251
|
Amini M, Hejazi M, Ghorban K, Mokhtarzadeh A, Baradaran B. Identification of functional methylated CpG loci in PD-L1 promoter as the novel epigenetic biomarkers for primary gastric cancer. Gene 2020; 772:145376. [PMID: 33359128 DOI: 10.1016/j.gene.2020.145376] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023]
Abstract
Gastric cancer (GC) is considered one of the most lethal malignancies worldwide due to poor prognosis. Aberrant methylation has been demonstrated to be involved in PD-L1 dysregulated expression in human cancers and possesses a great value as a diagnostic biomarker. Given that, in this study, we investigated the methylation status of PD-L1 as a promising biomarker in primary gastric tumors and identified functional CpG loci undergoing aberrant methylation through tumorigenesis of GC. PD-L1 methylation was initially evaluated in-silico using TCGA-STAD dataset. Pearson's correlation analysis was further employed to identify the most significant functional methylated CpG loci of PD-L1 gene in TCGA-STAD patient cohort. Methylation status and its correlation with PD-L1 expression were also validated using q-MSP and qRT-PCR in a set of internal samples, including 25 paired primary gastric tumors and adjacent normal tissues. The obtained results from TCGA-STAD showed that PD-L1 is significantly hypermethylated through gastric tumorigenesis, mostly in two CpG loci overlapping with cg19724470 and cg15837913 probes. Besides, PD-L1 DNA methylation was negatively correlated with PD-L1 expression in tumor samples. Furthermore, hypermethylation of cg19724470 and cg15837913 regions was validated in primary gastric tumors compared to adjacent normal samples. Also, ROC curve analysis illustrated the high diagnostic value of PD-L1 methylation for early detection of GC (AUC = 0.8110). In conclusion, the findings of this study suggested that PD-L1 expression is regulated by methylation in functional CpG loci and its methylation could be considered as a valuable diagnostic target for GC.
Collapse
Affiliation(s)
- Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hejazi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khodayar Ghorban
- Department of Immunology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
252
|
Park YL, Choi JH, Park SY, Oh HH, Kim DH, Seo YJ, So JK, Song K, Cho MS, Chung MW, Hong JY, Kim KH, Myung E, Myung DS, Cho SB, Lee WS, Park D, Joo YE. Engulfment and cell motility 1 promotes tumor progression via the modulation of tumor cell survival in gastric cancer. Am J Transl Res 2020; 12:7797-7811. [PMID: 33437361 PMCID: PMC7791502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/25/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND/AIM Engulfment and cell motility 1 (ELMO1) protein has been implicated in phagocytosis of apoptotic cells, cell migration, neurite outgrowth, cancer cell invasion and metastasis, and poor prognosis in various cancers. We investigated the role of ELMO1 in mediating the oncogenic behavior of gastric cancer (GC) cells. We also investigated the correlation between expression of ELMO1 in GC tissues and various clinicopathological parameters. METHODS We studied the impact of ELMO1 on tumor cell behavior using the pcDNA-myc vector and small interfering RNA in AGS and SNU1750 GC cell lines. We performed western blotting and immunohistochemistry to investigate the expression of ELMO1 in GC cells and tissues. RESULTS ELMO1 overexpression inhibited apoptosis via the modulation of PARP, caspase-3 and caspase-7 in GC cells. ELMO1 overexpression led to significant increase in the number of migrating and invading GC cells. The expression of E-cadherin decreased and that of Snail increased in GC cells upon ELMO1 overexpression. Phosphorylation of PI3K/Akt and GSK-3β was increased and that of β-catenin was decreased upon ELMO1 overexpression in GC cells. These results were reversed after ELMO1 knockdown. ELMO1 expression was significantly associated with tumor size, cancer stage, lymph node metastasis and survival. ELMO1-positive tumors had significantly higher mean of Ki-67 labeling index than ELMO1-negative tumors. There was no significant relationship between ELMO1 expression and the mean value of the apoptotic index. CONCLUSIONS Our results indicate that ELMO1 promotes tumor progression by modulating tumor cell survival in human GC.
Collapse
Affiliation(s)
- Young-Lan Park
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Jung-Ho Choi
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Sun-Young Park
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Hyung-Hoon Oh
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Dong-Ho Kim
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Yoon-Jin Seo
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Jae-Kyoung So
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Kaeun Song
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Min-Seok Cho
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Min-Woo Chung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Ji-Yun Hong
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Ki-Hyun Kim
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Eun Myung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Sung-Bum Cho
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Daeho Park
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and TechnologyGwangju, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| |
Collapse
|
253
|
Cui X, Zhang H, Chen T, Yu W, Shen K. Long Noncoding RNA SNHG22 Induces Cell Migration, Invasion, and Angiogenesis of Gastric Cancer Cells via microRNA-361-3p/HMGA1/Wnt/β-Catenin Axis. Cancer Manag Res 2020; 12:12867-12883. [PMID: 33364835 PMCID: PMC7751299 DOI: 10.2147/cmar.s281578] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background The correlation between long non-coding RNAs (lncRNAs) and gastric cancer (GC) has been indicated. As a newly found lncRNA, small nucleolar RNA host gene 22 (SNHG22) functions as an oncogene in ovarian carcinoma and breast cancer. However, its action has not been explored in GC. Herein, the purpose of the current research was to examine the influence of SNHG22 on GC development. Methods RT-qPCR was used to identify SNHG22 and microRNA-361-3p (miR-361-3p) in GC tissues and cells. Functional assays were implemented to measure changes on biological activities of GC cells under different transfections. Besides, after human umbilical vein endothelial cells (HUVECs) were co-cultured with supernatant of transfected GC cells, angiogenesis was assessed by tube formation assay in vitro. HMGA1 and β-catenin expression were determined. Finally, mechanistic assays, including RNA pull-down assay and dual-luciferase reporter assay, were employed to assess relationships among SNHG22, miR-361-3p, and HMGA1. Results SNHG22 and HMGA1 were highly expressed but miR-361-3p was poorly expressed in GC tissues. Mechanistically, SNHG22 bound to miR-361-3p, and miR-361-3p targeted HMGA1 to disrupt the Wnt/β-catenin pathway. Following SNHG22 or HMGA1 silencing or miR-361-3p upregulation, we observed a decline of proliferation, migration, and invasion of GC cells and HUVEC angiogenesis but acceleration of GC cell apoptosis and cell cycle arrest. Conclusion Collectively, SNHG22 silencing possessed tumor-suppressing potentials in GC development via Wnt/β-catenin pathway by binding to miR-361-3p and downregulating HMGA1, highlighting a new promising road for GC treatment development.
