existing treatment methods. Populations of treatment resistant cancer stem cells (CSCs) are well characterised in oral SCC. These populations of CSCs engage the cellular programme known as epithelial mesenchymal transition (EMT) to enhance metastatic spread and therapeutic resistance. EMT is characterised by specific morphological changes and the expression of certain cell surface markers that represent a transition from an epithelial phenotype to a mesenchymal phenotype. This process is regulated by several cellular pathways that interact both horizontally and hierarchically. The cellular changes in EMT occur along a spectrum, with sub-populations of cells displaying both epithelial and mesenchymal features. The unique features of these CSCs in terms of their EMT state, cell surface markers and metabolism may offer new druggable targets. In addition, these features could be used to identify more aggressive disease states and the opportunity to personalise therapy depending on the presence of certain CSC sub-populations.
Collapse
Affiliation(s)
- Henry Marles
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Adrian Biddle
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
6
|
Liu W, Meng J, Su R, Shen C, Zhang S, Zhao Y, Liu W, Du J, Zhu S, Li P, Wang Z, Li X. SP1-mediated up-regulation of lncRNA TUG1 underlines an oncogenic property in colorectal cancer. Cell Death Dis 2022; 13:433. [PMID: 35508523 PMCID: PMC9068916 DOI: 10.1038/s41419-022-04805-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022]
Abstract
The long non-coding RNA (lncRNA) taurine up-regulated gene 1 (TUG1) acts as tumor-promoting factor in colorectal cancer (CRC). We aimed to elucidate the mechanism by which the transcription factor specificity protein 1 (SP1) regulates TUG1 and microRNAs (miRs)/mRNAs in the context of CRC, which has not been fully studied before. Expression patterns of TUG1 and SP1 were determined in clinical CRC samples and cells, followed by identification of their interaction. Next, the functional significance of TUG1 in CRC was investigated. An in vivo CRC model was established to validate the effect of TUG1. The results demonstrated that TUG1 and SP1 were highly-expressed in CRC, wherein SP1 bound to the TUG1 promoter and consequently, positively regulated its expression. Silencing of TUG1 caused suppression of CRC cell growth and promotion of cell apoptosis. TUG1 could bind to miR-421 to increase KDM2A expression, a target gene of miR-421. TUG1 could activate the ERK pathway by impairing miR-421-targeted inhibition of KDM2A. Additionally, SP1 could facilitate the tumorigenesis of CRC cells in vivo by regulating the TUG1/miR-421/KDM2A/ERK axis. Altogether, the current study emphasizes the oncogenic role of TUG1 in CRC, and illustrates its interactions with the upstream transcription factor SP1 and the downstream modulatory axis miR-421/KDM2A/ERK, thus offering novel insights into the cancerogenic mechanism in CRC.
Collapse
Affiliation(s)
- Wei Liu
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Jin Meng
- Department of Fifth Treatment Areas of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang, 110002, P.R. China
| | - Rongjun Su
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Changjun Shen
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Shuai Zhang
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Yantao Zhao
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Wenqi Liu
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Jiang Du
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Shuai Zhu
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Pan Li
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China
| | - Zhigang Wang
- Department of Fifth Treatment Areas of Anorectal Disease, Shenyang Coloproctology Hospital, Shenyang, 110002, P.R. China
| | - Xiaoxia Li
- Department of General Surgery, Yan'an People's Hospital, Yan'an, 716000, P.R. China.
| |
Collapse
|
7
|
Guo Y, Ren Y, Dong X, Kan X, Zheng C. An Overview of Hepatocellular Carcinoma After Insufficient Radiofrequency Ablation. J Hepatocell Carcinoma 2022; 9:343-355. [PMID: 35502292 PMCID: PMC9056053 DOI: 10.2147/jhc.s358539] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022] Open
Abstract
Radiofrequency ablation (RFA) is a commonly used treatment for hepatocellular carcinoma (HCC), however, various complex conditions in clinical practice may lead to insufficient radiofrequency ablation (IRFA), allowing residual HCC to survive. In clinical practice and laboratory models, IRFA plays an important role in rapid tumor progression. Therefore, targeting the residual HCC and avoiding IRFA were worthwhile methods. A deeper understanding of IRFA is required; IRFA contributes to the improvement of proliferative activity, migration rates, and invasive capacity, and this may be due to the involvement of multiple complex processes or proteins, including epithelial mesenchymal transitions (EMTs), cancer stem cells (CSCs), autophagy, heat shock proteins (HSPs), changes of non-tumor cells and extracellular matrix, altered immune microenvironment, hypoxia-inducible factors (HIFs), growth factors, epigenetic alterations, and metabolic reprogramming. We focus on the processes of the above mechanisms and possible therapeutic approach, with a review of the literature. Additionally, we recapitulated the construction methods of various experimental models of IRFA (in vivo and in vitro).
Collapse
Affiliation(s)
- Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, People’s Republic of China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, People’s Republic of China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, People’s Republic of China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, People’s Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, People’s Republic of China
- Correspondence: Chuansheng Zheng, Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China, Tel/Fax +86-27-85726290, Email
| |
Collapse
|
8
|
Ge X, Yao Y, Li J, Li Z, Han X. Role of LncRNAs in the Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma. Front Oncol 2021; 11:690800. [PMID: 34113574 PMCID: PMC8185227 DOI: 10.3389/fonc.2021.690800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of primary liver cancer with a high incidence and mortality rate. HCC develops insidiously, and most newly diagnosed cases are in the middle and advanced stages. The epithelial-mesenchymal transition (EMT) is a vital mechanism underlying metastasis in patients with advanced HCC. EMT is a multistep and complex procedure. The promotion and inhibition of EMT directly affect the migration and invasion of HCC. LncRNAs are involved in the epigenetic modification of genes, regulation of gene transcription, and posttranslational modification of proteins. LncRNAs also play important roles in regulating EMT progression in HCC and are promising biomarkers and therapeutic targets. This review focused on summarizing the mechanism by which lncRNAs regulate EMT in HCC. In particular, lncRNAs were reported to primarily act as RNA sponges, and the regulation of EMT involves major signaling pathways. Finally, we reviewed the mechanisms by which lncRNAs are involved in drug resistance and discussed the clinical prospects and potential challenges of utilizing lncRNAs to treat HCC.
