1
|
Yang L, Bhat AM, Qazi S, Raza K. DLC1 as Druggable Target for Specific Subsets of Gastric Cancer: An RNA-seq-Based Study. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:514. [PMID: 36984515 PMCID: PMC10056469 DOI: 10.3390/medicina59030514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Background: Gastric cancer has been ranked the third leading cause of cancer death worldwide. Its detection at the early stage is difficult because patients mostly experience vague and non-specific symptoms in the early stages. Methods: The RNA-seq datasets of both gastric cancer and normal samples were considered and processed. The obtained differentially expressed genes were then subjected to functional enrichment analysis and pathway analysis. An implicit atomistic molecular dynamics simulation was executed on the selected protein receptor for 50 ns. The electrostatics, surface potential, radius of gyration, and macromolecular energy frustration landscape were computed. Results: We obtained a large number of DEGs; most of them were down-regulated, while few were up-regulated. A DAVID analysis showed that most of the genes were prominent in the KEGG and Reactome pathways. The most prominent GAD disease classes were cancer, metabolic, chemdependency, and infection. After an implicit atomistic molecular dynamics simulation, we observed that DLC1 is electrostatically optimized, stable, and has a reliable energy frustration landscape, with only a few maximum energy frustrations in the loop regions. It has a good functional and binding affinity mechanism. Conclusions: Our study revealed that DLC1 could be used as a potential druggable target for specific subsets of gastric cancer.
Collapse
Affiliation(s)
- Lianlei Yang
- Department of Gastroenterology, The First People’s Hospital of Linping District, Hangzhou 311100, China
| | - Adil Manzoor Bhat
- Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India
| | - Sahar Qazi
- Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
2
|
Gong H, Chen K, Zhou L, Jin Y, Chen W. Deleted in liver cancer 1 suppresses the growth of prostate cancer cells through inhibiting Rho-associated protein kinase pathway. Asian J Urol 2023; 10:50-57. [PMID: 36721699 PMCID: PMC9875144 DOI: 10.1016/j.ajur.2021.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 02/03/2023] Open
Abstract
Objective Deleted in liver cancer 1 (DLC1) is a GTPase-activating protein that is reported as a suppressor in certain human cancers. However, the detailed biological function of DLC1 is still unclear in human prostate cancer (PCa). In the present study, we aimed to explore the function of DLC1 in PCa cells. Methods Silencing and overexpression of DLC1 were induced in an androgen-sensitive PCa cell line (LNCaP) using RNA interference and lentiviral vector transduction. The Cell Counting Kit-8 assay was performed to determine cell proliferation. The cell cycle was examined by performing a propidium iodide staining assay. Results Our results indicated that DLC1 overexpression markedly suppressed the proliferation and cell cycle progression of LNCaP cells. Moreover, DLC1 expression was negatively correlated with Rho-associated protein kinase (ROCK) expression in LNCaP cells. Importantly, this study showed that the ROCK inhibitor Y27632 restored the function of DLC1 in LNCaP cells and reduced the tumorigenicity of LNCaP cells in vivo. Conclusion Our results indicated that DLC1 overexpression markedly suppressed the proliferation and cell cycle progression of PCa cells and negatively correlated with ROCK expression in PCa cells and tissue.
Collapse
Affiliation(s)
- Hua Gong
- Department of Urology, Zhoupu Hospital, Medicine and Health Sciences, Shanghai University, Shanghai, China
| | - Kang Chen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lan Zhou
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongchao Jin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Chen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China,Corresponding author.