Collapse
Affiliation(s)
- Xiaofeng Cui
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Huaiyu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Tong Chen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Wei Yu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Kexin Shen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| |
Collapse
|
254
|
Ando K, Ozonoff A, Lee SY, Voisine M, Parker JT, Nakanishi R, Nishimura S, Yang J, Grace Z, Tran B, Diefenbach TJ, Maehara Y, Yasui H, Irino T, Salgia R, Terashima M, Gibbs P, Ramanathan RK, Oki E, Mori M, Kulke M, Hartshorn K, Bharti A. Multicohort Retrospective Validation of a Predictive Biomarker for Topoisomerase I Inhibitors. Clin Colorectal Cancer 2020; 20:e129-e138. [PMID: 33731288 DOI: 10.1016/j.clcc.2020.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/24/2020] [Accepted: 11/29/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE The camptothecin (CPT) analogs topotecan and irinotecan specifically target topoisomerase I (topoI) and are used to treat colorectal, gastric, and pancreatic cancer. Response rate for this class of drug varies from 10% to 30%, and there is no predictive biomarker for patient stratification by response. On the basis of our understanding of CPT drug resistance mechanisms, we developed an immunohistochemistry-based predictive test, P-topoI-Dx, to stratify the patient population into those who did and did not experience a response. PATIENTS AND METHODS The retrospective validation studies included a training set (n = 79) and a validation cohort (n = 27) of gastric cancer (GC) patients, and 8 cohorts of colorectal cancer (CRC) patient tissue (n = 176). Progression-free survival for 6 months was considered a positive response to CPT-based therapy. Formalin-fixed, paraffin-embedded slides were immunohistochemically stained with anti-phospho-specific topoI-Serine10 (topoI-pS10), quantitated, and analyzed statistically. RESULTS We determined a threshold of 35% positive staining to offer optimal test characteristics in GC. The GC (n = 79) training set demonstrated 76.6% (95% confidence interval, 64-86) sensitivity; 68.8% (41-88) specificity; positive predictive value (PPV) 92.5% (81-98); and negative predictive value (NPV) 42.3% (24-62). The GC validation set (n = 27) demonstrated 82.4% (56-95) sensitivity and 70.0% (35-92) specificity. Estimated PPV and NPV were 82.4% (56-95) and 70.0% (35-92) respectively. In the CRC validation set (n = 176), the 40% threshold demonstrated 87.5% (78-94) sensitivity; 70.0% (59-79) specificity; PPV 70.7% (61-79); and NPV 87.0 % (77-93). CONCLUSION The analysis of retrospective data from patients (n = 282) provides clinical validity to our P-topoI-Dx immunohistochemical test to identify patients with disease that is most likely to respond to topoI inhibitors.
Collapse
Affiliation(s)
- Koji Ando
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Al Ozonoff
- Division of Infectious Diseases, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Shin-Yin Lee
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Michael Voisine
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Julian-Taylor Parker
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sho Nishimura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jing Yang
- Department of Pathology, Boston University School of Medicine, Boston, MA
| | - Zhao Grace
- Department of Pathology, Boston University School of Medicine, Boston, MA
| | - Ben Tran
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | | | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Yasui
- Division of Gastric Surgery and Division of Gastrointestinal Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Tomoyuki Irino
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, City of Hope, Duarte, CA
| | - Masanori Terashima
- Division of Gastric Surgery and Division of Gastrointestinal Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Peter Gibbs
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | | | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Matthew Kulke
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Kevan Hartshorn
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Ajit Bharti
- Division of Hematology Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA.
| |
Collapse
|
255
|
Farah A, Kabbage M, Atafi S, Gabteni AJ, Barbirou M, Madhioub M, Hamzaoui L, Mohamed MA, Touinsi H, Kchaou AO, Chelbi E, Boubaker S, Abderrazek RB, Bouhaouala-Zahar B. Selective expression of KCNA5 and KCNB1 genes in gastric and colorectal carcinoma. BMC Cancer 2020; 20:1179. [PMID: 33267786 PMCID: PMC7709444 DOI: 10.1186/s12885-020-07647-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/17/2020] [Indexed: 01/04/2023] Open
Abstract
Background Gastric and colorectal cancers are the most common malignant tumours, leading to a significant number of cancer-related deaths worldwide. Recently, increasing evidence has demonstrated that cancer cells exhibit a differential expression of potassium channels and this can contribute to cancer progression. However, their expression and localisation at the somatic level remains uncertain. In this study, we have investigated the expression levels of KCNB1 and KCNA5 genes encoding ubiquitous Kv2.1 and Kv1.5 potassium channels in gastric and colorectal tumours. Methods Gastric and colorectal tumoral and peritumoral tissues were collected to evaluate the expression of KCNB1 and KCNA5 mRNA by quantitative PCR. Moreover, the immunohistochemical staining profile of Kv2.1 and Kv1.5 was assessed on 40 Formalin-Fixed and Paraffin-Embedded (FFPE) gastric carcinoma tissues. Differences in gene expression between tumoral and peritumoral tissues were compared statistically with the Mann-Whitney U test. The association between the clinicopathological features of the GC patients and the expression of both Kv proteins was investigated with χ2 and Fisher’s exact tests. Results The mRNA fold expression of KCNB1 and KCNA5 genes showed a lower mean in the tumoral tissues (0.06 ± 0.17, 0.006 ± 0.009) compared to peritumoral tissues (0.08 ± 0.16, 0.16 ± 0.48, respectively) without reaching the significance rate (p = 0.861, p = 0.152, respectively). Interestingly, Kv2.1 and Kv1.5 immunostaining was detectable and characterised by a large distribution in peritumoral and tumoral epithelial cells. More interestingly, inflammatory cells were also stained. Surprisingly, Kv2.1 and Kv1.5 staining was undoubtedly and predominantly detected in the cytoplasm compartment of tumour cells. Indeed, the expression of Kv2.1 in tumour cells revealed a significant association with the early gastric cancer clinical stage (p = 0.026). Conclusion The data highlight, for the first time, the potential role of Kv1.5 and Kv2.1 in gastrointestinal-related cancers and suggests they may be promising prognostic markers for these tumours.
Collapse
Affiliation(s)
- Azer Farah
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia
| | - Maria Kabbage
- Biomedical Genomics and Oncogenetics Laboratory, LR11IPT05 Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Salsabil Atafi
- Laboratory of Human and Experimental Pathology, Institute Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Amira Jaballah Gabteni
- Biomedical Genomics and Oncogenetics Laboratory, LR11IPT05 Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Mouadh Barbirou
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia.,Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mouna Madhioub
- Gastroenterology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | - Lamine Hamzaoui
- Gastroenterology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | | | - Hassen Touinsi
- Surgical Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | | | - Emna Chelbi
- Pathology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | - Samir Boubaker
- Laboratory of Human and Experimental Pathology, Institute Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rahma Ben Abderrazek
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia.
