1
|
Liu C, Chen C, Lai H, Liang H, Zhong S, Guo G, Wang L, Li L. A New Method for Early Screening of Gastric Cancer (G17 and CA724 Dual-Labeled Time-Resolved Fluorescence Immunoassay). COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1704948. [PMID: 35912162 PMCID: PMC9328981 DOI: 10.1155/2022/1704948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022]
Abstract
Gastric carcinoma (GC) is one of the most common malignancies in the world with the great early screening challenges. The study is aimed at establishing a new detection method for early screening GC using time-resolved fluorescence immunoassay (TRFIA) via quantitative detection of gastrin-17 (G-17) and carbohydrate antigen 724 (CA724) in serum. Time-resolved analyzer measured the fluorescence intensity. The standards of G-17/CA724 were used for drawing the standard curve, which is used to calculate the concentration of G-17 and CA724 in serum sample. The sensitivity for G-17 was 0.54 pg/mL and for CA724 was 0.28 U/mL with a wide-range analyze concentration (0.1-1000) pg/mL or U/mL. The average recoveries ranged from 100.52% to 110.30% for G-17 and 103.02% to 116.00% for CA724. All CVs of the intra- and interassay were below 10% with high specificity. There was a high Pearson coefficient between this TRFIA method and the commercially available kits (Pearson r 0.9117 for G-17 and 0.9449 for CA724). Additionally, the cutoff value was 88.41 pg/mL and 5.47 U/mL for CA724 in health subjects. This study established a TRFIA method for simultaneous detection of the concentrations of G-17 and CA724 in serum, which provide a new method for sensitive, accurate, and specific early screening of gastric cancer.
Collapse
Affiliation(s)
- Chang Liu
- Heilongjiang Academy of Chinese Medicine Sciences, Harbin, 150036, China
| | - Cuicui Chen
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Hongrui Lai
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Huankun Liang
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Shuhai Zhong
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Guiling Guo
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Lei Wang
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| | - Laiqing Li
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou 510663, China
| |
Collapse
|
2
|
Abbas HS, Abo Zeina EA, Radwan HA, Shati AA, Alfaifi MY, Elbehairi SEI. Efficient Synthesis and Biological Evaluation of some new series of pyridine derivatives: Promising and Potent New Class of Anticancer Agents. J Heterocycl Chem 2022. [DOI: 10.1002/jhet.4533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Hebat‐Allah S. Abbas
- Chemistry Department, Faculty of Science King Khalid University Abha Saudi Arabia
- Photochemistry Department National Research Centre Dokki Cairo Egypt
| | - Esraa A. Abo Zeina
- Chemistry Department, Faculty of Science King Khalid University Abha Saudi Arabia
| | - Hayam A. Radwan
- Chemistry Department, Faculty of Women of Arts, Sciences and Educatin Ain Shams University Cairo, Egypt Abidah Saudi Arabia
| | - Ali A. Shati
- Biology Department, Faculty of Science King Khalid University Abha Saudi Arabia
| | - Mohammad Y. Alfaifi
- Biology Department, Faculty of Science King Khalid University Abha Saudi Arabia
| | - Serag Eldin I. Elbehairi
- Biology Department, Faculty of Science King Khalid University Abha Saudi Arabia
- Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El‐Zeraa St., Agouza Giza Egypt
| |
Collapse
|
3
|
A 1,10-phenanthroline derivative selectively targeting telomeric G-quadruplex induces cytoprotective autophagy, causing apoptosis of gastric cancer cells. Life Sci 2021; 287:120095. [PMID: 34715135 DOI: 10.1016/j.lfs.2021.120095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 01/14/2023]
Abstract
AIMS This study aimed to evaluate the ability of compound 13d to induce autophagy and to promote apoptosis of tumor cells and its interaction mechanism. MATERIALS AND METHODS Using CCK-8 assay, transwell assay, fluorescence resonance energy transfer melting analysis (FRET), transmission electron microscopy, flow cytometry assay, immunofluorescence assay, Western blot analysis, and wound healing assay. KEY FINDINGS The results indicated that compound 13d could induce autophagy and apoptosis of gastric cancer cells. Moreover, the findings of CCK-8 assay, colony formation, migration and invasion assay, and wound healing assay revealed that compound 13d would effectively inhibit cell proliferation, migration, and invasion. Its IC50 value is about 2.4 μM against gastric cancer cells, which is similar to positive drug‑platinum. 13d specific induction of telomere G-quadruplex formation was proved in extracellular FRET melting assay, and indirectly affected telomerase activity. G-quadruplex formation promoted cell apoptosis and autophagy. Upon incorporating the autophagy inhibitors 3-MA and HCQ, the expression of the autophagy marker protein LC3 was then checked, suggesting that the compound 13d influences the autophagy flux. Furthermore, knocking down the autophagy-related gene Atg5 to reduce the level of autophagy enhances the anti-tumor activity and increases apoptotic cells' proportion. Mechanistic experiments have shown that blocking the Akt/m-TOR signal pathway plays a crucial role in autophagy and G-quadruplex induced telomere dysfunction. DNA damage is the leading cause of autophagy. Compound 13d combined with autophagy inhibitor can inhibit tumor cells more effectively. SIGNIFICANCE Our findings demonstrate that compound 13d as a telomeric G-quadruplex ligand induces Telomere dysfunction, DNA damage response, autophagy, and apoptosis in gastric cancer cells by blocking the Akt/m-TOR signaling pathway.
Collapse
|
4
|
Tyczyńska M, Kędzierawski P, Karakuła K, Januszewski J, Kozak K, Sitarz M, Forma A. Treatment Strategies of Gastric Cancer-Molecular Targets for Anti-angiogenic Therapy: a State-of-the-art Review. J Gastrointest Cancer 2021; 52:476-488. [PMID: 33761051 PMCID: PMC8131337 DOI: 10.1007/s12029-021-00629-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2021] [Indexed: 12/19/2022]
Abstract
Purpose Recent studies have suggested that molecular targets for the anti-angiogenic therapy might constitute a basis for additional therapy in gastric cancer treatment. A vast number of molecules, receptors, pathways, specific interactions, and thus strategies that target gastric cancer angiogenesis specifically have been reported in numerous research articles and clinical trials. Methods We conducted a systematic literature review of molecularly targeted treatment strategies in gastric cancer on the following databases—PubMed, Google Scholar, and Scopus—on September 20, 2020. Multiple articles and evaluations were searched for studies reporting newly found and promising molecular anti-angiogenic therapy pathways. Eventually, 39 articles regarding the anti-angiogenic therapy in gastric cancer were included in the final analysis. Results As a consequence of the release of the pro-angiogenic molecules from the tumour cells, gastric cancer presents high angiogenic capability. Therefore, potential schemes for future treatment strategies include the decrease of the process ligands as well as the expression of their receptors. Moreover, the increase in the angiogenic inhibitor levels and direct aim for the inner walls of the endothelial cells appear as a promising therapeutic strategy. Beyond that, angiogenesis process inhibition seems to indirectly exaggerate the effects of chemotherapy in the considered patients. Conclusions The anti-angiogenic treatment in gastric cancer patients evaluates its significance especially in the early stages of the malignancy. The studies conducted so far show that most of the meaningful angiogenic factors and receptors with the potential molecular pathways should be further evaluated since they could potentially play a substantial role in future therapies.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Paweł Kędzierawski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Kaja Karakuła
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland
| | - Jacek Januszewski
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | - Krzysztof Kozak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
5
|
Ortiz N, Díaz C. Mevalonate pathway as a novel target for the treatment of metastatic gastric cancer. Oncol Lett 2020; 20:320. [PMID: 33093924 PMCID: PMC7573883 DOI: 10.3892/ol.2020.12183] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric mucosa tumors may present as two distinct major entities: Diffuse and intestinal subtypes. There is no standard treatment for advanced or metastatic gastric cancer. The mevalonate pathway and cholesterol homeostasis are important processes in cancer cells that may be highly relevant in terms of cell growth, survival and metastatic potential. Two model cell lines representing intestinal (NCI-N87) and diffuse (Hs746T) metastatic gastric tumor histological subtypes were treated with different drugs that alter membrane lipid metabolism to determine whether cell proliferation, viability and migration were affected. The results indicated that the cells exhibited significant differences in proliferation when treated with the cholesterol-lowering drug simvastatin, but not with terbinafine, another compound that affects cholesterol synthesis. Only simvastatin affected migration in both cell lines. Reposition studies with mevalonolactone, farnesyl pyrophosphate and geranylgeranyl pyrophosphate in the presence of high and low FBS concentrations indicated that both isoprenoids and cholesterol reversed the antiproliferative effects of simvastatin in gastric cancer cells. The cell lines used in the present study had different sensitivities to several potential anti-neoplastic agents that affect the synthesis of membrane lipids. The diffuse gastric cancer cells were particularly sensitive to simvastatin, suggesting it as an option for combination treatment.