| |
Collapse
|
3
|
Tian R, Sun Y, Han X, Wang J, Gu H, Wang W, Liang L. Identification and validation of prognostic autophagy-related genes associated with immune microenvironment in human gastric cancer. Aging (Albany NY) 2022; 14:7617-7634. [PMID: 36173625 PMCID: PMC9550254 DOI: 10.18632/aging.204313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 09/17/2022] [Indexed: 12/24/2022]
Abstract
Autophagy-related genes (ATGs) play critical roles in tumorigenesis and progression in gastric cancer (GC). The present study aimed to identify immune-based prognostic ATGs and verify their functions in tumor immune microenvironment (TIME) in GC. Macrophage infiltration was found to negatively correlate with prognosis in GC patients. After stratifying by infiltration levels of macrophages, we screened The Cancer Genome Atlas and Human Autophagy Database to identify the differentially expressed ATGs (DE-ATGs). Of 1,433 differentially expressed genes between the two groups, seven genes qualified as DE-ATGs. Of these, CXCR4, DLC1, and MAP1LC3C, exhibited strong prognostic prediction ability in Kaplan-Meier survival–log-rank test. High expression of these genes correlated with increased occurrence of advanced grade 3 tumors and poor prognoses. Furthermore, GSEA indicated that they were significantly associated with oncogenic and immune-related pathways. The comprehensive evaluation of TIME via GEPIA, ESTIMATE, CIBERSORT, and TIMER suggested that the three DE-ATGs were closely associated with immune condition, both in terms of immune cells and immune scores. Thus, the outcome of this study may aid in better understanding of the ATGs and their interaction with the immune microenvironment, which would allow the development of novel inhibitors, personalized treatment, and immunotherapy in gastric cancer.
Collapse
Affiliation(s)
- Ruyue Tian
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China.,Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, China
| | - Ya Sun
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China
| | - Xuedi Han
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China
| | - Jiajun Wang
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China
| | - Hongli Gu
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China
| | - Wenhai Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, China
| | - Lei Liang
- Department of Ultrasound, Aero Space Central Hospital, Beijing 100050, China
| |
Collapse
|
4
|
Hinsenkamp I, Köhler JP, Flächsenhaar C, Hitkova I, Meessen SE, Gaiser T, Wieland T, Weiss C, Röcken C, Mowat M, Quante M, Taxauer K, Mejias-Luque R, Gerhard M, Vogelmann R, Meindl-Beinker N, Ebert M, Burgermeister E. Functional antagonism between CagA and DLC1 in gastric cancer. Cell Death Dis 2022; 8:358. [PMID: 35963849 PMCID: PMC9376073 DOI: 10.1038/s41420-022-01134-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022]
Abstract
Helicobacter (H.) pylori-induced gastritis is a risk factor for gastric cancer (GC). Deleted-in-liver-cancer-1 (DLC1/ARHGAP7) inhibits RHOA, a downstream mediator of virulence factor cytotoxin-A (CagA) signalling and driver of consensus-molecular-subtype-2 diffuse GC. DLC1 located to enterochromaffin-like and MIST1+ stem/chief cells in the stomach. DLC1+ cells were reduced in H. pylori gastritis and GC, and in mice infected with H. pylori. DLC1 positivity inversely correlated with tumour progression in patients. GC cells retained an N-terminal truncation variant DLC1v4 in contrast to full-length DLC1v1 in non-neoplastic tissues. H. pylori and CagA downregulated DLC1v1/4 promoter activities. DLC1v1/4 inhibited cell migration and counteracted CagA-driven stress phenotypes enforcing focal adhesion. CagA and DLC1 interacted via their N- and C-terminal domains, proposing that DLC1 protects against H. pylori by neutralising CagA. H. pylori-induced DLC1 loss is an early molecular event, which makes it a potential marker or target for subtype-aware cancer prevention or therapy.
Collapse
Affiliation(s)
- Isabel Hinsenkamp
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jan P Köhler
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Flächsenhaar
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ivana Hitkova
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sabine Eberhart Meessen
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Timo Gaiser
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Röcken
- Institute of Pathology, Christian Albrechts University Kiel, Kiel, Germany
| | - Michael Mowat
- CancerCare Manitoba Research Institute, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Michael Quante
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Karin Taxauer
- Institute for Med. Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Raquel Mejias-Luque
- Institute for Med. Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Markus Gerhard
- Institute for Med. Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Roger Vogelmann
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadja Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,DKFZ-Hector Institute at the University Medical Center, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Mannheim Cancer Center (MCC), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elke Burgermeister
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
5
|
MiR-200a-3p promotes gastric cancer progression by targeting DLC-1. J Mol Histol 2021; 53:39-49. [PMID: 34751841 DOI: 10.1007/s10735-021-10037-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
Gastric cancer (GC) is one of the most common malignancies, ranking the third highest mortality rate worldwide. Due to the insidious symptoms and difficulty in early detection, patients with GS were mostly in the middle and late stages when they were diagnosed. Although ontogenetic or tumor-suppressive effects of miRNA-200a-3p have been demonstrated, the exact mechanism underlying GC is not clear. Therefore, the expression, effect, and mechanism of miRNA-200a-3p in GC progression were systematically investigated in this study. qRT-PCR, Western blotting, and immunohistochemical staining were applied to investigate the miRNA-200a-3p and deleted in liver cancer 1 (DLC-1) expression. Cell viability, proliferation, apoptosis, migration, and invasion capabilities of GC cells were assessed using cell counting kit-8 (CCK-8) colorimetry, EdU integration, flow cytometry, wound healing, and the transwell assay. The relationship between miRNA-200a-3p and tumor growth was investigated by tumor xenograft assay in vivo. A dual-luciferase reporter assay was estimated to verify the connection between miR-200-3p and DLC-1. The results showed that miRNA-200a-3p expression was significantly increased in both GC tissues and cells. Furthermore, via DLC-1, miRNA-200a-3p promotes tumor growth and development. miRNA-200a-3p, by targeting DLC-1, can function as an oncogene in GC cells. Collectively, our findings indicated that the miRNA-200a-3p/DLC axis might provide a theological basis for potential improvements in GC treatment strategies.