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia. .,Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
256
|
Jiang L, Hu LG. Serpin peptidase inhibitor clade A member 1-overexpression in gastric cancer promotes tumor progression in vitro and is associated with poor prognosis. Oncol Lett 2020; 20:278. [PMID: 33014156 PMCID: PMC7520747 DOI: 10.3892/ol.2020.12141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is the second most common cause of cancer-associated death in Asia. The incidence and mortality rates of gastric cancer have markedly increased in the past few decades. Therefore, the identification of novel gastric cancer biomarkers are needed to determine prognosis. The role of serpin peptidase inhibitor clade A member 1 (SERPINA1) has been studied in several types of cancer; however, little is known about its mechanism in gastric cancer. The present study aimed to evaluate SERPINA1 as a potential prognostic biomarker in gastric cancer and to identify the possible mechanisms underlying its action. The expression levels of SERPINA1 in several gastric cancer datasets were assessed, and it was identified that high expression of SERPINA1 was associated to poor clinical outcomes. Furthermore, using histochemical analysis, western blotting, apoptotic analysis, gap closure and invasion assays in cell lines, it was reported that silencing of SERPINA1 inhibited the formation of cellular pseudopodia and did not affect apoptosis, but promoted cell cycle S-phase entry. In addition, overexpression of SERPINA1 increased the migration and invasion of gastric cancer cells, whereas knockdown of SERPINA1 decreased these functions. Moreover, SERPINA1 overexpression increased the protein levels of SMAD4, which is a key regulator of the transforming growth factor (TGF)-β signaling pathway. Taken together, the present data demonstrated that SERPINA1 promotes gastric cancer progression through TGF-β signaling, and suggested that SERPINA1 may be a novel prognostic biomarker from tumor tissue biopsy in gastric cancer.
Collapse
Affiliation(s)
- Longchang Jiang
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Asia Research and Development Center, Shanghai 201210, P.R. China
| | - Liangbiao George Hu
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Asia Research and Development Center, Shanghai 201210, P.R. China
| |
Collapse
|
257
|
Karbalaei M, Keikha M. Potential association between the hopQ alleles of Helicobacter pylori and gastrointestinal diseases: A systematic review and meta-analysis. Meta Gene 2020; 26:100816. [DOI: 10.1016/j.mgene.2020.100816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
258
|
Nelson VK, Sahoo NK, Sahu M, Sudhan HH, Pullaiah CP, Muralikrishna KS. In vitro anticancer activity of Eclipta alba whole plant extract on colon cancer cell HCT-116. BMC Complement Med Ther 2020; 20:355. [PMID: 33225921 PMCID: PMC7681951 DOI: 10.1186/s12906-020-03118-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUNDS Colon cancer is the third most deadly and one of the most diagnosed diseases in the world. Although routine screening and early detection during last decades has improved the survival, colon cancer still claims hundreds of thousands lives each year worldwide. Surgery and chemotherapy is mainstay of current treatment, nevertheless toxicity associated with this treatment underscores the urgency of demand of a better therapeutics. Close to 50% of current chemotherapeutic drugs are direct or indirect descendants compounds isolated from medicinal plants, which indicate plants are great potential sources of novel therapeutics. In our literature review we found Eclipta alba to posses many pharmacological activities, including those with anticancer potentials. However, no study on anticancer activity of this kind has been reported. METHODS Phytochemicals were extracted by maceration method from shade dried whole plant of Eclipta alba using methanol as a solvent. The anticancer effect of extract was investigated on various cancer cell lines like human colorectal carcinoma (HCT-116), human prostate cancer (PC-3), Michigan cancer foundation-breast cancer (MCF-7) and renal cell carcinoma (RCC-45). We have also studied the effects on normal human embryonic lung fibroblast cell (WI-38) using MTT (methyl thiazoldiphenyltetrazolium bromide) assay, clonogenic (colony formation) and migration assay. Finally obtained results were analyzed using ANNOVA and Dunnett's test. RESULTS Results obtained from MTT assay revealed that the methanolic extract of Eclipta alba carried significant (p < 0.005) specificity against HCT-116 cells as compared to the other cancer cells. This extract also showed minimal or nontoxicity to WI-38 cells. Migration as well as clonogenic assays also confirmed the anticancer potential of the extract against HCT-116 cells. CONCLUSION This is a unique finding of its kind because the specific anticancer effect with minimal toxicity on normal cells has not been reported on Eclipta alba extract. Finally this finding opens up a great possibility to develop a novel antitumor drug candidate against deadly colon cancer in the future.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education & Research (Autonomous), Anantapuramu, Andhra Pradesh, 515721, India.
| | - Nalini Kanta Sahoo
- Marri Laxman Reddy Institute of Pharmacy, Medchal, Dundigal, Telangana, 500043, India
| | - Madhusmita Sahu
- Marri Laxman Reddy Institute of Pharmacy, Medchal, Dundigal, Telangana, 500043, India
| | - Hari Hara Sudhan
- Raghavendra Institute of Pharmaceutical Education & Research (Autonomous), Anantapuramu, Andhra Pradesh, 515721, India
| | - Chitikela P Pullaiah
- Department of Pharmacology, Siddha Central Research Institute, Chennai, Tamil Nadu, 60016, India
| | | |
Collapse
|
259
|
El Aamri M, Yammouri G, Mohammadi H, Amine A, Korri-Youssoufi H. Electrochemical Biosensors for Detection of MicroRNA as a Cancer Biomarker: Pros and Cons. BIOSENSORS 2020; 10:E186. [PMID: 33233700 PMCID: PMC7699780 DOI: 10.3390/bios10110186] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/23/2022]
Abstract
Cancer is the second most fatal disease in the world and an early diagnosis is important for a successful treatment. Thus, it is necessary to develop fast, sensitive, simple, and inexpensive analytical tools for cancer biomarker detection. MicroRNA (miRNA) is an RNA cancer biomarker where the expression level in body fluid is strongly correlated to cancer. Various biosensors involving the detection of miRNA for cancer diagnosis were developed. The present review offers a comprehensive overview of the recent developments in electrochemical biosensor for miRNA cancer marker detection from 2015 to 2020. The review focuses on the approaches to direct miRNA detection based on the electrochemical signal. It includes a RedOx-labeled probe with different designs, RedOx DNA-intercalating agents, various kinds of RedOx catalysts used to produce a signal response, and finally a free RedOx indicator. Furthermore, the advantages and drawbacks of these approaches are highlighted.