Collapse
Affiliation(s)
- Natalia Ortiz
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica
| | - Cecilia Díaz
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica.,Institute Clodomiro Picado, Faculty of Microbiology, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica
| |
Collapse
|
6
|
Chen Q, Chen R, Dong Y. Inhibitory effect of endostar combined with radiotherapy on gastric cancer animal models. World J Surg Oncol 2020; 18:165. [PMID: 32669133 PMCID: PMC7364530 DOI: 10.1186/s12957-020-01937-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/29/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Inhibitory effect of endostar combined with radiotherapy on gastric cancer (GC) animal models and its effect on transforming growth factor-β1 (TGF-β1) and inter-leukin-10 (IL-10) were evaluated. METHODS Forty mice of GC model xenograft tumors were prepared and randomly divided into blank control group, endostar group, radiotherapy group, and endostar combined with radiotherapy group (combination group). From the 14th day, a vernier caliper was used for measuring the long and short diameters of the xenograft tumors. The formula V = ab2/2 was used for calculating the tumor volume and to obtain its average value. Tumor growth curves were plotted to calculate the tumor inhibition rate. The growth of xenograft tumors and the behavioral changes of mice were observed. Enzyme-linked immunosorbent assay (ELISA) was used for detecting the expression levels of IL-10 and TGF-β1. RESULTS The tumor growth in the combination group was significantly inhibited, and the tumor volume was the smallest compared with the other groups (p < 0.05). Compared to the blank control group, the tumor inhibition rate was 11.8% in endostar group, 33.0% in radiotherapy group, and 52.1% in combination group (p < 0.01). Endostar combined with radiotherapy had an interaction in decreasing the expression levels of TGF-β1 and IL-10 (F = 4.35 and 5.12, p < 0.05). Leucocyte count was significantly higher in control and combination groups than that in endostar and radiotherapy groups. The body weight of mice in endostar and radiotherapy groups decreased after treatment (p < 0.05). The body weight of mice after treatment in control and combination groups increased, with a statistically significant difference compared to that before treatment (p < 0.05). There was a statistically significant difference among all groups after treatment (F = 198.1, p < 0.01). CONCLUSIONS Endostar combined with radiotherapy can inhibit tumor growth and downregulate the expression levels of TGF-β1 and IL-10 through synergistic action.
Collapse
Affiliation(s)
- Qitian Chen
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, Hubei, People's Republic of China
| | - Ran Chen
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, Hubei, People's Republic of China
| | - Youhong Dong
- Department of Oncology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, 15 Jiefang Road, Xiangyang, 441000, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Wang L, Wen X, Luan F, Fu T, Gao C, Du H, Guo T, Han J, Huangfu L, Cheng X, Ji J. EIF3B is associated with poor outcomes in gastric cancer patients and promotes cancer progression via the PI3K/AKT/mTOR signaling pathway. Cancer Manag Res 2019; 11:7877-7891. [PMID: 31686906 PMCID: PMC6708883 DOI: 10.2147/cmar.s207834] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose Eukaryotic translation initiation factor (EIF) plays a vital role in protein synthesis. EIF3B is a core subunit of the EIF3 family, and is overexpressed in many tumors. EIF3B is associated with an unfavorable prognosis, as well as the genesis and development of tumors. However, the potential role of EIF3B in gastric cancer (GC) remains unknown. In the current study, we explored the clinical significance and the possible mechanism of EIF3B in the progression of GC. Methods EIF3B expression was analyzed in 78 GC tissue samples through quantitative PCR and in 94 GC tissue samples through immunohistochemistry (IHC) staining. The correlation between EIF3B and clinicopathological features was analyzed in GC tissues. The role of EIF3B in GC progression was investigated through in vitro and in vivo assays. Results EIF3B expression was upregulated in GC tissues (73.4%, IHC). High expression of EIF3B was significantly correlated with the depth of tumor invasion, lymph node metastasis and TNM stage (P=0.000, 0.000 and 0.000, respectively). Multivariate analysis indicated that GC patients with high EIF3B expression suffered a poorer 5-year survival. EIF3B promoted GC cell proliferation and was strongly associated with proliferating cell nuclear antigen (PCNA) expression in GC samples (P=0.009). It also enhanced tumor cell migration and invasion, which were affected through epithelial-mesenchymal transition (EMT) and the Stat3 signaling pathway. Knockdown of EIF3B in GC cells suppressed the growth of xenograft tumors and lung metastatic colonization in vivo. Furthermore, gene set enrichment analysis (GSEA) and Western blot results demonstrated that EIF3B activated the PI3K/AKT/mTOR signaling pathway. Conclusion Our results suggest that EIF3B plays an oncogenic role in GC progression and serves as an independent prognostic factor for GC patients.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China.,Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Xianzi Wen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fengming Luan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Tao Fu
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Chao Gao
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ting Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jing Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Longtao Huangfu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Xiaojing Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China.,Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
8
|
Barati N, Momtazi-Borojeni AA, Majeed M, Sahebkar A. Potential therapeutic effects of curcumin in gastric cancer. J Cell Physiol 2019; 234:2317-2328. [PMID: 30191991 DOI: 10.1002/jcp.27229] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Despite recent advancements in understanding of the biology of gastric cancer, treatment of patients with advanced gastric cancer remains a major problem. Among different type of phytochemicals, curcumin, a welltable -known phytochemical, has been shown to be a promising cancer chemopreventive agent. Pharmacokinetics, safety, and efficacy of curcumin have been evaluated in several clinical trials against numerous diseases, and for the treatment of human cancer. In the present review, we have collected in vitro and in vivo investigations and studied the chemosensitizing and anticancer effects of curcumin against the gastric cancer cells. In summary, curcumin has been found to have efficient chemosensitizing effect and also inhibits viability, proliferation, and migration of gastric cancer cells mainly via cell cycle arrest and induction of apoptosis by both mitochondrial-dependent and -independent pathways.