Collapse
|
6
|
Tumor suppressor gene DLC1: Its modifications, interactive molecules, and potential prospects for clinical cancer application. Int J Biol Macromol 2021; 182:264-275. [PMID: 33836193 DOI: 10.1016/j.ijbiomac.2021.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Deleted in liver cancer 1 (DLC1) is a recognized tumor suppressor gene that negatively regulates Rho family proteins by hydrolyzing the active GTP-bound state to its inactive GDP-bound state. Active Rho proteins play a positive role in tumorigenesis. Numerous in vitro and in vivo experiments have shown that DLC1 is downregulated or inactivated in various solid tumors, which may be due to the following five reasons: genomic deletion, epigenetic modification and ubiquitin-dependent proteasomal degradation may cause DLC1 underexpression; phosphorylation at the post-translation level may cause DLC1 inactivation; and failure to localize at focal adhesions (FAs) may prevent DLC1 from exerting full activity. All of the causes could be attributed to molecular binding. Experimental evidence suggests that direct or indirect targeting of DLC1 is feasible for cancer treatment. Therefore, elucidating the interaction of DLC1 with its binding partners might provide novel targeted therapies for cancer. In this review, we summarized the binding partners of DLC1 at both the gene and protein levels and expounded a variety of anticancer drugs targeting DLC1 to provide information about DLC1 as a cancer diagnostic indicator or therapeutic target.
Collapse
|
7
|
Niu N, Ma X, Liu H, Zhao J, Lu C, Yang F, Qi W. DLC1 inhibits lung adenocarcinoma cell proliferation, migration and invasion via regulating MAPK signaling pathway. Exp Lung Res 2021; 47:173-182. [PMID: 33678109 DOI: 10.1080/01902148.2021.1885524] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung adenocarcinoma (LUAD), one of the most common cancers, is a major threat to people's health due to its high mortality, and the survival of most patients suffering LUAD remains poor. This study aimed to explore the mechanism of Deleted in Liver Cancer 1 (DLC1) as a tumor suppressor underlying the occurrence and progression of LUAD. As revealed by bioinformatics analysis and qRT-PCR, DLC1 was significantly down-regulated in LUAD tumor tissue and cells. A series of cellular experiments including CCK-8, wound healing and Transwell assays were performed to detect the effect of DLC1 on the biological function of LUAD cells. It was found that overexpressing DLC1 significantly inhibited LUAD cell proliferative, migratory and invasive abilities, while knockdown of DLC1 promoted these abilities. Gene Set Enrichment Analysis (GSEA) and dual-luciferase assay were used to explore the downstream signaling pathway of DLC1, finding that DLC1 could remarkably inhibit the activity of mitogen-activated protein kinase (MAPK) signaling pathway. Western blot implemented for MAPK signaling pathway-related proteins further identified that DLC1 restrained the activation of MAPK/ERK signaling pathway. Furthermore, rescue experiments suggested that DLC1 inhibited LUAD cell proliferation and invasion by suppressing the MAPK/ERK signaling pathway. Overall, our study discussed the DLC1-dependent mechanism involved in LUAD. We found that the up-regulation of DLC1 may inhibit the malignant progression of LUAD by suppressing MAPK signaling pathway, which supports the view that DLC1 may serve as a molecular target for the targeted therapy of LUAD patients.