Collapse
Affiliation(s)
- Maliana El Aamri
- Laboratory of Process Engineering & Environment, Faculty of Sciences and Techniques, Hassan II, University of Casablanca, B.P.146, Mohammedia 28806, Morocco; (M.E.A.); (G.Y.); (H.M.)
| | - Ghita Yammouri
- Laboratory of Process Engineering & Environment, Faculty of Sciences and Techniques, Hassan II, University of Casablanca, B.P.146, Mohammedia 28806, Morocco; (M.E.A.); (G.Y.); (H.M.)
| | - Hasna Mohammadi
- Laboratory of Process Engineering & Environment, Faculty of Sciences and Techniques, Hassan II, University of Casablanca, B.P.146, Mohammedia 28806, Morocco; (M.E.A.); (G.Y.); (H.M.)
| | - Aziz Amine
- Laboratory of Process Engineering & Environment, Faculty of Sciences and Techniques, Hassan II, University of Casablanca, B.P.146, Mohammedia 28806, Morocco; (M.E.A.); (G.Y.); (H.M.)
| | - Hafsa Korri-Youssoufi
- Université Paris-Saclay, CNRS, Institut de Chimie Moléculaire et des Matériaux d’Orsay (ICMMO), Equipe de Chimie Biorganique et Bioinorganique (ECBB), Bât 420, 2 Rue du Doyen Georges Poitou, 91400 Orsay, France;
| |
Collapse
|
260
|
Xu Z, Ran J, Gong K, Hou Y, Li J, Guo Y. LncRNA SUMO1P3 regulates the invasion, migration and cell cycle of gastric cancer cells through Wnt/β-catenin signaling pathway. J Recept Signal Transduct Res 2020; 41:574-581. [PMID: 33179980 DOI: 10.1080/10799893.2020.1836494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zhong Xu
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jing Ran
- Department of Gynaecology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Kai Gong
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yihan Hou
- Department of ICU, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ji Li
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yijuan Guo
- Department of Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
261
|
Luo T, Du Y, Duan J, Liang C, Chen G, Jiang K, Chen Y, Chen Y. Development and Validation of a Scoring System Based on 9 Glycolysis-Related Genes for Prognosis Prediction in Gastric Cancer. Technol Cancer Res Treat 2020; 19:1533033820971670. [PMID: 33161837 PMCID: PMC7658532 DOI: 10.1177/1533033820971670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gastric cancer is a malignant tumor with high morbidity and mortality worldwide. However, increasing evidences have revealed the correlation between the glycolysis process and tumorigenesis. This study is aim to develop a list of glycolysis-related genes for risk stratification in gastric cancer patients. We included 500 patients’ sample data from GSE62254 and GSE26942 datasets, and classified patients into training (n = 350) and testing sets (n = 150) at a ratio of 7: 3. Univariate and multivariate Cox regression analysis were performed to screen genes having prognostic value. Based on HALLMARK gene sets, we identified 9 glycolysis-related genes (BPNT1, DCN, FUT8, GMPPA, GPC3, LDHC, ME2, PLOD2, and UGP2). On the basis of risk score developed by the 9 genes, patients were classified into high- and low-risk groups. The survival analysis showed that the high-risk patients had a worse prognosis (p < 0.001). Similar finding was observed in the testing cohort and 2 independent cohorts (GSE13861 and TCGA-STAD, all p < 0.001). The multivariate Cox regression analysis showed that the risk score was an independent prognostic factor for overall survival (p < 0.001). Furthermore, we constructed a nomogram that integrated the risk score and tumor stage, age, and adjuvant chemotherapy. Through comparing the results of the receiver operating characteristic curves and decision curve analysis, we found that the nomogram had a superior predictive accuracy than conventional TNM staging system, suggesting that the risk score combined with other clinical factors (age, tumor stage, and adjuvant chemotherapy) can develop a robust prediction for survival and improve the individualized clinical decision making of the patient. In conclusion, we identified 9 glycolysis-related genes from hallmark glycolysis pathway. Based on the 9 genes, gastric cancer patients were separated into different risk groups related to survival.
Collapse
Affiliation(s)
- Tianqi Luo
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yufei Du
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jinling Duan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Experimental Research (Cancer Institute), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chengcai Liang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guoming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kaiming Jiang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Yingbo Chen, and Yongming Chen, Dongfeng East Road 651, Guangzhou, Guangdong 510060, China. Emails: ;
| | - Yingbo Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Yingbo Chen, and Yongming Chen, Dongfeng East Road 651, Guangzhou, Guangdong 510060, China. Emails: ;
| |
Collapse
|
262
|
Bian J, Long JY, Yang X, Yang XB, Xu YY, Lu X, Sang XT, Zhao HT. Signature based on molecular subtypes of deoxyribonucleic acid methylation predicts overall survival in gastric cancer. World J Gastroenterol 2020; 26:6414-6430. [PMID: 33244202 PMCID: PMC7656213 DOI: 10.3748/wjg.v26.i41.6414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/17/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks as the third leading cause of cancer-related death worldwide. Epigenetic alterations contribute to tumor heterogeneity in early stages.
AIM To identify the specific deoxyribonucleic acid (DNA) methylation sites that influence the prognosis of GC patients and explore the prognostic value of a model based on subtypes of DNA methylation.
METHODS Patients were randomly classified into training and test sets. Prognostic DNA methylation sites were identified by integrating DNA methylation profiles and clinical data from The Cancer Genome Atlas GC cohort. In the training set, unsupervised consensus clustering was performed to identify distinct subgroups based on methylation status. A risk score model was built based on Kaplan-Meier, least absolute shrinkage and selector operation, and multivariate Cox regression analyses. A test set was used to validate this model.
RESULTS Three subgroups based on DNA methylation profiles in the training set were identified using 1061 methylation sites that were significantly associated with survival. These methylation subtypes reflected differences in T, N, and M category, age, stage, and prognosis. Forty-one methylation sites were screened as specific hyper- or hypomethylation sites for each specific subgroup. Enrichment analysis revealed that they were mainly involved in pathways related to carcinogenesis, tumor growth, and progression. Finally, two methylation sites were chosen to generate a prognostic model. The high-risk group showed a markedly poor prognosis compared to the low-risk group in both the training [hazard ratio (HR) = 2.24, 95% confidence interval (CI): 1.28-3.92, P < 0.001] and test (HR = 2.12, 95%CI: 1.19-3.78, P = 0.002) datasets.
CONCLUSION DNA methylation-based classification reflects the epigenetic heterogeneity of GC and may contribute to predicting prognosis and offer novel insights for individualized treatment of patients with GC.