Collapse
Affiliation(s)
- Nastaran Barati
- Deputy of Research and Technology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir A Momtazi-Borojeni
- Nanotechnology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Irantab
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Zhang Y, Liu S, Feng Q, Huang X, Wang X, Peng Y, Zhao Z, Liu Z. Perilaldehyde activates AMP-activated protein kinase to suppress the growth of gastric cancer via induction of autophagy. J Cell Biochem 2019; 120:1716-1725. [PMID: 30378150 DOI: 10.1002/jcb.27491] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND AIM Perillaldehyde (PAH), one of the major oil components in Perilla frutescens, is very critical to health maintenance, for a wide range of human chronic diseases, including cancers. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. This study was designed to explore whether PAH prevents gastric cancer growth and to investigate the molecular mechanism. METHODS AND RESULTS In cultured mouse gastric cancer cell line MFCs and human gastric cancer cell lines GC9811-P, PAH activated AMPK by increasing the Thr172 phosphorylation and activity in a time-/concentration-dependent manner. Furthermore, incubation of MFCs with PAH also increased autophagy as determined by monodansylcadaverine (MDC) staining, which was reversed by AMPK inhibitor compound C. PAH further decreased MFCs cell survival, which was abolished by compound C or autophagy inhibitor 3-Methyladenine (3-MA). In vivo studies indicated that 4-week administration of PAH (100 mg/kg/day) suppressed the growth of gastric cancer and increased the levels of autophagy-related proteins, including beclin-1, LC3-II, cathepsin, caspase-3, p53, and cathepsin in tumors isolated from the xenograft model of gastric cancer in mice. Moreover, these anticancer effects produced by PAH were abolished by coadministration of compound C or 3-MA in vivo. CONCLUSIONS PAH increases AMPK phosphorylation and activity to induce gastric cancer cell autophagy to inhibit the growth of gastric cancer. In perspective, therapy of PAH should be applied to treat patients with gastric cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Suosi Liu
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Qin Feng
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Xiuyun Huang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Xiangyang Wang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Ya Peng
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Zhihong Zhao
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Zhan Liu
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
10
|
Li L, Cui Y, Ye L, Zhao Z, Jiang WG, Ji J. Psoriasin overexpression confers drug resistance to cisplatin by activating ERK in gastric cancer. Int J Oncol 2018; 53:1171-1182. [PMID: 29956751 DOI: 10.3892/ijo.2018.4455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/17/2018] [Indexed: 11/06/2022] Open
Abstract
Psoriasin, a member of the S100 multigenic family, which is aberrantly expressed in a variety of human tumors, is considered as an attractive molecular target for cancer treatment. The present study aimed to characterize the role of psoriasin in gastric cancer (GC), the associated pathways through which it contributes to cancer development and progression, and the effect of psoriasin on cellular response to pre-operative chemotherapy in patients with GC. Expression of psoriasin mRNA and protein were analyzed using quantitative polymerase chain reaction and immunohistochemistry of gastric cancer cohorts, respectively. Gastric cancer cell models with differential expression of psoriasin were generated using stable cell lines that overexpressed psoriasin. The in vitro biological functions of the cells in response to psoriasin overexpression and to chemotherapeutic agents were assessed using various cell-based assays. Psoriasin was overexpressed in patients with advanced GC, and high psoriasin levels led to poor clinical outcomes. Increasing psoriasin expression in GC cell lines promoted cell proliferation, migration and invasion in vitro. Furthermore, psoriasin overexpression caused alterations in the levels of epithelial-mesenchymal transition-associated proteins, and activated the extracellular signal-regulated kinase signaling pathway. Additionally, higher levels of psoriasin expression were significantly associated a lack of response to neoadjuvant chemotherapy in patients with GC. Psoriasin overexpression tended to decrease the sensitivity of GC cells to cisplatin, potentially by inhibiting apoptosis or increasing the S-phase population. Taken together, these results indicate that psoriasin may be a promising therapeutic target for GC treatment, and a potential molecular marker to predict patient response to pre-operative chemotherapy.
Collapse
Affiliation(s)
- Liting Li
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Peking University, Beijing 100142, P.R. China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Zehang Zhao
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Peking University, Beijing 100142, P.R. China
| |
Collapse
|
11
|
Wang D, Sun Y, Li W, Ye F, Zhang Y, Guo Y, Zhang DY, Suo J. Antiproliferative effects of the CDK6 inhibitor PD0332991 and its effect on signaling networks in gastric cancer cells. Int J Mol Med 2018; 41:2473-2484. [PMID: 29436583 PMCID: PMC5846637 DOI: 10.3892/ijmm.2018.3460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/20/2017] [Indexed: 12/31/2022] Open
Abstract
PD0332991 (palbociclib/Ibrance®) is a cyclin-dependent kinase (CDK)4/6 inhibitor that has recently been approved for the treatment of estrogen receptor‑positive advanced breast cancer. The present study investigated the antiproliferative effects of PD0332991 on gastric cancer (GC) cells and the underlying molecular mechanisms. The activity of PD0332991 was tested in several GC cell lines, including AGS, KATO‑Ⅲ, NCI‑N87 and HS746T. Growth inhibitory activity of PD0332991, alone or in combination with fluorouracil (5‑FU), was measured by MTT assay. The effects of PD0332991 on cell cycle progression were analyzed by flow cytometry and western blotting. Protein pathway array and Ingenuity Pathway Analysis were used to identify signaling pathways that may mediate the antiproliferative effects of PD0332991. PD0332991 inhibited proliferation in a dose‑dependent manner and enhanced the activity of 5‑FU in all GC cell lines tested. Cells treated with PD0332991 exhibited cell cycle arrest in G1 phase of the cell cycle, whereas the number of cells in G2/M phase was decreased. PD0332991 also inhibited CDK6‑specific phosphorylation of retinoblastoma on Ser780, reduced the expression of cyclin D1, and induced expression of p53 and p27. Furthermore, 31 proteins were identified, the expression of which was significantly altered following treatment with PD0332991 in at least three cell lines. Pathway analysis indicated that the altered proteins were frequently associated with cell death, cell cycle and the molecular mechanism of cancer. The results of the present study indicated that PD0332991 may inhibit cell proliferation via modulation of the cell cycle, and may affect numerous oncogenic signaling pathways. Therefore, PD0332991 may be considered effective for the treatment of GC.