Collapse
Affiliation(s)
- Niu Niu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Xingjie Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Haitao Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Junjie Zhao
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Chao Lu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Fan Yang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| | - Weibo Qi
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), Jiaxing, Zhejiang, China
| |
Collapse
|
8
|
Nie Y, Liang X, Liu S, Guo F, Fang N, Zhou F. WASF3 Knockdown Sensitizes Gastric Cancer Cells to Oxaliplatin by Inhibiting ATG12-Mediated Autophagy. Am J Med Sci 2020; 359:287-295. [PMID: 32359534 DOI: 10.1016/j.amjms.2020.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric cancer is one of the most aggressive tumors, usually resulting in metastasis, and therapies for advanced gastric cancer remain limited. Drug resistance is the main reason for chemotherapeutic failure in gastric cancer. Wiskott-Aldrich syndrome protein family member 3 (WASF3) is required for invasion and metastasis of different cancers. However, there has been little study of WASF3 expression involvement in gastric cancer. In this study, we explored the role of WASF3 in the sensitivity of gastric cancer to oxaliplatin, and the underlying mechanisms. METHODS We silenced WASF3 using WASF3-siRNA in MGC803 cells. Then, CCK-8, flow cytometry and transwell assay were performed to study the effect of WASF3 silencing on proliferation, migration, invasiveness and apoptosis of MGC803 cells. Moreover, we evaluated the potential mechanism in vitro to determine the sensitization to oxaliplatin induced by WASF3. RESULTS WASF3 silencing by small interfering RNA inhibited the proliferation, migration and invasiveness of gastric cancer cells. We also observed that WASF3 knockdown promoted cell apoptosis and enhanced oxaliplatin sensitivity. Furthermore, the sensitization to oxaliplatin induced by WASF3 knockdown depended on the inhibition of Atg12-mediated autophagy. CONCLUSIONS Our analysis demonstrates WASF3 targeting is a new potential therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Yanli Nie
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China; Department of Gastrointestinal Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinjun Liang
- Department of Gastrointestinal Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanhe Liu
- Department of Gastrointestinal Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Guo
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Fang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuxiang Zhou
- Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.
| |
Collapse
|
9
|
Lin L, Liu Y, Pan C, Zhang J, Zhao Y, Shao R, Huang Z, Su Y, Shi M, Bin J, Liao Y, Li N, Wang C, Liao W. Gastric cancer cells escape metabolic stress via the DLC3/MACC1 axis. Am J Cancer Res 2019; 9:2100-2114. [PMID: 31037159 PMCID: PMC6485279 DOI: 10.7150/thno.29538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
Metabolic stress usually occurs in rapidly growing gastric cancer (GC) when the energy demand exceeds the supply. Interestingly, cancer cells can somehow escape this stress. Some small Rho GTPases regulating cell migration can be activated by metabolic stress. DLC3 is a RhoA-specific GTPase-activating protein of unclear function in cancer. We hypothesized that it participated in metabolic stress escape. Methods: Metabolic stress in GC cells was induced by glucose deprivation, and DLC3 expression was detected. Based on the prognostic value, cell viability, motility and glycolysis were detected in DLC3 differently expressed GC cells in vitro and in vivo. DLC3 downstream targets were screened and verified. Chemotactic ability was evaluated to study DLC3 and its downstream signaling on metabolic stress escape. In addition, therapeutic strategies targeting DLC3 were explored. Results: DLC3 expression was lowered by metabolic stress in GC cells. DLC3 downregulation indicated poor cancer prognosis, and silencing DLC3 promoted GC cell proliferation and invasion. MACC1, an oncogene promoting GC growth and metastasis, was proved to be the downstream target of DLC3. Low DLC3 expression and high MACC1 expression indicated high recurrence rate after GC resection. DLC3 transcriptionally inhibited MACC1 expression via RhoA/JNK/AP-1 signaling, and subsequently suppressed GC cell glycolysis and survival under metabolic stress. The DLC3/MACC1 axis modulated the chemotaxis of GC cells from energy deficient area to glucose abundant area. Finally, lovastatin was found to be a promising therapeutic drug targeting the DLC3/MACC1 axis. Conclusions: The DLC3/MACC1 axis modulates GC glycolysis and chemotaxis to escape glucose deprivation. Lovastatin may inhibit GC by targeting the DLC3/MACC1 axis.