Collapse
Affiliation(s)
- Xiaoyong Ge
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Yao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaonan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Kuo CY, Weng TS, Kumar KJS, Tseng YH, Tung TW, Wang SY, Wang HC. Ethanol Extracts of Dietary Herb, Alpinia nantoensis, Exhibit Anticancer Potential in Human Breast Cancer Cells. Integr Cancer Ther 2020; 18:1534735419866924. [PMID: 31409145 PMCID: PMC6696839 DOI: 10.1177/1534735419866924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent advances in mammography screening, chemotherapy, and adjuvant treatment modalities have improved the survival rate of women with breast cancer. Nevertheless, the breast tumor with metastatic progression is still life-threatening. Indeed, combination therapy with Ras-ERK and PI3K inhibitors is clinically effective in malignant breast cancer treatment. Constituents from genus Alpinia plants have been implicated as potent anticancer agents in terms of their efficacy of inhibiting tumor cell metastasis. In this study, we tested the effects of ethanol extracts of Alpinia nantoensis (rhizome, stem, and leaf extracts) in cultured human breast cancer cells and particularly focused on the Ras-ERK and PI3K/AKT pathways. We found that the rhizome and leaf extracts from A nantoensis inhibited cell migration, invasion, and sphere formation in MCF-7 and MDA-MB-231 cells. The potency was extended with the inhibition of serum-induced PI3K/AKT and Ras-ERK activation and epidermal growth factor (EGF)-mediated EGFR activation in MDA-MB-231 cells. These results indicate that extracts of A nantoensis could inhibit signal transduction at least involved in EGFR as well as the PI3K/AKT and Ras-ERK pathways, which are crucial players of tumor cell migration and invasion. Our study strongly supports that the extracts of A nantoensis could be a novel botanical drug lead for the development of an antimetastatic agent for the treatment of human malignant breast cancer.
Collapse
Affiliation(s)
- Ching-Ying Kuo
- 1 Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Teng-Song Weng
- 1 Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,2 Chi Mei Medical Center, Liouying, Tainan 73657, Taiwan
| | - K J Senthil Kumar
- 3 Department of Forestry, National Chung-Hsing University, Taichung 40227, Taiwan
| | - Yen-Hsueh Tseng
- 3 Department of Forestry, National Chung-Hsing University, Taichung 40227, Taiwan
| | - Ta-Wei Tung
- 1 Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sheng-Yang Wang
- 3 Department of Forestry, National Chung-Hsing University, Taichung 40227, Taiwan.,4 Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Hui-Chun Wang
- 1 Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,5 Department of Medical Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.,6 Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|
10
|
Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol 2019; 20:69-84. [PMID: 30459476 DOI: 10.1038/s41580-018-0080-4] [Citation(s) in RCA: 2209] [Impact Index Per Article: 441.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular programme that is known to be crucial for embryogenesis, wound healing and malignant progression. During EMT, cell-cell and cell-extracellular matrix interactions are remodelled, which leads to the detachment of epithelial cells from each other and the underlying basement membrane, and a new transcriptional programme is activated to promote the mesenchymal fate. In the context of neoplasias, EMT confers on cancer cells increased tumour-initiating and metastatic potential and a greater resistance to elimination by several therapeutic regimens. In this Review, we discuss recent findings on the mechanisms and roles of EMT in normal and neoplastic tissues, and the cell-intrinsic signals that sustain expression of this programme. We also highlight how EMT gives rise to a variety of intermediate cell states between the epithelial and the mesenchymal state, which could function as cancer stem cells. In addition, we describe the contributions of the tumour microenvironment in inducing EMT and the effects of EMT on the immunobiology of carcinomas.
Collapse
Affiliation(s)
- Anushka Dongre
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA. .,MIT Ludwig Center for Molecular Oncology, Cambridge, MA, USA. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
11
|
Zinn R, Otterbein H, Lehnert H, Ungefroren H. RAC1B: A Guardian of the Epithelial Phenotype and Protector Against Epithelial-Mesenchymal Transition. Cells 2019; 8:cells8121569. [PMID: 31817229 PMCID: PMC6952788 DOI: 10.3390/cells8121569] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
The small GTPase Ras-related C3 botulinum toxin substrate 1B (RAC1B) has been shown to potently inhibit transforming growth factor (TGF)-β1-induced cell migration and epithelial-mesenchymal transition (EMT) in pancreatic and breast epithelial cells, but the underlying mechanism has remained obscure. Using a panel of pancreatic ductal adenocarcinoma (PDAC)-derived cell lines of different differentiation stages, we show that RAC1B is more abundantly expressed in well differentiated as opposed to poorly differentiated cells. Interestingly, RNA interference-mediated knockdown of RAC1B decreased expression of the epithelial marker protein E-cadherin, encoded by CDH1, and enhanced its TGF-β1-induced downregulation, whereas ectopic overexpression of RAC1B upregulated CDH1 expression and largely prevented its TGF-β1-induced silencing of CDH1. Conversely, knockdown of RAC1B, or deletion of the RAC1B-specific exon 3b by CRISPR/Cas-mediated genomic editing, enhanced basal and TGF-β1-induced upregulation of mesenchymal markers like Vimentin, and EMT-associated transcription factors such as SNAIL and SLUG. Moreover, we demonstrate that knockout of RAC1B enhanced the cells’ migratory activity and derepressed TGF-β1-induced activation of the mitogen-activated protein kinase ERK2. Pharmacological inhibition of ERK1/2 activation in RAC1B-depleted cells rescued cells from the RAC1B knockdown-induced enhancement of cell migration, TGF-β1-induced downregulation of CDH1, and upregulation of SNAI1. We conclude that RAC1B promotes epithelial gene expression and suppresses mesenchymal gene expression by interfering with TGF-β1-induced MEK-ERK signaling, thereby protecting cells from undergoing EMT and EMT-associated responses like acquisition of cell motility.