Collapse
Affiliation(s)
| | - Yabin Sun
- Department of Ophthalmology, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Li
- Departments of Gastric and Colorectal Surgery
| | - Fei Ye
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yang Zhang
- Departments of Gastric and Colorectal Surgery
| | - Yuchen Guo
- Departments of Gastric and Colorectal Surgery
| | - David Y. Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Suo
- Departments of Gastric and Colorectal Surgery
| |
Collapse
|
12
|
Liu XF, Hao JL, Xie T, Pant OP, Lu CB, Lu CW, Zhou DD. The BRAF activated non-coding RNA: A pivotal long non-coding RNA in human malignancies. Cell Prolif 2018; 51:e12449. [PMID: 29484737 DOI: 10.1111/cpr.12449] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/13/2018] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) participate in the complex network of cancer and play an important role in tumourigenesis and progression. BRAF activated non-coding RNA (BANCR), a 4-exon transcript of 693-bp, was first discovered as an oncogenic long non-coding RNA in BRAFV600E melanomas cells in 2012 and was related to melanoma cell migration. Besides melanoma, increasing evidence has explored the potential role of BANCR in the development and progression of multiple other human malignancies, such as retinoblastoma, lung cancer, gastric cancer etc. since its discovery. The expression pattern of BANCR varies in different types of cancers, either as a tumour suppressor or as an accelerator. Functional BANCR may serve as a promising biomarker for cancer diagnosis as well as prognosis evaluation. BANCR-targeted intervention may also become a valuable novel therapeutic tool against human malignancies. This review summarized the advanced research progresses concerning the expression and role of BANCR in different human malignancies.
Collapse
Affiliation(s)
- Xiu-Fen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Ji-Long Hao
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Tian Xie
- Department of. Neurosurgery, The People's Hospital of Jilin Province, Jilin, China
| | - Om Prakash Pant
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Cheng-Bo Lu
- Department of Cardiology, The First Hospital of Jiamusi University, Heilongjiang, China
| | - Cheng-Wei Lu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Dan-Dan Zhou
- Department of Radiology, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
13
|
Riihimäki M, Hemminki A, Sundquist K, Sundquist J, Hemminki K. Metastatic spread in patients with gastric cancer. Oncotarget 2018; 7:52307-52316. [PMID: 27447571 PMCID: PMC5239553 DOI: 10.18632/oncotarget.10740] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
Background The epidemiology of metastatic gastric cancer is unexplored because cancer registries seldom cover metastatic involvement apart from “present or not”. We used a novel approach by utilizing Swedish registers to assess metastatic spread in gastric cancer. To our knowledge, this is the first nationwide description of metastases in gastric cancer. Results The most common sites of metastasis were liver (in 48% of metastatic cancer patients), peritoneum (32%), lung (15%), and bone (12%). Metastases to the lung, nervous system, and bone were more frequent in cardia cancer and men, whereas non-cardia cancer more frequently metastasized within the peritoneum. Signet ring adenocarcinomas more frequently metastasized within the peritoneum, bone and ovaries, and less frequently to the lungs and liver compared with generic adenocarcinoma. The liver and the peritoneum were commonly single metastases while lung metastases occurred frequently together with liver metastases. The median survival in metastatic gastric cancer was 3 months, worst among those with bone and liver metastases (2 months). Methods A total of 7,559 patients with gastric cancer were identified. Metastatic patterns and survival depending on sex, age, stage, anatomical location (cardia and non-cardia), and histological type were assessed. Conclusions The patterns of metastasis differ notably depending on histological type. Cardia cancer exhibits a completely different metastatic behavior than non-cardia cancer. Awareness of the differing patterns may guide in tailored diagnosis of metastases. Survivors from cardia cancer would benefit from increased surveillance of extraperitoneal metastases. Bone metastases should be considered in patients with signet ring adenocarcinoma if symptoms emerge.
Collapse
Affiliation(s)
- Matias Riihimäki
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
14
|
Qian Y, Lu S, Shi Y, Zhao X, Yang T, Jin F, Liu Y. Celastrus orbiculatus extracts induce apoptosis and inhibit invasion by targeting the maspin gene in human gastric adenocarcinoma cells. Oncol Lett 2017; 15:243-249. [PMID: 29387218 PMCID: PMC5768137 DOI: 10.3892/ol.2017.7341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/03/2017] [Indexed: 12/17/2022] Open
Abstract
Celastrus orbiculatus Thunb. has been used as a remedy against cancer and inflammatory diseases for thousands of years in China. Maspin is expressed in normal cells and downregulated in prostate tumor cells. The underlying mechanisms between C. orbiculatus extract (COE) and maspin remain unclear. In the present study, 3 target-specific 19–25 nucleotide maspin small interfering RNAs were designed and synthesized to knockdown maspin expression. The effects of COE on MGC-803/maspin− cell proliferation were evaluated by the MTT assay. Apoptosis was measured by flow cytometry. Invasive activity was measured with the Transwell assay and the associated molecular mechanisms were assessed by western blot analysis. The results demonstrated that COE significantly promoted the expression of maspin (P<0.01) to induce apoptosis and inhibit invasion and migration in MGC803 cells. The expression levels of phosphorylated (p)-p38 mitogen-activated protein kinase (MAPK), phospho-extracellular regulated protein kinase (Erk), B cell lymphoma-2-associated X protein and caspase-3 were increased in the MGC-803/maspin− cells in a dose-dependent manner. The Erk, B-cell lymphoma 2, p-Akt, Akt and p-mechanistic target of rapamycin (mTOR) protein in MGC-803/maspin− cells were reduced in a dose-dependent manner. This indicated that COE may inhibit invasion and migration through phosphoinositide 3-kinase/Akt/mTOR and MAPK signaling pathways in MGC-803/maspin− cells. In conclusion, COE has the ability to improve the expression of maspin to induce apoptosis and inhibit invasion and migration in human gastric adenocarcinoma cells.
Collapse
Affiliation(s)
- Yayun Qian
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, P.R. China
| | - Songhua Lu
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Youyang Shi
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Xueyu Zhao
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Ting Yang
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Feng Jin
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Yanqing Liu
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| |
Collapse
|
15
|
Zhou DD, Liu XF, Lu CW, Pant OP, Liu XD. Long non-coding RNA PVT1: Emerging biomarker in digestive system cancer. Cell Prolif 2017; 50. [PMID: 29027279 DOI: 10.1111/cpr.12398] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/22/2017] [Indexed: 12/19/2022] Open
Abstract
The digestive system cancers are leading cause of cancer-related death worldwide, and have high risks of morbidity and mortality. More and more long non-coding RNAs (lncRNAs) have been studied to be abnormally expressed in cancers and play a key role in the process of digestive system tumour progression. Plasmacytoma variant translocation 1 (PVT1) seems fairly novel. Since 1984, PVT1 was identified to be an activator of MYC in mice. Its role in human tumour initiation and progression has long been a subject of interest. The expression of PVT1 is elevated in digestive system cancers and correlates with poor prognosis. In this review, we illustrate the various functions of PVT1 during the different stages in the complex process of digestive system tumours (including oesophageal cancer, gastric cancer, colorectal cancer, hepatocellular carcinoma and pancreatic cancer). The growing evidence shows the involvement of PVT1 in both proliferation and differentiation process in addition to its involvement in epithelial to mesenchymal transition (EMT). These findings lead us to conclude that PVT1 promotes proliferation, survival, invasion, metastasis and drug resistance in digestive system cancer cells. We will also discuss PVT1's potential in diagnosis and treatment target of digestive system cancer. There was a great probability PVT1 could be a novel biomarker in screening tumours, prognosis biomarkers and future targeted therapy to improve the survival rate in cancer patients.