Collapse
|
10
|
Huang Z, Liu Y, Yang C, Li X, Pan C, Rao J, Li N, Liao W, Lin L. Combined neutrophil/platelet/lymphocyte/differentiation score predicts chemosensitivity in advanced gastric cancer. BMC Cancer 2018; 18:515. [PMID: 29720123 PMCID: PMC5932840 DOI: 10.1186/s12885-018-4414-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/19/2018] [Indexed: 12/17/2022] Open
Abstract
Background Gastric cancer is common in developing regions, and we hope to find out an economical but practical prognostic indicator. It was reported that pre-treatment peripheral neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR), as well as differentiation status, were associated with cancer progression. Hence, we introduced a novel combined Neutrophil/platelet/lymphocyte/differentiation Score (cNPLDS) to improve the prediction value of palliative chemotherapeutic response in advanced gastric cancer. Methods According to statistical sample size estimation, 136 primary diagnosed unresectable advanced ptaients were included for a retrospective study. The follow-up end-point was progression free survival (PFS) during the first-line palliative chemotherapy. Differentiation stratified patients into well, medium and poor groups by score 1 to 3, while patients with neither elevated NLR and PLR, only one elevated, or both elevated were of the combined NLR-PLR score (cNPS) 1 to 3, respectively. The cNPLDS was calculated by multiplying the tumor differentiation score and cNPS. Results Determined by the receiver operating characteristic (ROC) curve, the optimal cut-off points for NLR and PLR were 3.04 and 223. Through univariate analysis and survival analysis, poor differentiation, high NLR, high PLR, high cNPS, and high cNPLDS respectively indicated inferior PFS during the first-line palliative chemotherapy. Patients were furhter classified into low to high risk groups by cNPLDS. Groups of elevated NLR, PLR, cNPS, and cNPLDS showed lower disease control rate. Compared to other parameters, cNPLDS significantly improved the accuracy in predicing the first-progression. Conclusions This study indicates that the novel parameter cNPLDS is superior to NLR or PLR alone, or even cNPS, in predicting the first-line chemosensitivity in advanced gastric cancer.
Collapse
Affiliation(s)
- Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yantan Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chen Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyin Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Changqie Pan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jinjun Rao
- Key laboratory of new drug screening of Guangdong Province, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Nailin Li
- Department of Medicine-Solna, Karolinska Institute, Clinical Pharmacology Group, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Wen Q, Chen Z, Chen Z, Chen J, Wang R, Huang C, Yuan W. EphA2 affects the sensitivity of oxaliplatin by inducing EMT in oxaliplatin-resistant gastric cancer cells. Oncotarget 2018. [PMID: 28624791 PMCID: PMC5564621 DOI: 10.18632/oncotarget.18208] [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] [Indexed: 12/14/2022] Open
Abstract
Erythropoietin-producing hepatocellular receptor A2 (EphA2) is upregulated in gastric cancer tissues and cells, which is accompanied by epithelial–mesenchymal transition (EMT). The current study was designed to establish the oxaliplatin-resistant human gastric cancer cell line SGC-7901/L-OHP, to determine if EMT in these cells could be reversed, and to determine if the susceptibility of these cells to oxaliplatin was affected by silencing EphA2 expression. We found that EphA2 expression levels were upregulated in gastric cancer and associated with chemotherapy sensitivity. EphA2 and the EMT molecular markers N-cadherin and Snail were upregulated in SGC-7901/L-OHP cells, while silencing of EphA2 using small interfering RNA had the opposite effect. Moreover, silencing of EphA2 inhibited cell migration and invasion, and significantly enhanced the sensitivity of oxaliplatin-resistant gastric cancer cells to oxaliplatin. These observations demonstrate that EphA2 affects the sensitivity to oxaliplatin by inducing EMT in oxaliplatin-resistant gastric cancer cells.