Collapse
Affiliation(s)
- Rabea Zinn
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hannah Otterbein
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Ungefroren
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Correspondence: ; Tel.: +49-451-3101-7866
| |
Collapse
|
12
|
Jeong YJ, Park YY, Park KK, Choi YH, Kim CH, Chang YC. Bee Venom Suppresses EGF-Induced Epithelial-Mesenchymal Transition and Tumor Invasion in Lung Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1869-1883. [PMID: 31786944 DOI: 10.1142/s0192415x19500952] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Bee venom of Apis mellifera is a traditional medicine in Asia. It has been used with promoting results for the treatment of pain, rheumatoid, and cancer disease. The purpose of this study was to investigate the effects of bee venom on epidermal growth factor (EGF)-induced epithelial-mesenchymal transition (EMT) in non-small cell lung cancer (NSCLC) and determine possible signaling pathway affected in EGF-induced EMT in A549 cells. Bee venom inhibited EGF-induced F-actin reorganization and cell invasion, and suppressed EGF-induced EMT, processes associated with tumor metastasis in NSCLC. Bee venom enhanced the upregulation of E-cadherin and the downregulation of vimentin and inhibited EGF-induced ERK, JNK, FAK, and mTOR phosphorylation in A549 cells. However, the inhibition of JNK phosphorylation by bee venom was not related to the inhibitory effects of EMT. Furthermore, we found that bee venom suppressed the EMT-related transcription factors ZEB2 and Slug by blocking EGF-induced ERK, FAK and mTOR phosphorylation. Bee venom inhibits EGF-induced EMT by blocking the phosphorylation of ERK, FAK, and mTOR, resulting in the suppression of ZEB2 and Slug. These data suggest bee venom as a potential antimetastatic agent for NSCLC.
Collapse
Affiliation(s)
- Yun-Jeong Jeong
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Korea
| | - Yoon-Yub Park
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Korea
| | - Kwan-Kyu Park
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, Sungkyunkwan University, Suwon, Kyunggi-Do 16419, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 42472, Korea
| |
Collapse
|
13
|
Wu JR, You RI, Hu CT, Cheng CC, Rudy R, Wu WS. Hydrogen peroxide inducible clone-5 sustains NADPH oxidase-dependent reactive oxygen species-c-jun N-terminal kinase signaling in hepatocellular carcinoma. Oncogenesis 2019; 8:40. [PMID: 31387985 PMCID: PMC6684519 DOI: 10.1038/s41389-019-0149-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/18/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022] Open
Abstract
Target therapy aiming at critical molecules within the metastatic signal pathways is essential for prevention of hepatocellular carcinoma (HCC) progression. Hic-5 (hydrogen peroxide inducible clone-5) which belongs to the paxillin superfamily, can be stimulated by a lot of metastatic factors, such as transforming growth factor (TGF-β), hepatocyte growth factor (HGF), and reactive oxygen species (ROS). Previous studies implicated Hic-5 cross-talks with the ROS-c-jun N-terminal kinase (JNK) signal cascade in a positive feedback manner. In this report, we addressed this issue in a comprehensive manner. By RNA interference and ectopic Hic-5 expression, we demonstrated Hic-5 was essential for activation of NADPH oxidase and ROS generation leading to activation of downstream JNK and c-jun transcription factor. This was initiated by interaction of Hic-5 with the regulator and adaptor of NADPH oxidase, Rac1 and Traf4, respectively, which may further phosphorylate the nonreceptor tyrosine kinase Pyk2 at Tyr881. On the other hand, promoter activity assay coupled with deletion mapping and site directed mutagenesis strategies demonstrated the distal c-jun and AP4 putative binding regions (943–1126 bp upstream of translational start site) were required for transcriptional activation of Hic-5. Thus Hic-5 was both downstream and upstream of NADPH oxidase-ROS-JNK-c-jun cascade. This signal circuit was essential for regulating the expression of epithelial mesenchymal transition (EMT) factors, such as Snail, Zeb1, E-cadherin, and matrix metalloproteinase 9, involved in HCC cell migration and metastasis. Due to the limited expression of Hic-5 in normal tissue, it can be a promising therapeutic target for preventing HCC metastasis.
Collapse
Affiliation(s)
- Jia-Ru Wu
- Department of Molecular Biology and Human Genetics, Hualien, Taiwan
| | - Ren-In You
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chi-Tan Hu
- Division of Gastroenterology, Department of Medicine, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan.,Research Centre for Hepatology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Chuan-Chu Cheng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Rudy Rudy
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sheng Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan. .,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
14
|
Hu H, Dong Z, Wang X, Bai L, Lei Q, Yang J, Li L, Li Q, Liu L, Zhang Y, Ji Y, Guo L, Liu Y, Cui H. Dehydrocorydaline inhibits cell proliferation, migration and invasion via suppressing MEK1/2-ERK1/2 cascade in melanoma. Onco Targets Ther 2019; 12:5163-5175. [PMID: 31456643 PMCID: PMC6620435 DOI: 10.2147/ott.s183558] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/01/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: Alkaloids are naturally occurring chemical compounds that are widely distributed in plants, and have pharmaceutical values and low toxicity. In recent years, some of them have been demonstrated to be promising therapeutic drug candidates for cancer treatment. Herein, we tried to explore the antitumor effect of dehydrocorydaline (DHC), a natural alkaloid isolated from Corydalis, on malignant melanoma. Methods: We treated two malignant metastatic melanoma cell lines, A375 and MV3, and a normal melanocyte cell line, PIG1, with various concentrations of DHC for set amounts of time, and detected cell proliferation, migration, and invasion by using MTT, BrdU, transwell, Western blot and soft agar assay in vitro and tumorigenicity in the xenografts in vivo. Results: Our results showed that DHC dramatically blocked cell proliferation and led to cell cycle arrest at G0/G1 phase and downregulated the expressions of cell cycle regulators CDK6 and Cyclin D1 in melanoma cells. However, DHC had little inhibitory effect on normal melanocyte cell line PIG-1. Meanwhile, DHC suppressed cell invasion and migration through modulating the epithelial–mesenchymal transition (EMT) markers including E-cadherin, vimentin, as well as β-catenin. In addition, DHC also significantly attenuated tumor growth in vivo. The expressions of cell cycle-related and metastasis-related proteins were further confirmed by immunohistochemical staining in the xenografts. Importantly, MEK1/2-ERK1/2 cascade was inactivated after DHC treatment and ERK activator t-butylhydroquinone (tBHQ) treatment rescued DHC-induced cell proliferation inhibition. Conclusions: Our results indicated that DHC inhibited cell proliferation and migration/invasion via inactivating MAPK signaling, and showed that DHC might be a potential novel drug to treat malignant melanoma.