Collapse
Affiliation(s)
- Dan-Dan Zhou
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China.,Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiu-Fen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Cheng-Wei Lu
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Om Prakash Pant
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Dong Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
16
|
Zhu ML, Lu JX, Pan GP, Ping S, Zhao FR, Qi HT, Yu HY, Jian X, Wan GR, Li P. Traditional Chinese medicine Ka-Sai-Ping suppresses the growths of gastric cancers via induction of autophagy. Oncotarget 2017; 8:95075-95082. [PMID: 29221112 PMCID: PMC5707006 DOI: 10.18632/oncotarget.18041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/10/2017] [Indexed: 11/25/2022] Open
Abstract
Traditional Chinese medication is increasingly used to treat a wide range of human chronic diseases like cardiovascular diseases and cancers. This study was designed to explore whether ka-sai-ping (KSP), a novel traditional Chinese medicine developed by us, prevents gastric cancer growths and to investigate the underlying mechanism. The xenograft model of mouse gastric cancer was established by injecting MFCs into nude mouse subcutaneously. Cell autophagy was assessed by MDC staining. Lysosome and mitochondria were detected by Lyso-Tracker Red and Mito-Traker Green staining. Incubation of cultured mouse gastric cancer cell line MFCs with KSP for 48 hours, concentration-dependently reduced cell survivals and activated autophagy, which were accompanied with damaged lysosomes and mitochondria. In vivo studies indicated that KSP therapy (20 ml/kg/day) for two weeks suppressed the growth of gastric cancer, increased the protein levels of LC3-II, beclin-1, cathepsin L, bcl-2, p53, and capase-3 in tumor tissues from the xenograft model of mouse gastric cancer. Importantly, all these effects induced by KSP were abolished by co-administration of autophagy inhibitor 3-MA. In conclusion, KSP activates cell autophagy to suppress gastric cancer growths. Clinically, KSP is potentially considered as a medicine to treat patients with gastric cancer.
Collapse
Affiliation(s)
- Mo-Li Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Jun-Xiu Lu
- Department of Histology and Embryology, Xinxiang Medical University, Xinxiang, China
| | - Guo-Pin Pan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Song Ping
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Fan-Rong Zhao
- San-Quan College of Xinxiang Medical University, Xinxiang, China
| | - Heng-Tian Qi
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Hai-Ya Yu
- Department of Neurology, The People's Hospital of Xishui County, Huangang, Hubei, China
| | - Xu Jian
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Guang-Rui Wan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
Hsu JT, Liao JA, Chuang HC, Chen TD, Chen TH, Kuo CJ, Lin CJ, Chou WC, Yeh TS, Jan YY. Palliative gastrectomy is beneficial in selected cases of metastatic gastric cancer. BMC Palliat Care 2017; 16:19. [PMID: 28288593 PMCID: PMC5348866 DOI: 10.1186/s12904-017-0192-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 03/07/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Salvage chemotherapy is the mainstay of treatment for metastatic gastric cancer (mGC). This study aimed to clarify the effects of palliative gastrectomy (PG) and identify prognostic factors in mGC patients undergoing PG. METHODS This was a retrospective review of 333 mGC patients receiving PG or a non-resection procedure (NR) between 2000 and 2010. Clinicopathological factors affecting the prognosis of these patients were collected prospectively and analyzed. RESULTS One hundred and ninety-three patients underwent PG and 140 NR. The clinicopathological characteristics were comparable between the two groups except for metastatic pattern. There were no significant differences in postoperative morbidity and mortality between the two groups. The PG group had a significantly longer median overall survival compared with the NR group (7.7 months vs. 4.9 months). In the PG group, age ≤58 years, preoperative albumin level >3 g/dL, ratio of metastatic to examined lymph nodes ≤0.58, and administration of chemotherapy were independent prognostic factors in multivariate analysis. CONCLUSIONS Patients undergoing PG had better outcomes than those undergoing NR. Among the patients undergoing resection, age ≤58 years, a better preoperative nutritional status, less nodal involvement and postoperative chemotherapy independently affected patient survival.
Collapse
Affiliation(s)
- Jun-Te Hsu
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, #5, Fushing Street, Kweishan District, Taoyuan City, 333 Taiwan
| | - Jian-Ann Liao
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, #5, Fushing Street, Kweishan District, Taoyuan City, 333 Taiwan
| | - Huei-Chieh Chuang
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Tai-Di Chen
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Tsung-Hsing Chen
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Chia-Jung Kuo
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Chun-Jung Lin
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, 333 Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, #5, Fushing Street, Kweishan District, Taoyuan City, 333 Taiwan
| | - Yi-Yin Jan
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, #5, Fushing Street, Kweishan District, Taoyuan City, 333 Taiwan
| |
Collapse
|
18
|
The long noncoding RNA PVT1 functions as a competing endogenous RNA by sponging miR-186 in gastric cancer. Biomed Pharmacother 2017; 88:302-308. [PMID: 28122299 DOI: 10.1016/j.biopha.2017.01.049] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/07/2017] [Accepted: 01/09/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Recent evidence has highlighted the key regulatory roles of long non-coding RNAs (lncRNAs) in tumor development and progression including gastric cancer (GC).The long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) has been identified as an oncogene in some tumors. However, the potential biological roles and regulatory mechanisms of PVT1 involved in GC remained poorly understood. METHODS Quantitative real-time PCR (QRT-PCR) was used to determine the expression of PVT1 and miR-186 in GC tissues. The MTT cell proliferation and transwell invasion assays were used to detect the cell proliferation and invasion abilities. Western-blotting analysis was used to detect the protein expression of PCNA and HIF-1α. To understand the tumorigenic mechanism of PVT1, luciferase reporter assays were performed to evaluate whether the miR-186 was a target of PVT1 in GC cells. RESULTS In the current study, we showed that PVT1 expression was markedly upregulated in GC tissues and cell lines, and high expression levels of PVT1 were obviously correlated with advanced tumor stage and lymph node metastasis. Further functional experiments indicated up-regulation of PVT1 promoted the GC cell proliferation and invasion, while down-regulation of PVT1 inhibited cell proliferation and invasion. In addition, PVT1 could directly interact with miR-186 in GC cells and this interaction lead to the inhibition of downstream of HIF-1α expression. CONCLUSIONS These results suggested that PVT1 acted as a key role in GC pathogenesis and may serve as a potential therapeutic target for GC.
Collapse
|
19
|
Li S, Li Z, Guo T, Xing XF, Cheng X, Du H, Wen XZ, Ji JF. Maternal embryonic leucine zipper kinase serves as a poor prognosis marker and therapeutic target in gastric cancer. Oncotarget 2017; 7:6266-80. [PMID: 26701722 PMCID: PMC4868755 DOI: 10.18632/oncotarget.6673] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/07/2015] [Indexed: 12/28/2022] Open
Abstract
Maternal embryonic leucine zipper kinase (MELK) is upregulated in a variety of human tumors, and is considered an attractive molecular target for cancer treatment. We characterized the expression of MELK in gastric cancer (GC) and measured the effects of reducing MELK mRNA levels and protein activity on GC growth. MELK was frequently overexpressed in primary GCs, and higher MELK levels correlated with worse clinical outcomes. Reducing MELK expression or inhibiting kinase activity resulted in growth inhibition, G2/M arrest, apoptosis and suppression of invasive capability of GC cells in vitro and in vivo. MELK knockdown led to alteration of epithelial mesenchymal transition (EMT)-associated proteins. Furthermore, targeting treatment with OTSSP167 in GC patient-derived xenograft (PDX) models had anticancer effects. Thus, MELK promotes cell growth and invasiveness by inhibiting apoptosis and promoting G2/M transition and EMT in GC. These results suggest that MELK may be a promising target for GC treatment.