Collapse
Affiliation(s)
- Jin Bian
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun-Yu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao-Bo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yi-Yao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xin-Ting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
263
|
Development and validation of metabolism-related gene signature in prognostic prediction of gastric cancer. Comput Struct Biotechnol J 2020; 18:3217-3229. [PMID: 33209209 PMCID: PMC7649605 DOI: 10.1016/j.csbj.2020.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the most common malignant tumours in the world. As one of the crucial hallmarks of cancer reprogramming of metabolism and the relevant researches have a promising application in the diagnosis treatment and prognostic prediction of malignant tumours. This study aims to identify a group of metabolism-related genes to construct a prediction model for the prognosis of gastric cancer. A large cohort of gastric cancer cases (1121 cases) from public database was included in our analysis and classified patients into training and testing cohorts at a ratio of 7: 3. After identifying a list of metabolism-related genes having prognostic value, we constructed a risk score based on metabolism-related genes using LASSO-COX method. According to the risk score, patients were divided into high- and low-risk groups. Our results revealed that high-risk patients had a significantly worse prognosis than low-risk patients in both the training (high-risk vs low-risk patients; five years overall survival: 37.2% vs 72.2%; p < 0.001) and testing cohorts (high-risk vs low-risk patients; five years overall survival: 42.9% vs 62.9%; p < 0.001). This observation was validated in the external validation cohort (high-risk vs. low-risk patients; five years overall survival: 30.2% vs 40.4%; p = 0.007). To reinforce the predictive ability of the model, we integrated risk score, age, adjuvant chemotherapy, and TNM stage into a nomogram. According to the result of receiver operating characteristic curves and decision curves analysis, we found that the nomogram score had a superior predictive ability than conventional factors, indicating that the risk score combined with clinicopathological features can develop a robust prediction for survival and improve the individualized clinical decision making of the patient. In conclusion, we identified a list of metabolic genes related to survival and developed a metabolism-based predictive model for gastric cancer. Through a series of bioinformatics and statistical analyses, the predictive ability of the model was confirmed.
Collapse
|
264
|
Rahimi AM, Nabavizadeh F, Ashabi G, Halimi S, Rahimpour M, Vahedian J, Panahi M. Probiotic Lactobacillus rhamnosus Supplementation Improved Capecitabine Protective Effect against Gastric Cancer Growth in Male BALB/c Mice. Nutr Cancer 2020; 73:2089-2099. [PMID: 33955797 DOI: 10.1080/01635581.2020.1832237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The gastric cancer (GC) is biologically and genetically heterogeneous with a poorly understood carcinogenesis at the molecular level. Herein, we studied the effects of probiotics (Lactobacillus rhamnosus) on subcutaneous implantation of xenograft GC. Moreover, the effect of probiotics (L. rhamnosus) was compared with the capecitabine drug as known used drug against GC. Human GC tissue was obtained from patients with gastric adenocarcinoma and grafted into mice armpit. Probiotic (L. rhamnosus) was given to animals by gavage 2 weeks prior to GC and 4 weeks after GC induction. Also, capecitabine was orally added through feeding tube at the last week of treatment procedure. All grafted animals received cyclosporine a day before the surgery and during the study period to prevent graft rejection. Capecitabine-probiotic complex reduced the size of the axillary implanted GC when compared with control group. Furthermore, combination of capecitabine and probiotic increased apoptotic and necrotic responses in the grafted tumor, blood cells (red blood cells, white blood cells, and platelet counts) in comparison with capecitabine. Probiotic (L. rhamnosus) administration effectively improved the therapeutic index and outcomes, and also, improved the therapeutic effects of the capecitabine.
Collapse
Affiliation(s)
- Ahmad Mustafa Rahimi
- Department of Physiology, Medical school, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Department of Physiology, Medical school, Tehran University of Medical Sciences, Tehran, Iran.,Electrophysiology Research Center, Neurosciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, Medical school, Tehran University of Medical Sciences, Tehran, Iran.,Electrophysiology Research Center, Neurosciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahnaz Halimi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Rahimpour
- Department of Physiology, Medical school, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalal Vahedian
- Department of Surgery, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Panahi
- Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
265
|
Yamamoto H, Watanabe Y, Sato Y, Maehata T, Itoh F. Non-Invasive Early Molecular Detection of Gastric Cancers. Cancers (Basel) 2020; 12:E2880. [PMID: 33036473 PMCID: PMC7600616 DOI: 10.3390/cancers12102880] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is a significant source of global cancer death with a high mortality rate, because the majority of patients with GC are diagnosed at a late stage, with limited therapeutic choices and poor outcomes. Therefore, development of minimally invasive or noninvasive biomarkers which are specific to GC is crucially needed. The latest advancements in the understanding of GC molecular landscapes and molecular biological methods have accelerated attempts to diagnose GC at an early stage. Body fluids, including peripheral blood, saliva, gastric juice/wash, urine, and others, can be a source of biomarkers, offering new methods for the early detection of GC. Liquid biopsy-based methods using circulating sources of cancer nucleic acids could also be considered as alternative strategies. Moreover, investigating gastric juices/washes could represent an alternative for the detection of GC via invasive biopsy. This review summarizes recently reported biomarkers based on DNA methylation, microRNA, long noncoding RNA, circular RNA, or extracellular vesicles (exosomes) for the detection of GC. Although the majority of studies have been conducted to detect these alterations in advanced-stage GC and only a few in population studies or early-stage GC, some biomarkers are potentially valuable for the development of novel approaches for an early noninvasive detection of GC.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan; (Y.W.); (Y.S.); (T.M.); (F.I.)
| | - Yoshiyuki Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan; (Y.W.); (Y.S.); (T.M.); (F.I.)
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki 210-0806, Japan
| | - Yoshinori Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan; (Y.W.); (Y.S.); (T.M.); (F.I.)
| | - Tadateru Maehata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan; (Y.W.); (Y.S.); (T.M.); (F.I.)
| | - Fumio Itoh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan; (Y.W.); (Y.S.); (T.M.); (F.I.)
| |
Collapse
|
266
|
Namikawa T, Yokota K, Iwabu J, Munekage M, Uemura S, Tsujii S, Maeda H, Kitagawa H, Karashima T, Kumon M, Inoue K, Kobayashi M, Hanazaki K. Incidence and risk factors of osteoporotic status in outpatients who underwent gastrectomy for gastric cancer. JGH Open 2020; 4:903-908. [PMID: 33102762 PMCID: PMC7578282 DOI: 10.1002/jgh3.12347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/14/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Disorders in bone metabolism have long been recognized as typical sequelae of gastrectomy; however, the pathogenesis has not been fully elucidated, resulting in a variation of reported incidence. This study aimed to evaluate current bone health by measuring bone mineral density (BMD) in patients treated by gastrectomy for gastric cancer, with a focus on incidence and risk factors of osteoporosis. METHODS The study enrolled 81 patients who underwent gastrectomy for gastric cancer at Kochi Medical School. BMD of the lumbar spine was measured by dual-energy X-ray mineral absorptiometry, with the results expressed as a percentage of the young adult mean (YAM). Clinical data were also obtained to investigate associations with BMD. RESULTS Of the 81 study patients, 12 (14.8%) were deemed to have osteoporosis, defined by a percentage of YAM <70, with a dominance of females over males (66.7% vs 17.4%; P < 0.001). The median body weight, hemoglobin concentration, and serum alkaline phosphatase (ALP) level of the patients with osteoporosis was significantly lower than in those with a percentage of YAM ≥70 group (39.6 kg vs 53.1 kg, P < 0.001; 10.9 mg/dL vs 12.5 mg/dL, P = 0.010; 210 U/L vs 251 U/L, P = 0.002). Further analyses revealed a significant positive correlation between body weight and percentage of YAM (r = 0.441, P < 0.001). Despite the administration of bisphosphonates in these patients during this study, one acquired a bone fracture. CONCLUSION Osteoporosis was found in 14.8% of postoperative gastric cancer patients, with female gender, low body weight, and low ALP proposed as risk factors for osteoporosis and thus future bone fracture.