Collapse
Affiliation(s)
- Huanrong Hu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Xianxing Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Longchang Bai
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Qian Lei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Qian Li
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Lichao Liu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Yanli Zhang
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Yacong Ji
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Leiyang Guo
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Yaling Liu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
15
|
Tong Y, Wang M, Dai Y, Bao D, Zhang J, Pan H. LncRNA HOXA-AS3 Sponges miR-29c to Facilitate Cell Proliferation, Metastasis, and EMT Process and Activate the MEK/ERK Signaling Pathway in Hepatocellular Carcinoma. HUM GENE THER CL DEV 2019; 30:129-141. [PMID: 30963785 DOI: 10.1089/humc.2018.266] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignant tumor with high morbidity and mortality across the world. Recent findings have suggested that long noncoding (lnc)RNA HOXA-AS3 plays an important role in tumorigenesis and metastasis in a variety of cancers. However, the role of lncRNA HOXA-AS3 in the initiation and progression of HCC remains largely unclear. In the present study, HOXA-AS3 was highly expressed in HCC tumor tissues and cell lines. High HOXA-AS3 expression was correlated with low survival of HCC patients. Loss-of-function experiments showed that knockdown of HOXA-AS3 inhibited cell proliferation, migration, invasion, the epithelial-mesenchymal transition (EMT) process, and the mitogen-activated protein kinase/extracellular regulated protein kinase (MEK/ERK) signaling pathway in HCC. Molecular mechanism exploration uncovered that HOXA-AS3 could directly interact with and negatively regulate miR-29c. BMP1 is a downstream target gene of miR-29c, and HOXA-AS3 could regulate BMP1 expression by targeting miR-29c. miR-29c negatively regulated and BMP1 promoted the progression of HCC. Rescue experiments revealed that miR-29c inhibitor could partially counteract the impact induced by HOXA-AS3 knockdown in HCC. Taken together, our study is the first to show the interaction of HOXA-AS3 with miR-29c in facilitating cell proliferation, metastasis, EMT process, and MEK/ERK signaling pathway in HCC.
Collapse
Affiliation(s)
- Yongxi Tong
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| | - Mingshan Wang
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| | - Yining Dai
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| | - Dujing Bao
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| | - Jiajie Zhang
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| | - Hongying Pan
- Department of Infection Diseases, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Tai Y, Zhang LH, Gao JH, Zhao C, Tong H, Ye C, Huang ZY, Liu R, Tang CW. Suppressing growth and invasion of human hepatocellular carcinoma cells by celecoxib through inhibition of cyclooxygenase-2. Cancer Manag Res 2019; 11:2831-2848. [PMID: 31114336 PMCID: PMC6497485 DOI: 10.2147/cmar.s183376] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/23/2019] [Indexed: 02/05/2023] Open
Abstract
Purpose: Biomarkers are lacking in hepatocellular carcinoma (HCC). Cyclooxygenase-2 (COX-2) and its metabolites play crucial roles in the process of inflammation-tumor transformation. This study was aimed to detect COX-2 expression in HCC tissues and evaluate the effects of a COX-2 inhibitor, celecoxib, on biological behaviors of HCC cell lines in vitro. Methods: COX-2 expression was detected by immunohistochemistry on a human HCC tissue microarray. The correlations of COX-2 expression with tumor clinicopathological variables and overall survival were analyzed. The proliferation, apoptosis, cell cycle distribution, invasion capacity, and related signaling molecules of HCC cells after incubated with COX-2 inhibitor celecoxib were evaluated in vitro. Results: Expression levels of COX-2 in HCC tissues were significantly higher than those in paracancerous tissues. The TNM stage III-IV, tumor size >5 cm, lymphovascular invasion and distant metastasis was higher in high COX-2 expression group compared with that in low COX-2 expression group. Patients with low COX-2 expression achieved better 5-year overall survival than those with high COX-2 expression. Treatment with celecoxib was sufficient to inhibit cell proliferation, promote apoptosis, and induce G0/G1 cell cycle arrest in HCC cells with concentration- and time-dependent manners. Celecoxib up-regulated E-cadherin protein through inhibiting COX-2-prostaglandin E2 (PGE2)-PGE2 receptor 2 (EP2)-p-Akt/p-ERK signaling pathway to suppress HCC cells migration and invasion. Conclusion: High COX-2 expression was associated with advanced TNM stage, larger tumor size, increased lymphovascular invasion and short survival. Targeting inhibition of COX-2 by celecoxib exhibited anti-tumor activities by suppressing proliferation, promoting apoptosis, and inhibiting the aggressive properties of HCC cells.
Collapse
Affiliation(s)
- Yang Tai
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Lin-Hao Zhang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Jin-Hang Gao
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Chong Zhao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Huan Tong
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Cheng Ye
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Zhi-Yin Huang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Rui Liu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| | - Cheng-Wei Tang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China.,Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, People's Republic of China
| |
Collapse
|
17
|
Liang X, Qi M, Wu R, Liu A, Chen D, Tang L, Chen J, Hu X, Li W, Zhan L, Shao C. Long non-coding RNA CUDR promotes malignant phenotypes in pancreatic ductal adenocarcinoma via activating AKT and ERK signaling pathways. Int J Oncol 2018; 53:2671-2682. [PMID: 30272271 DOI: 10.3892/ijo.2018.4574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/10/2018] [Indexed: 11/06/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies, with a marked potential for invasion and metastasis. Emerging evidence has suggested that dysregulation of long non-coding RNAs (lncRNAs) is associated with the development of multiple types of cancer. However, the function of lncRNAs in PDAC is poorly known. In the present study, a microarray assay was used to screen for differently expressed lncRNAs in PDAC and it was identified that cancer upregulated drug resistance (CUDR) was upregulated in PDAC. CUDR increased PDAC cell proliferation, migration and invasion, inhibited apoptosis, and promoted drug resistance; it also regulated the PDAC cell epithelial-mesenchymal transition. The CUDR-induced PDAC malignant phenotypes is via the protein kinase B and extracellular-signal-regulated kinase signaling pathways. Downregulation of CUDR may be a novel therapeutic strategy to prevent PDAC development and drug resistance in the future.