Collapse
Affiliation(s)
- Shen Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China.,Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiao-Fang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaojing Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xian-Zi Wen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jia-Fu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital & Institute, Beijing, China.,Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
20
|
Sun W, Yan L. Gastric cancer: current and evolving treatment landscape. CHINESE JOURNAL OF CANCER 2016; 35:83. [PMID: 27581465 PMCID: PMC5006607 DOI: 10.1186/s40880-016-0147-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/04/2016] [Indexed: 12/23/2022]
Abstract
Gastric (including gastroesophageal junction) cancer is the third leading cause of cancer-related death in the world. In China, an estimated 420,000 patients were diagnosed with gastric cancer in 2011, ranking this malignancy the second most prevalent cancer type and resulting in near 300,000 deaths. The treatment landscape of gastric cancer has evolved in recent years. Although systemic chemotherapy is still the mainstay treatment of metastatic disease, the introduction of agents targeting human epidermal growth factor receptor 2 and vascular endothelial growth factor/vascular endothelia growth factor receptor has brought this disease into the molecular and personalized medicine era. The preliminary yet encouraging clinical efficacy observed with immune checkpoint inhibitors, e.g., anti-programmed cell death protein 1/programmed death-ligand 1, will further shape the treatment landscape for gastric cancer. Molecular characterization of patients will play a critical role in developing new agents, as well as in implementing new treatment options for this disease.
Collapse
Affiliation(s)
- Weijing Sun
- University of Pittsburgh School of Medicine, Pittsburgh, PA, 15232, USA.
| | - Li Yan
- US Chinese Anti-Cancer Association, Martinez, CA, 94553, USA. .,Beijing Cancer Hospital and Institute, Peking University School of Oncology, Beijing, 100142, P. R. China.
| |
Collapse
|
21
|
Xu C, Li M, Zhang L, Bi Y, Wang P, Li J, Jiang X. MicroRNA-205 suppresses the invasion and epithelial-mesenchymal transition of human gastric cancer cells. Mol Med Rep 2016; 13:4767-73. [PMID: 27082508 DOI: 10.3892/mmr.2016.5118] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 11/24/2015] [Indexed: 11/06/2022] Open
Abstract
Distant metastasis is the predominant pattern of gastric cancer (GC) recurrence, and is the most common cause of cancer‑associated mortality. Accumulating evidence has suggested that aberrant activation of epithelial‑mesenchymal transition has a crucial role in the genesis, invasion and metastasis of various types of cancer, including GC. Using Cell Counting kit‑8 and Transwell assays, the effects of microRNA (miR)‑205 on the proliferation, migration and invasion of NCI‑H87 GC cells were determined, and the potential underlying mechanisms were explored. The results of the present study demonstrated that miR‑205, which has been reported to function as a tumor suppressor in various types of cancer, significantly suppressed the migration and invasion of GC cells, which may be correlated with its suppressive effects on EMT. Upon transfection with miR‑205, the epithelial marker CDH1 (E‑cadherin) was upregulated, and the mesenchymal markers CDH2 (N‑cadherin) and vimentin were suppressed. Furthermore, zinc‑finger E‑box‑binding homeobox factor‑1 (ZEB1) was identified as a putative target gene of miR‑205 in GC, which may be associated with its suppressive effects. The results of the present study may provide novel diagnostic and therapeutic options for the treatment of human GC.
Collapse
Affiliation(s)
- Chang Xu
- Department of Radiology, Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Ming'e Li
- Department of Gerontology, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Lin Zhang
- Department of Radiology, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Yueyang Bi
- Department of Respiratory Medicine, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Peiyuan Wang
- Department of Radiology, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Jun Li
- Department of Radiology, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Xingyue Jiang
- Department of Radiology, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
22
|
Kanat O, O’Neil B, Shahda S. Targeted therapy for advanced gastric cancer: A review of current status and future prospects. World J Gastrointest Oncol 2015; 7:401-410. [PMID: 26690491 PMCID: PMC4678387 DOI: 10.4251/wjgo.v7.i12.401] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 09/18/2015] [Accepted: 10/23/2015] [Indexed: 02/05/2023] Open
Abstract
In the West in particular, the vast majority of gastric cancer (GC) patients present with advanced-stage disease. Although combination chemotherapy is still the most important component of treatment for these patients, it confers a modest survival advantage. Recently, increased knowledge of the key molecular signaling pathways involved in gastric carcinogenesis has led to the discovery of specific molecular-targeted therapeutic agents. Some of these agents such as trastuzumab and ramucirumab have changed the treatment paradigm for this disease. In this paper, we will summarize the current clinical status of targeted drug therapy in the management of GC.
Collapse
|
23
|
LI AODI, QU XIUJUAN, LI ZHI, QU JINGLEI, SONG NA, MA YANJU, LIU YUNPENG. Secreted protein acidic and rich in cysteine antagonizes bufalin-induced apoptosis in gastric cancer cells. Mol Med Rep 2015; 12:2926-32. [DOI: 10.3892/mmr.2015.3676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 08/14/2014] [Indexed: 11/06/2022] Open
|
24
|
You T, Gao W, Wei J, Jin X, Zhao Z, Wang C, Li Y. Overexpression of LIMK1 promotes tumor growth and metastasis in gastric cancer. Biomed Pharmacother 2014; 69:96-101. [PMID: 25661344 DOI: 10.1016/j.biopha.2014.11.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/09/2014] [Indexed: 01/07/2023] Open
Abstract
Gastric cancer is the second-leading cause of cancer death in Asia. Despite improvement of therapies, the outcome in patients remains extremely poor because of metastasis. In the present study, we found that LIMK1 is overexpressed in gastric cancer, and its expression level correlate with tumor size, lymph node metastasis and TNM stage. Knockdown of LIMK1 expression could inhibit cell proliferation, migration and invasion in vitro, as well as suppress the activation of FAK/paxillin pathway. Moreover, knockdown of LIMK1 expression retarded tumor growth and peritoneal ametastasis in vivo. This highlights that LIMK1 might be used as a potential target in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Tiangeng You
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Wei Gao
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Jun Wei
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Xiaoli Jin
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Zhongxin Zhao
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Congjun Wang
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China
| | - Yang Li
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, NO. 150 Jimo Rd., Shanghai 200120, People's Republic of China.