Collapse
Affiliation(s)
| | | | - Jun Iwabu
- Department of SurgeryKochi Medical SchoolNankokuJapan
| | | | - Sunao Uemura
- Department of SurgeryKochi Medical SchoolNankokuJapan
| | | | | | | | | | | | - Keiji Inoue
- Department of UrologyKochi Medical SchoolNankokuJapan
| | - Michiya Kobayashi
- Department of Human Health and Medical SciencesKochi Medical SchoolNankokuJapan
| | | |
Collapse
|
267
|
Shahidi S, Hejazi J, Moghimi M, Borji S, Zabihian S, Fathi M. Circulating Irisin Levels and Redox Status Markers in Patients with Gastric Cancer: A Case-Control Study. Asian Pac J Cancer Prev 2020; 21:2847-2851. [PMID: 33112539 PMCID: PMC7798161 DOI: 10.31557/apjcp.2020.21.10.2847] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Irisin, mostly known as an exercise-induced fat browning myokine, has been recently detected in several cancer cells, and its potential for being utilized as a biomarker for early diagnosis of some cancers, such as Gastric cancer (GC), is the subject of speculation. The present study aims to compare serum irisin levels in GC patients and healthy controls and assess the interrelation between irisin and oxidative stress markers. METHODS In this case-control study, 22 newly diagnosed GC patients and 29 healthy controls were recruited based on the inclusion criteria. Serum levels of irisin were quantified in duplicates by ELISA. Oxidative stress indices, including total antioxidant power in sera, thiol group, malondialdehyde, and superoxide dismutase concentrations, were also measured in both groups. An independent-sample t-test was used to compare the means between the two studied groups. RESULTS Serum levels of irisin were significantly higher in the GC group compared with those of their healthy counterparts (p =0.032). No significant differences were observed between the two groups in terms of the serum total antioxidant power or the oxidative stress marker, including MDA, thiol groups, and SOD concentration in sera. Furthermore, there was no significant association between irisin, FRAP, the Thiol group, and the SOD activity. CONCLUSION According to the finding, the increased serum levels of irisin in GC patients can play a potential role in the early diagnosis of the GC patients; hence, this peptide can be employed as a new diagnostic indicator of GC. .
Collapse
Affiliation(s)
- Shabnam Shahidi
- Student Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Jalal Hejazi
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Minoosh Moghimi
- Department of Hemato Oncology, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Soheila Borji
- Department of Radiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Saeed Zabihian
- Ayatollah Mousavi Clinical and Educational Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mojtaba Fathi
- Department of Biochemistry. School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
268
|
Azangou-Khyavy M, Ghasemi M, Khanali J, Boroomand-Saboor M, Jamalkhah M, Soleimani M, Kiani J. CRISPR/Cas: From Tumor Gene Editing to T Cell-Based Immunotherapy of Cancer. Front Immunol 2020; 11:2062. [PMID: 33117331 PMCID: PMC7553049 DOI: 10.3389/fimmu.2020.02062] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/29/2020] [Indexed: 12/26/2022] Open
Abstract
The clustered regularly interspaced short palindromic repeats system has demonstrated considerable advantages over other nuclease-based genome editing tools due to its high accuracy, efficiency, and strong specificity. Given that cancer is caused by an excessive accumulation of mutations that lead to the activation of oncogenes and inactivation of tumor suppressor genes, the CRISPR/Cas9 system is a therapy of choice for tumor genome editing and treatment. In defining its superior use, we have reviewed the novel applications of the CRISPR genome editing tool in discovering, sorting, and prioritizing targets for subsequent interventions, and passing different hurdles of cancer treatment such as epigenetic alterations and drug resistance. Moreover, we have reviewed the breakthroughs precipitated by the CRISPR system in the field of cancer immunotherapy, such as identification of immune system-tumor interplay, production of universal Chimeric Antigen Receptor T cells, inhibition of immune checkpoint inhibitors, and Oncolytic Virotherapy. The existing challenges and limitations, as well as the prospects of CRISPR based systems, are also discussed.
Collapse
Affiliation(s)
| | - Mobina Ghasemi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Khanali
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Monire Jamalkhah
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Masoud Soleimani
- Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
269
|
Yue L, Lu Y, Li Y, Wang Y. Prognostic Value of C-Reactive Protein to Albumin Ratio in Gastric Cancer: A Meta-Analysis. Nutr Cancer 2020; 73:1864-1871. [PMID: 32940549 DOI: 10.1080/01635581.2020.1817510] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE C-reactive protein to albumin ratio (CRP/Alb) is investigated as a prognostic marker in gastric cancer in previous studies, with presence of inconsistent data. Therefore, this study aimed to explore the prognostic role of CRP/Alb in gastric cancer through meta-analysis. METHODS This meta-analysis systemically retrieved PubMed, Embase, Web of Science, the Chinese National Knowledge Infrastructure (CNKI), and Wanfang up to July 4, 2020. Pooled hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were conducted to evaluate the association between CRP/Alb and survival outcomes. RESULTS A total of nine studies with 3346 patients were included in the present meta-analysis. The pooled HR and 95%CI were: HR = 1.89, 95%CI = 1.64-2.19, p < 0.001 for overall survival (OS) and HR = 2.15, 95%CI = 1.72-2.70, p < 0.001 for disease-free survival (DFS). Subgroup analysis demonstrate that an elevated CRP/Alb remain a significant prognostic factor for poor OS and DFS irrespective of sample size, nationality of patients, or cutoff value resource (p < 0.05 in all subgroups). CONCLUSIONS The present meta-analysis suggests that high CRP/Alb is predictive of poor OS and DFS in gastric cancer. CRP/Alb is therefore a potential prognostic factor in the management of patients with gastric cancer.