Collapse
Affiliation(s)
- Xing Liang
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Meiyan Qi
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of The Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Rui Wu
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai 200433, P.R. China
| | - Anan Liu
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Danlei Chen
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Liang Tang
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jun Chen
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xiangui Hu
- Department of Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Wei Li
- General Surgical Department, Sir Run Run Shaw Hospital affiliated with The Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Lixing Zhan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of The Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Chenghao Shao
- Department of Pancreatic-Biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
18
|
Chen X, Hu X, Li Y, Zhu C, Dong X, Zhang R, Ma J, Huang S, Chen L. Resveratrol inhibits Erk1/2-mediated adhesion of cancer cells via activating PP2A-PTEN signaling network. J Cell Physiol 2018; 234:2822-2836. [PMID: 30066962 DOI: 10.1002/jcp.27100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Abstract
Resveratrol, a natural polyphenol compound, has been shown to possess anticancer activity. However, how resveratrol inhibits cancer cell adhesion has not been fully elucidated. Here, we show that resveratrol suppressed the basal or type I insulin-like growth factor (IGF)-1-stimulated adhesion of cancer cells (Rh1, Rh30, HT29, and HeLa cells) by inhibiting the extracellular signal-regulated kinase 1/2 (Erk1/2) pathway. Inhibition of Erk1/2 with U0126, knockdown of Erk1/2, or overexpression of dominant-negative mitogen-activated protein kinase kinase 1 (MKK1) strengthened resveratrol's inhibition of the basal or IGF-1-stimulated of Erk1/2 phosphorylation and cell adhesion, whereas ectopic expression of constitutively active MKK1 attenuated the inhibitory effects of resveratrol. Further research revealed that both protein phosphatase 2A (PP2A) and phosphatase and tensin homolog (PTEN)-Akt were implicated in resveratrol-inactivated Erk1/2-dependent cell adhesion. Inhibition of PP2A with okadaic acid or overexpression of dominant-negative PP2A rendered resistance to resveratrol's suppression of the basal or IGF-1-stimulated phospho-Erk1/2 and cell adhesion, whereas expression of wild-type PP2A enhanced resveratrol's inhibitory effects. Overexpression of wild-type PTEN or dominant-negative Akt or inhibition of Akt with Akt inhibitor X strengthened resveratrol's inhibition of the basal or IGF-1-stimulated Erk1/2 phosphorylation and cell adhesion. Furthermore, inhibition of mechanistic/mammalian target of rapamycin (mTOR) with rapamycin or silencing mTOR enhanced resveratrol's inhibitory effects on the basal and IGF-1-induced inhibition of PP2A-PTEN, activation of Akt-Erk1/2, and cell adhesion. The results indicate that resveratrol inhibits Erk1/2-mediated adhesion of cancer cells via activating PP2A-PTEN signaling network. Our data highlight that resveratrol has a great potential in the prevention of cancer cell adhesion.
Collapse
Affiliation(s)
- Xin Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaoyu Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yue Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Cuilan Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jing Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
19
|
Zhong C, Zhuang M, Wang X, Li J, Chen Z, Huang Y, Chen F. 12-Lipoxygenase promotes invasion and metastasis of human gastric cancer cells via epithelial-mesenchymal transition. Oncol Lett 2018; 16:1455-1462. [PMID: 30008824 PMCID: PMC6036329 DOI: 10.3892/ol.2018.8808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 03/22/2018] [Indexed: 12/13/2022] Open
Abstract
The role of 12-lipoxygenase (12-LOX) in tumorigenesis has been well established in several types of human cancer, including gastric cancer. It was reported that epithelial-mesenchymal transition (EMT) contributes to tumor invasion and metastasis. However, whether 12-LOX promotes the invasion and metastasis of human gastric cancer cells via EMT remains to be elucidated. In the present study, the expression of 12-LOX and EMT markers, N-cadherin and E-cadherin, was evaluated in gastric cancer and adjacent normal mucosa samples by immunohistochemical analysis. 12-LOX-overexpressing gastric cancer cells were established via lentiviral transfection of SCG-7901 cells. Wound-healing and Transwell assays were performed to examine the regulation of cell metastasis and invasion by 12-LOX. Furthermore, the regulation of N-cadherin expression by 12-LOX was evaluated using reverse transcription-quantitative polymerase chain reaction and western blotting. The results revealed that the expression of 12-LOX and N-cadherin was significantly higher in gastric cancer compared with that in adjacent normal mucosa tissues (P<0.05). By contrast, the expression of E-cadherin was significantly decreased in gastric cancer compared with that in adjacent normal mucosa tissues (P<0.05). Furthermore, the expression of 12-LOX was positively associated with N-cadherin expression in gastric cancer tissues. 12-LOX-overexpressing gastric cancer cells exhibited significantly increased invasion and migration abilities compared with the empty vector and control groups. The expression of N-cadherin in 12-LOX-overexpressing gastric cancer cells was increased compared with that in the empty vector and control groups. The present study suggests that EMT may be involved in the promotion of the invasion and metastasis of human gastric cancer cells by 12-LOX.
Collapse
Affiliation(s)
- Canmei Zhong
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Mingkai Zhuang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xiazhong Wang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jianying Li
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Zhixin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yuehong Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Fenglin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
20
|
Luo Y, Yu T, Zhang Q, Fu Q, Hu Y, Xiang M, Peng H, Zheng T, Lu L, Shi H. Upregulated N-cadherin expression is associated with poor prognosis in epithelial-derived solid tumours: A meta-analysis. Eur J Clin Invest 2018; 48:e12903. [PMID: 29405291 PMCID: PMC5887888 DOI: 10.1111/eci.12903] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/28/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND N-cadherin is an important molecular in epithelial-mesenchymal transition (EMT) and has been reported to be associated with aggressive behaviours of tumours. However, prognostic value of N-cadherin in solid malignancies remains controversially. MATERIALS AND METHODS The Pubmed/MELINE and EMBASE databases were used for a comprehensive literature searching. Pooled risk ratio (RR) and hazard ratio (HR) with their corresponding 95% confidence intervals (CIs) were employed to quantify the prognostic role. RESULTS Involving 36 studies with 5705 patients were performed to investigate relationships between N-cadherin upregulation and clinicopathological features, survival. Results suggested upregulated N-cadherin was associated with lymph node metastasis (RR = 1.16, 95% CI [1.00, 1.35]), higher histological grade (RR = 1.36, 95%CI [1.14, 1.62]), angiolymphatic invasion (RR = 1.19, 95% CI [1.06, 1.34]) and advanced clinical stage (RR = 1.32, 95% CI [1.06, 1.64]), while upregulated N-cadherin was apt to be associated with distant metastasis (RR = 1.43, 95% CI [0.99, 2.05]). Moreover, N-cadherin was correlated with poor prognosis of 3-year survival (HR = 1.78, 95% CI [1.51, 2.10]), 5-year survival (HR = 1.57, 95% CI [1.17, 2.10]) and overall survival (OS) (HR = 1.32, 95% CI [1.20, 1.44]). Subgroup analyses according to cancer types were also conducted for applying these conclusions to some tumours more properly. No publication bias was found except subgroup analysis of distant metastasis (P = .652 for Begg's test and 0.023 for Egger's test). CONCLUSIONS Taken together, upregulation of N-cadherin is associated with more aggressive behaviours of epithelial-derived solid malignancies and can be regarded as a predictor of poor survival.