| |
Collapse
|
25
|
Zhu YD, Liu YQ, Qian YY, Zhang H, Li GQ, Yang L. Extracts of Celastrus orbiculatus exhibit anti-proliferative and anti-invasive effects on human gastric adenocarcinoma cells. Chin J Integr Med 2014. [PMID: 25382615 DOI: 10.1007/s11655-014-1951-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To assess the effect of Celastrus orbiculatus (COE) on growth, invasion and migration of human gastric cancer MGC-803 cells and to explore the possible mechanism. METHODS The effect of COE on cell viability, apoptosis, adhesion, invasion and migration were studied by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, flow cytometric, cell adhesion and transwell assay, respectively. The activity and expression of matrix metalloproteinase-9 (MMP-9) were determined by gelatin zymography, Western blot and quantitative real-time polymerase chain reaction analysis. Meanwhile, effects of COE on the expression of mitogen-activated protein kinases (MAPKs), serine threonine kinase (Akt), nuclear factor κB (NF-κB) were investigated with Western blot analysis. RESULTS COE inhibited proliferation and induced apoptosis of MGC-803 cells in a dose-dependent manner. When treated with low-toxic (below 80 μg/mL) doses of COE, cell adhesion, invasion and migration were markedly suppressed. Furthermore, the gelatinolytic activity and expression of MMP-9 were also remarkably suppressed in a dose-dependent manner. In addition, upstream signaling pathways, including the phosphatidylinositol-3 kinase (PI3K)/Akt and NF-κB, were suppressed by COE. Additionally, the PI3K/Akt inhibitor, LY294002, in treating MGC-803 cells potently suppressed cell invasion and migration as well as expression of MMP-9. Similarly, the combined treatment with COE and LY294002 showed a synergistic effect compared with the treatment with COE or LY294002 alone in MGC-803 cells. CONCLUSIONS COE inhibits invasion and migration of MGC-803 cells by reducing MMP-9 expression. It also inhibit PI3K/Akt and NF-κB signaling pathways, which may offer a novel approach for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Yao-Dong Zhu
- Institute of Traditional Chinese Medicine and Western Medicine, School of Medicine, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | | | | | | | | | | |
Collapse
|
26
|
Schildberg CW, Weidinger T, Hohenberger W, Wein A, Langheinrich M, Neurath M, Boxberger F. Metastatic adenocarcinomas of the stomach or esophagogastric junction (UICC stage IV) are not always a palliative situation: a retrospective analysis. World J Surg 2014; 38:419-25. [PMID: 24146196 DOI: 10.1007/s00268-013-2293-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gastric cancer is one of the most common cancers. Unfortunately, it is often diagnosed at the advanced stage International Union Against Cancer stage IV. This induced us to carry out an interdisciplinary analysis of this patient group with the Department of Internal Medicine 1. Our aim was to discuss cancers classified initially as unresectable in a meeting of the interdisciplinary tumor board after palliative chemotherapy, and to refer selected patients for surgery after establishing resectability. The outcome of the chemotherapy, operation method, complication rate, and long-term survival were analyzed. METHODS From 1999 to 2008, 76 patients with metastatic gastric cancer or carcinoma of the esophagogastric junction were discussed by the interdisciplinary tumor board of the University of Erlangen and classified initially as unresectable. The patients then received palliative chemotherapy according to the AIO regimen (weekly high-dose 5-fluorouracil/folinic acid [FU/FA] in a 24 h infusion), plus irinotecan. If the tumor was subsequently classified as resectable, the patient underwent either gastric resection or gastrectomy with DII-III dissection. Metastases were resected depending on their location (liver). Peritoneal carcinomatosis was treated additionally by HIPEC. Statistical analysis was with SPSSS version 20. RESULTS Surgical and general complications and hospital mortality were acceptable. There were no cases of anastomotic leak, but one patient died of fulminant pneumonia. The R0 resection rate was 69 %, and four patients had long-term survival of more than 60 months. There were significant survival advantages. CONCLUSIONS Metastatic gastric cancer or carcinoma of the esophagogastric junction can become resectable after downsizing the tumor with palliative chemotherapy. Long-term survival is achieved in some cases. Therefore, every patient with this type of cancer should be discussed by the interdisciplinary tumour board after palliative chemotherapy to provide him with a chance of cure after re-evaluation.
Collapse
Affiliation(s)
- Claus W Schildberg
- Department of Surgery, University of Erlangen/Nürnberg, Krankenhausstrasse 12, 91054, Erlangen, Germany,
| | | | | | | | | | | | | |
Collapse
|
27
|
Zou L, Qian J. Decline of serum CA724 as a probable predictive factor for tumor response during chemotherapy of advanced gastric carcinoma. Chin J Cancer Res 2014; 26:404-9. [PMID: 25232212 DOI: 10.3978/j.issn.1000-9604.2014.07.02] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/20/2014] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To evaluate the predictive value of decline in the serum level of carbohydrate antigen 724 (CA724) on tumor response during the chemotherapy in patients with advanced gastric carcinoma (GC). METHODS The serum CA724 level was determined by electrochemiluminescence immunoassay, while the objective response rate (ORR) was assessed according to response evaluation criteria in solid tumors (RECIST). The association of the changes of serum concentration of CA724 with ORR was analyzed. RESULTS The ORR in CA724 (pretreatment serum level) high and low groups was 32.3% (20/62) and 52.8% (19/36), respectively (P=0.045). The relationship between the reduction of CA724 and the ORR was statistically significant (P=0.044). Receiver operating characteristic (ROC) curve established the best cutoff value of the decrease ratio of CA724 as 20.5%. CONCLUSIONS CA724 decline seems to indicate chemotherapy efficacy in patients with advanced GC, and an average drop of 20.5% in serum CA724 appears to predict the sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Li Zou
- 1 Department of Oncology, 2 Department of Thoracic Surgery, Yancheng City No. 1 People's Hospital, Yancheng 224000, China
| | - Jun Qian
- 1 Department of Oncology, 2 Department of Thoracic Surgery, Yancheng City No. 1 People's Hospital, Yancheng 224000, China
| |
Collapse
|
28
|
The role of hepatectomy in the management of metastatic gastric adenocarcinoma: a systematic review. Surg Oncol 2014; 23:177-85. [PMID: 25263794 DOI: 10.1016/j.suronc.2014.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/03/2014] [Accepted: 08/02/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gastric cancer has a high mortality, with many patients presenting with advanced disease. Many patients who undergo curative gastrectomy will subsequently develop metastatic disease. Hepatectomy has an established place in treating metastases from a variety of cancers but its role in gastric cancer is not clear. This review sought to systematically appraise the literature to establish the role of hepatectomy in treating gastric cancer metastases. METHOD Medline and EMBASE were searched for all papers publishing data on survival of patients with metastatic gastric adenocarcinoma who underwent hepatectomy. RESULTS Seventeen studies with 438 patients were included. There were no randomised controlled trials. Perioperative mortality was 2%, with morbidity between 17 and 60%. Patients with solitary metastases appeared to have better survival. Other favourable survival characteristics included unilobar disease, and metachronous presentation. No advantage was demonstrated with either adjuvant or neoadjuvant chemotherapy. DISCUSSION Few patients with hepatic metastases from gastric cancer are suitable for hepatectomy, but for those suitable there appears to be survival benefit. Patients with synchronous, multiple or bilobar metastases have worse survival. CONCLUSION The evidence supporting the role of hepatectomy in the treatment of hepatic metastases from gastric cancer is weak. However in a selected group there appears to be a survival advantage; patients with solitary metastases had better survival outcomes than those with multiple metastases and metachronous presentation was associated with a better prognosis than synchronous presentation. Hepatectomy should be considered in these patients in the setting of a randomised trial.