Collapse
Affiliation(s)
- Liang Yue
- Faculty of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Lu
- Faculty of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulin Li
- Faculty of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yilin Wang
- Department of Thoracic Surgery, Chongqing University Three Gorges Hospital & Chongqing Three Gorges Central Hospital, Chongqing, China
| |
Collapse
|
270
|
Asgharipour H, Nourian M, Iravani S, Saidi S, Mehrvar N, Chaleshi V, Zamani N, Etemadi A, Sadeghi Y, Naghoosi H. Expression analysis of FOXC1 & FOXCUT genes in patients with gastric cancer. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
271
|
Zeng X, Wang HY, Wang YP, Bai SY, Pu K, Zheng Y, Guo QH, Guan QL, Ji R, Zhou YN. COL4A family: potential prognostic biomarkers and therapeutic targets for gastric cancer. Transl Cancer Res 2020; 9:5218-5232. [PMID: 35117889 PMCID: PMC8799138 DOI: 10.21037/tcr-20-517] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Background The type IV collagen alpha chain (COL4A) family is a major component of the basement membrane (BM) that has recently been found to be involved in tumor angiogenesis and progression. However, the expression levels and the exact roles of distinct COL4A family members in gastric cancer (GC) have not been completely understood. Methods Here, the expression levels of COL4As in GC and normal gastric tissues were calculated by using TCGA datasets and the predicted prognostic values by the GEPIA tool. Furthermore, the cBioPortal and Metascape tools were integrated to analyze the genetic alterations, correlations and potential functions of COL4As, and their frequently altered neighboring genes in GC. Results Notably, the expression levels of COL4A1/2/4 in GC were higher to those in normal gastric tissues, while the expression levels of COL4A3/5/6 were lower in GC than normal. Survival analysis revealed that lower expression levels of COL4A1/5 led to higher overall survival (OS) rate. Multivariate analysis using the Cox proportional-hazards model indicated that age, gender, pathological grade, metastasis and COL4A5 expression, are independent prognostic factors for OS. However, TNM stage, lymph node metastasis, Lauren’s classification, COL4A1-4 and COL4A6 were associated with poor OS but not independent prognostic factors. Function-enriched analysis of COL4As and their frequently altered neighboring genes was involved in tumor proliferation and metastasis in GC. Conclusions These results implied that COL4A1/2 were potential therapeutic targets for GC. COL4A3/4/6 might have an impact on gastric carcinogenesis and subsequent progression, whereas COL4A5 was an independent prognostic marker for GC.
Collapse
Affiliation(s)
- Xi Zeng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Hao-Ying Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Yu-Ping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Su-Yang Bai
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Ke Pu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Qing-Hong Guo
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Quan-Lin Guan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Rui Ji
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| | - Yong-Ning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
272
|
Demographic Profile of Gastric Cancer in Afghanistan. Indian J Surg Oncol 2020; 11:344-347. [DOI: 10.1007/s13193-020-01133-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
|
273
|
Halajzadeh J, Dana PM, Asemi Z, Mansournia MA, Yousefi B. An insight into the roles of piRNAs and PIWI proteins in the diagnosis and pathogenesis of oral, esophageal, and gastric cancer. Pathol Res Pract 2020; 216:153112. [PMID: 32853949 DOI: 10.1016/j.prp.2020.153112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/28/2020] [Accepted: 07/07/2020] [Indexed: 12/31/2022]
Abstract
P-Element induced wimpy testis (PIWI)-interacting RNA (piRNA) is a member of the non-coding RNAs family. Four PIWI proteins are found to be expressed in humans. The number of studies focusing on the roles of piRNAs and PIWI proteins in the field of cancer is increasing. Oral, esophageal, and gastric cancers are considered as important causes of death. PIWI proteins and piRNAs are suggested to be involved in the pathogenesis of these diseases. Thus, studying these molecules may be beneficial for finding new therapeutics. Since it is shown that currently used biomarkers for these cancers have low sensitivity and specificity, there is a necessity for identifying novel non-invasive biomarkers which are highly sensitive and specific. This paper will provide an insight into current knowledge about the functions of PIWI proteins and piRNAs in the oral, esophageal, and gastric cancer. We discuss how PIWI proteins and piRNAs can be involved in the pathogenesis of these cancers. Moreover, we review the studies concerning with the roles of PIWI proteins and piRNAs as biomarkers which are used for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Jamal Halajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Science, Maragheh, Iran.
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran.
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bahman Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
274
|
Behar D, Boublenza L, Chabni N, Hassaine H, Dahmani B, Masdoua N, Nahet A, Meguenni K. Retrospective epidemiological study on stomach cancer in a region of western Algeria: about 394 cases between 2011 and 2015. J Gastrointest Cancer 2020; 52:706-710. [PMID: 32705578 DOI: 10.1007/s12029-020-00459-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Stomach cancer is a major global health problem; it is one of the ten most common cancers with poor survival, and its incidence is characterized by wide variation. The aim of this work is to carry out a retrospective epidemiological study on gastric cancer in the wilaya of Tlemcen (West Algeria) over a period of 5 years (2011-2015). METHODS The data set was provided by the cancer registry of the Tlemcen wilaya. The statistical analysis was performed using software SPSS.21. RESULTS During this period, 394 cases of gastric cancers were collected, including 199 patients with gastric adenocarcinoma (50.50%); gastric cancer was ranked in 5th rank of the ten most answered cancers in the wilaya. The average age of the patients was 60.5 ± 14.208 years (60.618 ± 13.556 men, 56.654 ± 14.8761 women) with an extremity ranging from 18 to 91 years. A predominance of men has been observed with 60.4% against 39.6% of women, with a sex ratio of 1.5 and a significant difference between the two sexes (p = 0.08). Patients over 50 years of age represented the predominant age group (73.4%). The tumor topography was antropyloric in 6.09% of the cases, and the vast majority of the diagnosed cases was in the local stage (17.01%). CONCLUSION Stomach cancer remains one of the top ten cancers in the Tlemcen willaya, so extensive research on the risk factors for gastric cancer remains important to plan effective preventive and curative strategies to reduce this burden.
Collapse
Affiliation(s)
- Dalale Behar
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria.