Collapse
Affiliation(s)
- Yong Luo
- State Key Laboratory of Biotherapy and Department of Head and Neck OncologyWest China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Ting Yu
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Qiongwen Zhang
- State Key Laboratory of Biotherapy and Department of Head and Neck OncologyWest China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Qingyu Fu
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Yuzhu Hu
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Mengmeng Xiang
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Haoning Peng
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Tianying Zheng
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| | - Li Lu
- College of Computer ScienceSichuan UniversityChengduSichuanChina
| | - Huashan Shi
- State Key Laboratory of Biotherapy and Department of Head and Neck OncologyWest China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
- West China HospitalWest China School of MedicineSichuan UniversityChengduSichuanChina
| |
Collapse
|
21
|
Dong W, Li N, Pei X, Wu X. Differential expression of DUSP2 in left- and right-sided colon cancer is associated with poor prognosis in colorectal cancer. Oncol Lett 2018; 15:4207-4214. [PMID: 29541187 PMCID: PMC5835964 DOI: 10.3892/ol.2018.7881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/08/2017] [Indexed: 01/18/2023] Open
Abstract
Dual-specificity phosphatase-2 (DUSP2), a negative regulator of extracellular-regulated kinase activity, has been identified as an important kinase with emerging roles in cancer. However, the clinical significance of DUSP2 in colorectal cancer (CRC) remains to be fully elucidated. In the present study, the expression of DUSP2 was investigated using immunohistochemistry in 96 patients with CRC. Cell viability was estimated using a cell counting kit-8 assay, and cell apoptosis by flow cytometry. The relationship between DUSP2 expression and patient characteristics, including overall survival, were studied retrospectively in these patients. It was found that DUSP2 was differentially expressed between left-sided colon carcinoma (LSCC) and right-sided colon carcinoma (RSCC). It was also found that decreased expression of DUSP2 was correlated with significantly shorter overall survival (P=0.001) and short distant-metastasis-free survival (P=0.002). In univariate comparisons, the decreased expression of DUSP2 was found to be an independent risk factor for poor survival rate (HR 3.55, CI 1.092-9.896; P=0.002). It was also found that the enforced overexpression of DUSP2 sensitized CRC cells to cetuximab. In conclusion, the findings demonstrated that DUSP2 was differentially expressed between RSCC and LSCC, and that the overexpression of DUSP2 increased the inhibitory effect of cetuximab in CRC, suggesting that DUSP2 may be a novel biomarker and therapeutic target in CRC therapy.
Collapse
Affiliation(s)
- Wenjie Dong
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Na Li
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiufeng Pei
- Department of Internal Medical Oncology, Tumor Hospital of Baotou, Baotou, Inner Mongolia 014030, P.R. China
| | - Xinai Wu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
22
|
Kang JH, Choi MY, Cui YH, Kaushik N, Uddin N, Yoo KC, Kim MJ, Lee SJ. Regulation of FBXO4-mediated ICAM-1 protein stability in metastatic breast cancer. Oncotarget 2017; 8:83100-83113. [PMID: 29137327 PMCID: PMC5669953 DOI: 10.18632/oncotarget.20912] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/27/2017] [Indexed: 12/20/2022] Open
Abstract
Advanced or progressive cancers share common traits such as altered transcriptional modulation, genetic modification, and abnormal post-translational regulation. These processes influence protein stability and cellular activity. Intercellular adhesion molecule-1 (ICAM-1) is involved in the malignant progression of various human cancers, including breast, liver, renal, and pancreatic cancers, but protein stability has not been deal with in metastatic breast cancer. Additionally, the relevance of the stability maintenance of ICAM-1 protein remains obscure. Here, we identified a novel interaction of E3 ligase FBXO4 that is specifically presented to ICAM-1. To understand how FBXO4 modulates ICAM-1 stability, we investigated ICAM-1-overexpressing or knockdown metastatic/non-metastatic breast cancers. ICAM-1 was found to influence tumor progression and metastasis, whereas FBXO4 regulated aggressive tumorigenic conditions. These results demonstrate that FBXO4 is a major regulator of ICAM-1 stability and that alterations in the stability of ICAM-1 can influence therapeutic outcome in metastatic cancer.
Collapse
Affiliation(s)
- Jae-Hyeok Kang
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Mi-Young Choi
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Yan-Hong Cui
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Neha Kaushik
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Nizam Uddin
- Centre of Excellence in Molecular Biology (CEMB), University of The Panjab, Lahore, Pakistan
| | - Ki-Chun Yoo
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Min-Jung Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
23
|
Bartolowits MD, Brown W, Ali R, Pedley AM, Chen Q, Harvey KE, Wendt MK, Davisson VJ. Selective Inhibition of STAT3 Phosphorylation Using a Nuclear-Targeted Kinase Inhibitor. ACS Chem Biol 2017; 12:2371-2378. [PMID: 28787571 DOI: 10.1021/acschembio.7b00341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The discovery of compounds that selectively modulate signaling and effector proteins downstream of EGFR could have important implications for understanding specific roles for pathway activation. A complicating factor for receptor tyrosine kinases is their capacity to be translocated to the nucleus upon ligand engagement. Once localized in subcellular compartments like the nucleus, the roles for EGFR take on additional features, many of which are still being revealed. Additionally, nuclear localization of EGFR has been implicated in downstream events that have significance for therapy resistance and disease progression. The challenges to addressing the differential roles for EGFR in the nucleus motivated experimental approaches that can selectively modulate its subcellular function. By adding modifications to the established EGFR kinase inhibitor gefitinib, an approach to small molecule conjugates with a unique nuclear-targeting peptoid sequence was tested in both human and murine breast tumor cell models for their capacity to inhibit EGF-stimulated activation of ERK1/2 and STAT3. While gefitinib alone inhibits both of these downstream effectors, data acquired here indicate that compartmentalization of the gefitinib conjugates allows for pathway specific inhibition of STAT3 while not affecting ERK1/2 signaling. The inhibitor conjugates offered a more direct route to evaluate the role of EGF-stimulated epithelial-to-mesenchymal transition in these breast cancer cell models. These conjugates revealed that STAT3 activation is not involved in EGF-induced EMT, and instead utilization of the cytoplasmic MAP kinase signaling pathway is critical to this process. This is the first example of a conjugate kinase inhibitor capable of partitioning to the nucleus and offers a new approach to enhancing kinase inhibitor specificity.