Collapse
|
29
|
Cheng Y, Li Y, Liu D, Zhang R, Zhang J. miR-137 effects on gastric carcinogenesis are mediated by targeting Cox-2-activated PI3K/AKT signaling pathway. FEBS Lett 2014; 588:3274-81. [PMID: 25064845 DOI: 10.1016/j.febslet.2014.07.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 12/15/2022]
Abstract
The discovery of microRNAs (miRNAs) provided a new avenue for early diagnosis and treatment of GC. MiR-137 has been reported to be under-expressed and involved in various cell processes. However, the role of miR-137 in GC is less known. In this study, we show that miR-137 is under-expressed in GC and functions as a tumor suppressor through targeting Cyclooxygenase-2 (Cox-2), which subsequently suppresses the activation of PI3K/AKT signaling pathway both in vitro and in vivo. Moreover, restored Cox-2 expression partially abolished the tumor suppressive effects of miR-137 in GC cells, suggesting miR-137 may suppress GC carcinogenesis by targeting Cox-2.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Digestive Diseases, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Yang Li
- Department of Otolaryngology-head and neck surgery, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Dong Liu
- Department of Digestive Diseases, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Rong Zhang
- Department of Digestive Diseases, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Jun Zhang
- Department of Digestive Diseases, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China.
| |
Collapse
|
30
|
Epidermal growth factor-like domain-containing protein 7 (EGFL7) enhances EGF receptor-AKT signaling, epithelial-mesenchymal transition, and metastasis of gastric cancer cells. PLoS One 2014; 9:e99922. [PMID: 24945379 PMCID: PMC4063792 DOI: 10.1371/journal.pone.0099922] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/19/2014] [Indexed: 12/16/2022] Open
Abstract
Epidermal growth factor-like domain-containing protein 7 (EGFL7) is upregulated in human epithelial tumors and so is a potential biomarker for malignancy. Indeed, previous studies have shown that high EGFL7 expression promotes infiltration and metastasis of gastric carcinoma. The epithelial-mesenchymal transition (EMT) initiates the metastatic cascade and endows cancer cells with invasive and migratory capacity; however, it is not known if EGFL7 promotes metastasis by triggering EMT. We found that EGFL7 was overexpressed in multiple human gastric cancer (GC) cell lines and that overexpression promoted cell invasion and migration as revealed by scratch wound and transwell migration assays. Conversely, shRNA-mediated EGFL7 knockdown reduced invasion and migration. Furthermore, EGFL7-overexpressing cells grew into larger tumors and were more likely to metastasize to the liver compared to underexpressing CG cells following subcutaneous injection in mice. EGFL7 overexpression protected GC cell lines against anoikis, providing a plausible mechanism for this enhanced metastatic capacity. In excised human gastric tumors, expression of EGFL7 was positively correlated with expression levels of the mesenchymal marker vimentin and the EMT-associated transcription repressor Snail, and negatively correlated with expression of the epithelial cell marker E-cadherin. In GC cell lines, EGFL7 knockdown reversed morphological signs of EMT and decreased both vimentin and Snail expression. In addition, EGFL7 overexpression promoted EGF receptor (EGFR) and protein kinase B (AKT) phospho-activation, effects markedly suppressed by the EGFR tyrosine kinase inhibitor AG1478. Moreover, AG1478 also reduced the elevated invasive and migratory capacity of GC cell lines overexpressing EGFL7. Collectively, these results strongly suggest that EGFL7 promotes metastasis by activating EMT through an EGFR-AKT-Snail signaling pathway. Disruption of EGFL7-EGFR-AKT-Snail signaling may a promising therapeutic strategy for gastric cancer.
Collapse
|
31
|
Du T, Qu Y, Li J, Li H, Su L, Zhou Q, Yan M, Li C, Zhu Z, Liu B. Maternal embryonic leucine zipper kinase enhances gastric cancer progression via the FAK/Paxillin pathway. Mol Cancer 2014; 13:100. [PMID: 24885567 PMCID: PMC4113179 DOI: 10.1186/1476-4598-13-100] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 04/28/2014] [Indexed: 12/15/2022] Open
Abstract
Background Elevated MELK expression is featured in multiple tumors and correlated with tumorigenesis and tumor development. This study is aimed to investigate the mechanisms of MELK-mediated development of gastric cancer. Methods MELK expression levels in human gastric cancer were determined by quantitative-PCR and immunohistochemistry. The effect of MELK on cell activity was explored by knockdown and overexpression experiments. Cell growth was measured using the CCK-8 assay. Apoptosis and cell cycle distributions were analyzed by flow cytometry. Migration and invasion were tested using a transwell migration assay. Cytoskeletal changes were analyzed by immunofluorescence. To explore the molecular mechanism and effect of MELK on migration and invasion, Western blotting was used to analyze the FAK/Paxillin pathway and pull down assays for the activity of small Rho GTPases. In vivo tumorigenicity and peritoneal metastasis experiments were performed by tumor cell engraftment into nude mice. Results MELK mRNA and protein expression were both elevated in human gastric cancer, and this was associated with chemoresistance to 5-fluorouracil (5-FU). Knockdown of MELK significantly suppressed cell proliferation, migration and invasion of gastric cancer both in vitro and in vivo, decreased the percentages of cells in the G1/G0 phase and increased those in the G2/M and S phases. Moreover, knockdown of MELK decreased the amount of actin stress fibers and inhibited RhoA activity. Finally, knockdown of MELK decreased the phosphorylation of the FAK and paxillin, and prevented gastrin-stimulated FAK/paxillin phosphorylation. By contrast, MELK overexpression had the opposite effect. Conclusions MELK promotes cell migration and invasion via the FAK/Paxillin pathway, and plays an important role in the occurrence and development of gastric cancer. MELK may be a potential target for treatment against gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Bingya Liu
- Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No 197 Ruijin er Road, Shanghai 200025, China.
| |
Collapse
|
32
|
Deng WL, Lv SQ, Ren XJ, Wang XY, Wang QL, Lu M. Effect of Yi-Qi-Fu-Sheng Recipe on mucosal barrier in an orthotopic transplant mouse model of gastric cancer. Shijie Huaren Xiaohua Zazhi 2013; 21:951-956. [DOI: 10.11569/wcjd.v21.i11.951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Yi-Qi-Fu-Sheng Recipe (YQFS) on tumor growth and on intestinal mucosal barrier function in mice with experimental gastric cancer.
METHODS: An orthotopic transplant mouse model of gastric cancer was created by injecting mouse MFC cells. Mice were randomly divided into five groups: NS control group, model group, 5-Fu group, YQFS group and combination therapy group. After intragastric administration of drugs for 14 d, tumor growth and the reduced rate of tumor growth were evaluated, the changes in D-lactic acid, diamine oxidase (DAO), endotoxin and IgG were evaluated by ELISA, and the expression of ZO-1 was detected by gelatin zymography.
RESULTS: Compared to the model group, tumor weight in the YQFS group and combination therapy group were significantly decreased (2.29 g ± 0.14 g vs 1.47 g ± 0.10 g, 1.24 g ± 0.11 g, both P < 0.05). Compared to the model group, D-lactic acid, DAO, endotoxin and IgG were decreased after treatment with YQFS (all P < 0.05). Immunohistochemistry analysis revealed that the levels of ZO-1 in the YQFS group, 5-Fu group and combination therapy group were significantly increased compared to the model group (all P < 0.05).
CONCLUSION: YQFS significantly inhibits tumor growth and protects the intestinal mucosal barrier function in mouse with gastric cancer possibly via mechanisms associated with altering the expression of ZO-1.
Collapse
|