Collapse
Affiliation(s)
- Qiaocheng Wen
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zihua Chen
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhikang Chen
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiang Chen
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ran Wang
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Changhao Huang
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Yuan
- General Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Peng XC, Zeng Z, Huang YN, Deng YC, Fu GH. Clinical significance of TM4SF1 as a tumor suppressor gene in gastric cancer. Cancer Med 2018; 7:2592-2600. [PMID: 29665316 PMCID: PMC6010756 DOI: 10.1002/cam4.1494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 12/14/2022] Open
Abstract
Transmembrane‐4‐L‐six‐family member‐1 (TM4SF1), a tumor‐associated antigen, is overexpressed in most epithelial cell carcinomas and a potential target for antibody‐mediated therapy. However, the role of TM4SF1 in gastric cancer has not been elucidated. The aim of this study was to investigate the clinical significance of TM4SF1 expression in gastric carcinoma (GC) tissues using 152 GC tissue samples and matched adjacent nontumor tissue samples analyzed by immunohistochemistry, and 13 fresh GC tissue samples analyzed by Western blotting. The results showed that TM4SF1 was heterogeneously expressed in normal gastric mucosa, with a high expression rate in fundus mucosa. Higher levels and strong expression rate of TM4SF1 were associated with GC tissues of higher‐grade differentiation. TM4SF1 levels were lower in gastric cancer tissues than gastric noncancerous tissues. Expression of TM4SF1 was not correlated with USP10 (P = 0.157), S100A12 (P = 0.479), p53 (P = 0.249), or Ki67 (P = 0.166) in GC. The expression of TM4SF1 was significantly and negatively correlated with depth of invasion (P = 0.031), nodal metastasis (P = 0.042), TNM stage (P = 0.030), and Lauren classification (P = 0.026). There was no significant correlation between TM4SF1 expression and age, gender, tumor size, or distant metastasis (P > 0.05). The expression of TM4SF1 was associated with well overall survival (P = 0.0164). The 5‐year survival rate for patients with GC showing TM4SF1 positive was 58.82% (10/17), and the median survival time was 78 months, higher than that (12.90%, 12/93) of patients who were TM4SF1 negative, whose median survival time was 62 months. These data suggested that low expression of TM4SF1 is associated with carcinogenesis and development, tumor progression and invasion of gastric cancer, and poor overall survival of patients with GC. TM4SF1 is a tumor suppressor for GC and a novel prognostic marker for patients with GC.
Collapse
Affiliation(s)
- Xing-Chun Peng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China.,School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, No.99, Ziyang Road, Wuchang District, Wuhan, 430060, Hubei Province, China
| | - Yu-Ning Huang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China
| | - Yun-Chao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No.99, Ziyang Road, Wuchang District, Wuhan, 430060, Hubei Province, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China
| |
Collapse
|
13
|
Ma H, Wu Z, Peng J, Li Y, Huang H, Liao Y, Zhou M, Sun L, Huang N, Shi M, Bin J, Liao Y, Rao J, Wang L, Liao W. Inhibition of SLC1A5 sensitizes colorectal cancer to cetuximab. Int J Cancer 2018; 142:2578-2588. [PMID: 29363109 DOI: 10.1002/ijc.31274] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/13/2017] [Accepted: 01/09/2018] [Indexed: 12/15/2022]
Abstract
Cetuximab resistance is a key barrier in treating metastatic colorectal cancer (mCRC). Targeting of metabolic resources import could resensitize drug-resistant cancer cells to anticancer treatments. Here we showed that the expression of the glutamine transporter solute carrier 1 family member 5 (SLC1A5) in clinical CRC samples of patients resisted to cetuximab was significantly higher than in those of patients responded to cetuximab. Inhibition of SLC1A5 by shRNA-mediated gene silencing or pharmacological inhibitor significantly suppressed the growth of CRC. Moreover, inhibition of SLC1A5 significantly enhanced the inhibitory efficacy of cetuximab on CRC proliferation both in vitro and in vivo. Mechanistically, SLC1A5 inhibition facilitated EGFR degradation through the ubiquitin-proteasome pathway, and decreased the expression of nuclear EGFR, both of which might have contribution to the improved response to cetuximab. This study provides the metabolic molecule SLC1A5 as a potential therapeutic target to increase the efficacy of cetuximab on CRC.
Collapse
Affiliation(s)
- Huanrong Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenzhen Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianjun Peng
- Department of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongxiang Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minyu Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinjun Rao
- Key laboratory of new drug screening of Guangdong province, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|