Collapse
Affiliation(s)
- Matthew D. Bartolowits
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Wells Brown
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Remah Ali
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anthony M. Pedley
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Qingshou Chen
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kyle E. Harvey
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Michael K. Wendt
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Vincent Jo Davisson
- Department of Medicinal Chemistry
and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
24
|
Tiran V, Lindenmann J, Brcic L, Heitzer E, Stanzer S, Tabrizi-Wizsy NG, Stacher E, Stoeger H, Popper HH, Balic M, Dandachi N. Primary patient-derived lung adenocarcinoma cell culture challenges the association of cancer stem cells with epithelial-to-mesenchymal transition. Sci Rep 2017; 7:10040. [PMID: 28855609 PMCID: PMC5577216 DOI: 10.1038/s41598-017-09929-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 08/01/2017] [Indexed: 12/20/2022] Open
Abstract
The cancer stem cell (CSC) and epithelial-to-mesenchymal transition (EMT) models have been closely associated and used to describe both the formation of metastasis and therapy resistance. We established a primary lung cell culture from a patient in a clinically rare and unique situation of primary resistant disease. This culture consisted of two biologically profoundly distinct adenocarcinoma cell subpopulations, which differed phenotypically and genotypically. One subpopulation initiated and sustained in spheroid cell culture (LT22s) whereas the other subpopulation was only capable of growth and proliferation under adherent conditions (LT22a). In contrast to our expectations, LT22s were strongly associated with the epithelial phenotype, and expressed additionally CSC markers ALDH1 and CD133, whereas the LT22a was characterized as mesenchymal with lack of CSC markers. The LT22s cells also demonstrated an invasive behavior and mimicked gland formation. Finally, LT22s were more resistant to Cisplatin than LT22a cells. We demonstrate a primary lung adenocarcinoma cell culture derived from a patient with resistant disease, with epithelial aggressive subpopulation of cells associated with stem cell features and therapy resistance. Our findings challenge the current model associating CSC and disease resistance mainly to mesenchymal cells and may have important clinical implications.
Collapse
Affiliation(s)
- Verena Tiran
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic and Hyperbaric Surgery, Medical University of Graz, A-8036, Graz, Austria
| | - Luka Brcic
- Institute of Pathology, Medical University of Graz, A-8036, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Medical University of Graz, A-8010, Graz, Austria
| | - Stefanie Stanzer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036, Graz, Austria
| | | | - Elvira Stacher
- Institute of Pathology, Medical University of Graz, A-8036, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, A-8010, Graz, Austria
| | - Herbert Stoeger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036, Graz, Austria
| | - Helmut H Popper
- Institute of Pathology, Medical University of Graz, A-8036, Graz, Austria
| | - Marija Balic
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036, Graz, Austria.
- Research Unit Circulating Tumor Cells and Cancer Stem Cells, Medical University of Graz, A-8036, Graz, Austria.
| | - Nadia Dandachi
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036, Graz, Austria.
- Research Unit Epigenetic and Genetic Cancer Biomarkers, Medical University of Graz, A-8036, Graz, Austria.
| |
Collapse
|
25
|
Cytosolic THUMPD1 promotes breast cancer cells invasion and metastasis via the AKT-GSK3-Snail pathway. Oncotarget 2017; 8:13357-13366. [PMID: 28076326 PMCID: PMC5355103 DOI: 10.18632/oncotarget.14528] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/27/2016] [Indexed: 01/05/2023] Open
Abstract
Human THUMP domain-containing protein 1 (THUMPD1) is a specific adaptor protein that modulates tRNA acetylation through interaction with NAT10. Immunohistochemical analysis of 146 breast cancer specimens (82 triple-negative and 64 non-triple-negative cases) indicated THUMPD1 expression is higher in breast cancer tissues (60.9%, 89/146) than normal breast tissues (28.3%, 15/53; p < 0.001). Overall and cytosolic, but not nuclear, THUMPD1 expression in breast cancer correlated with advanced TNM stage (p = 0.003 and p < 0.001, respectively), lymph node metastasis (p = 0.001 and p < 0.001, respectively), and poor patient prognosis (p = 0.001 and p < 0.001, respectively). THUMPD1 interacted and co-localized with YAP, but did not affect Hippo pathway activity. THUMPD1 overexpression enhanced breast cancer cells invasion and migration in vivo and in vitro, possibly through activation of AKT, GSK3β and Snail, and inhibition of E-cadherin. Treatment with the AKT inhibitor, LY294002, reduced the effects of THUMPD1 overexpression in breast cancer cells. These results indicate that THUMPD1 promotes breast cancer cells invasion and migration via the AKT-GSK3β-Snail pathway.
Collapse
|
26
|
Hu X, Zhai Y, Kong P, Cui H, Yan T, Yang J, Qian Y, Ma Y, Wang F, Li H, Cheng C, Zhang L, Jia Z, Li Y, Yang B, Xu E, Wang J, Yang J, Bi Y, Chang L, Wang Y, Zhang Y, Song B, Li G, Shi R, Liu J, Zhang M, Cheng X, Cui Y. FAT1 prevents epithelial mesenchymal transition (EMT) via MAPK/ERK signaling pathway in esophageal squamous cell cancer. Cancer Lett 2017; 397:83-93. [DOI: 10.1016/j.canlet.2017.03.033] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/07/2017] [Accepted: 03/25/2017] [Indexed: 02/06/2023]
|
27
|
Ying HY, Gong CJ, Feng Y, Jing DD, Lu LG. Serine protease inhibitor Kazal type 1 (SPINK1) downregulates E-cadherin and induces EMT of hepatoma cells to promote hepatocellular carcinoma metastasis via the MEK/ERK signaling pathway. J Dig Dis 2017; 18:349-358. [PMID: 28544403 DOI: 10.1111/1751-2980.12486] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/22/2017] [Accepted: 05/17/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate serine protease inhibitor Kazal type 1 (SPINK1) expression and its influence on the prognosis of human hepatocellular carcinoma (HCC) and to explore the underlying molecular mechanisms involved. METHODS Altogether 80 patients with HCC who underwent curative resection were followed up for a median of 58.6 months. SPINK1 expression was detected in the primary HCC samples by immunohistochemistry. Its role in tumor invasion and metastasis was evaluated in vitro by gene silencing using a small interfering RNA-mediated approach, recombinant SPINK1 and U0126 (an inhibitor of MEK/ERK). The proteins in the MEK/ERK signaling pathway were detected by Western blot. RESULTS Patients with high SPINK1 expression showed poor overall survival (P = 0.0001) and recurrence-free survival (P = 0.001) compared with those with low SPINK1 expression. The suppression of SPINK1 resulted in reduced cell migration and invasion. SPINK1 overexpression was significantly associated with increased cell migration and invasion in vitro. Furthermore, SPINK1 promoted cancer cells motility and epithelial-mesenchymal transition (EMT) via the mitogen-activated protein kinase kinase (MAPK) and extracellular regulated kinase (ERK) pathway, resulting in increased vimentin expression and decreased E-cadherin expression. CONCLUSION SPINK1 may be an oncogene that induces EMT via the MEK/ERK pathway and is a potential target for HCC therapy.
Collapse
Affiliation(s)
- Hai Yan Ying
- Department of Geriatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Jie Gong
- Department of Geriatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Feng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Da Dao Jing
- Department of Geriatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lun Gen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Niu X, Liu F, Zhou Y, Zhou Z, Zhou D, Wang T, Li Z, Ye X, Yu Y, Weng X, Zhang H, Ye J, Liao M, Liu Y, Chen Z, Lu S. Genome-wide DNA Methylation Analysis Reveals GABBR2 as a Novel Epigenetic Target for EGFR 19 Deletion Lung Adenocarcinoma with Induction Erlotinib Treatment. Clin Cancer Res 2017; 23:5003-5014. [PMID: 28490462 DOI: 10.1158/1078-0432.ccr-16-2688] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/05/2017] [Accepted: 05/02/2017] [Indexed: 11/16/2022]
Abstract
Purpose: The past decade has witnessed the rapid development of personalized targeted therapies in lung cancer. It is still unclear whether epigenetic changes are involved in the response to tyrosine kinase inhibitor (TKI) treatment in epidermal growth factor receptor (EGFR)-mutated lung cancer.Experimental Design: Methyl-sensitive cut counting sequencing (MSCC) was applied to investigate the methylation changes in paired tissues before and after erlotinib treatment for 42 days with partial response (PR) from stage IIIa (N2) lung adenocarcinoma patients (N = 2) with EGFR 19 deletion. The Sequenom EpiTYPER assay was used to validate the changed methylated candidate genes. Up- or downregulation of the candidate gene was performed to elucidate the potential mechanism in the regulation of erlotinib treatment response.Results: Sixty aberrant methylated genes were screened using MSCC sequencing. Two aberrant methylated genes, CBFA2T3 and GABBR2, were clearly validated. A same differential methylated region (DMR) between exon 2 and exon 3 of GABBR2 gene was confirmed consistently in both patients. GABBR2 was significantly downregulated in EGFR 19 deletion cells, HCC4006 and HCC827, but remained conserved in EGFR wild-type A549 cells after erlotinib treatment. Upregulation of GABBR2 expression significantly rescued erlotinib-induced apoptosis in HCC827 cells. GABBR2 was significantly downregulated, along with the reduction of S6, p-p70 S6, and p-ERK1/2, demonstrating that GABBR2 may play an important role in EGFR signaling through the ERK1/2 pathway.Conclusions: We demonstrated that GABBR2 gene might be a novel potential epigenetic treatment target with induction erlotinib treatment for stage IIIa (N2) EGFR 19 deletion lung adenocarcinoma. Clin Cancer Res; 23(17); 5003-14. ©2017 AACR.
Collapse
Affiliation(s)
- Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Fatao Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yi Zhou
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Zhen Zhou
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Daizhan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ting Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Ziming Li
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiangyun Ye
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yongfeng Yu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiaoling Weng
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Hong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Junyi Ye
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China.,Key Laboratory of Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Meilin Liao
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yun Liu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Zhiwei Chen
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China.
| |
Collapse
|
29
|
Li D, Chen X, Wang H, Yu Y, Liu J, Wang Y, Zhang J, Zheng M, Zhi J. Nanodiamonds as Raman probes for specifically targeted bioimaging: visualization and mechanism study of the biorecognition between nanodiamonds-EGF and EGFR. RSC Adv 2017. [DOI: 10.1039/c6ra28139g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Targeting ligand EGF induced cell morphology change and potential cell migration risks visualized with Raman mapping using NDs as probes.
Collapse
Affiliation(s)
- Dandan Li
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
- University of Chinese Academy of Sciences
| | - Xin Chen
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
- University of Chinese Academy of Sciences
| | - Hong Wang
- Department of Pharmaceutical Analysis
- School of Pharmaceutical Sciences
- Peking University
- PR China
| | - Yuan Yu
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
| | - Jie Liu
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
| | - Yu Wang
- Beijing Center for Physical and Chemical Analysis
- PR China
| | - Jinghua Zhang
- Beijing Center for Physical and Chemical Analysis
- PR China
| | - Meiling Zheng
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
| | - Jinfang Zhi
- Laboratory of Photochemical Conversion and Optoelectronic Materials
- Technical Institute of Physics and Chemistry
- Chinese Academy of Sciences
- PR China
| |
Collapse
|
30
|
Ci Y, Qiao J, Han M. Molecular Mechanisms and Metabolomics of Natural Polyphenols Interfering with Breast Cancer Metastasis. Molecules 2016; 21:E1634. [PMID: 27999314 PMCID: PMC6273039 DOI: 10.3390/molecules21121634] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 01/22/2023] Open
Abstract
Metastatic cancers are the main cause of cancer-related death. In breast primary cancer, the five-year survival rate is close to 100%; however, for metastatic breast cancer, that rate drops to a mere 25%, due in part to the paucity of effective therapeutic options for treating metastases. Several in vitro and in vivo studies have indicated that consumption of natural polyphenols significantly reduces the risk of cancer metastasis. Therefore, this review summarizes the research findings involving the molecular mechanisms and metabolomics of natural polyphenols and how they may be blocking breast cancer metastasis. Most natural polyphenols are thought to impair breast cancer metastasis through downregulation of MMPs expression, interference with the VEGF signaling pathway, modulation of EMT regulator, inhibition of NF-κB and mTOR expression, and other related mechanisms. Intake of natural polyphenols has been shown to impact endogenous metabolites and complex biological metabolic pathways in vivo. Breast cancer metastasis is a complicated process in which each step is modulated by a complex network of signaling pathways. We hope that by detailing the reported interactions between breast cancer metastasis and natural polyphenols, more attention will be directed to these promising candidates as effective adjunct therapies against metastatic breast cancer in the clinic.
Collapse
Affiliation(s)
- Yingqian Ci
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China.
| | - Jinping Qiao
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China.
| | - Mei Han
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China.
| |
Collapse
|