| | - Lamia Boublenza
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria
- Department of Medicine Cancer Laboratory, Abou-Bekr Belkaid University, Tlemcen, Algeria
| | - Nafissa Chabni
- Department of Medicine Cancer Laboratory, Abou-Bekr Belkaid University, Tlemcen, Algeria
- Department of Epidemiology, Dr Tidjani Damerdji University Hospital, Abou-Bekr Belkaid University, Tlemcen, Algeria
| | - Hafida Hassaine
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria
| | - Bouchra Dahmani
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria
| | - Nabila Masdoua
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria
| | - Amira Nahet
- Department of Biology, Laboratory of Microbiology Applied to the Food Industry, to Biomedical and to the Environment (LAMAABE), University Abou-Bekr Belkaid, Ex biomedical complex Imama Bloc C BP 119, 13000, Tlemcen, Algeria
| | - Kaouel Meguenni
- Department of Medicine Cancer Laboratory, Abou-Bekr Belkaid University, Tlemcen, Algeria
- Department of Epidemiology, Dr Tidjani Damerdji University Hospital, Abou-Bekr Belkaid University, Tlemcen, Algeria
| |
Collapse
|
275
|
Chen Y, Chen W, Dai X, Zhang C, Zhang Q, Lu J. Identification of the collagen family as prognostic biomarkers and immune-associated targets in gastric cancer. Int Immunopharmacol 2020; 87:106798. [PMID: 32693357 DOI: 10.1016/j.intimp.2020.106798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Gastric cancer has extremely high morbidity and mortality. Currently, it is lack of effective biomarkers and therapeutic targets for guiding clinical treatment. In this study, we aimed to identify novel biomarkers and therapeutic targets for gastric cancer. METHODS Differentially expressed genes (DEGs) between gastric cancer and normal tissues were obtained from Gene Expression Omnibus (GEO). Core genes were identified by constructing protein-protein interaction network of DEGs. The expression of core genes was verified in Gene Expression Profiling Interactive Analysis (GEPIA), UALCAN and clinical samples. Further, the mutation, DNA methylation, prognostic value, and immune infiltration of core genes were validated by cBioPortal, MethSurv, Kaplan-Meier plotter, and Tumor Immune Estimation Resource (TIMER) databases. Additionally, drug response analysis was performed by Cancer Therapy Response Portal (CTRP). RESULTS A total of seven collagen family members were identified as core genes among upregulated genes. And copy number amplification may be involved in the upregulation of COL1A1 and COL1A2. Importantly, the collagen family was associated with the poor prognosis of patients with metastasis. Among them, COL1A1 had a higher hazard ratio (HR) for overall survival than other members (HR = 2.33). The correlation between DNA methylation levels at CpG sites of collagen family members and the prognosis was verified in gastric cancer. Besides, collagen family expression was positively correlated with macrophages infiltration and the expression of M2 macrophages markers. Further, collagen expression was related to the sensitivity and resistance of gastric cancer cell lines to certain drugs. CONCLUSIONS The collagen family, especially COL1A1, COL1A2, and COL12A1, may act as potential prognostic biomarkers and immune-associated therapeutic targets in gastric cancer.
Collapse
Affiliation(s)
- Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Chengjuan Zhang
- Department of Pathology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province 450003, PR China
| | - Qiushuang Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China.
| |
Collapse
|
276
|
Liu XY, Zhang J. A protocol of systematic review and meta-analysis of narrow band imaging endoscopy in detection of early gastric cancer. Medicine (Baltimore) 2020; 99:e21269. [PMID: 32702914 PMCID: PMC7373608 DOI: 10.1097/md.0000000000021269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although previous studies have utilized narrow band imaging endoscopy diagnosis (NBIED) in detection of patients with early gastric cancer (EGC), there are still inconsistent results. Thus, this study will explore the accuracy of NBIED in detection of patients with EGC. METHODS We propose to perform literature search of potential studies investigating the accuracy of NBIED in detection of patients with EGC in MEDLINE, EMBASE, Cochrane Library, Web of Science, WANGFANG, VIP database and China National Knowledge Infrastructure from the beginning of each database to January 31, 2020 without restrictions to language and publication time. Two authors will independently scrutinize these databases to identify studies that satisfy all predefined eligibility criteria. We will check study quality and analyze outcome data using Quality Assessment of Diagnostic Accuracy Studies tool, and RevMan 5.3 software respectively. RESULTS We anticipate the results of this study will afford additional insight into the appraising of the accuracy of NBIED in patients with EGC. CONCLUSION The findings of this study will be useful informing diagnostic decisions for the diagnosis of patients with EGC.PROSPERO registration number: PROSPERO CRD42020171053.
Collapse
Affiliation(s)
- Xiao-yu Liu
- Department of Endoscopy Center, Yulin No.2 Hospital, Yulin
| | - Jun Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, China
| |
Collapse
|
277
|
Cai WY, Dong ZN, Fu XT, Lin LY, Wang L, Ye GD, Luo QC, Chen YC. Identification of a Tumor Microenvironment-relevant Gene set-based Prognostic Signature and Related Therapy Targets in Gastric Cancer. Am J Cancer Res 2020; 10:8633-8647. [PMID: 32754268 PMCID: PMC7392024 DOI: 10.7150/thno.47938] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022] Open
Abstract
Rationale: The prognosis of gastric cancer (GC) patients is poor, and there is limited therapeutic efficacy due to genetic heterogeneity and difficulty in early-stage screening. Here, we developed and validated an individualized gene set-based prognostic signature for gastric cancer (GPSGC) and further explored survival-related regulatory mechanisms as well as therapeutic targets in GC. Methods: By implementing machine learning, a prognostic model was established based on gastric cancer gene expression datasets from 1699 patients from five independent cohorts with reported full clinical annotations. Analysis of the tumor microenvironment, including stromal and immune subcomponents, cell types, panimmune gene sets, and immunomodulatory genes, was carried out in 834 GC patients from three independent cohorts to explore regulatory survival mechanisms and therapeutic targets related to the GPSGC. To prove the stability and reliability of the GPSGC model and therapeutic targets, multiplex fluorescent immunohistochemistry was conducted with tissue microarrays representing 186 GC patients. Based on multivariate Cox analysis, a nomogram that integrated the GPSGC and other clinical risk factors was constructed with two training cohorts and was verified by two validation cohorts. Results: Through machine learning, we obtained an optimal risk assessment model, the GPSGC, which showed higher accuracy in predicting survival than individual prognostic factors. The impact of the GPSGC score on poor survival of GC patients was probably correlated with the remodeling of stromal components in the tumor microenvironment. Specifically, TGFβ and angiogenesis-related gene sets were significantly associated with the GPSGC risk score and poor outcome. Immunomodulatory gene analysis combined with experimental verification further revealed that TGFβ1 and VEGFB may be developed as potential therapeutic targets of GC patients with poor prognosis according to the GPSGC. Furthermore, we developed a nomogram based on the GPSGC and other clinical variables to predict the 3-year and 5-year overall survival for GC patients, which showed improved prognostic accuracy than clinical characteristics only. Conclusion: As a tumor microenvironment-relevant gene set-based prognostic signature, the GPSGC model provides an effective approach to evaluate GC patient survival outcomes and may prolong overall survival by enabling the selection of individualized targeted therapy.
Collapse
|
278
|
Necula L, Matei L, Dragu D, Pitica I, Neagu AI, Bleotu C, Dima S, Popescu I, Diaconu CC, Chivu-Economescu M. High plasma levels of COL10A1 are associated with advanced tumor stage in gastric cancer patients. World J Gastroenterol 2020; 26:3024-3033. [PMID: 32587446 PMCID: PMC7304107 DOI: 10.3748/wjg.v26.i22.3024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) remains an aggressive malignancy with a high rate of mortality, being the third leading cause of cancer-related death. More than one million newly diagnosed cases and 782685 deaths due to GC were reported in 2018. GC is characterized by limited effective treatment options and the lack of consistent biomarkers for the diagnosis and prognosis of these patients. The discovery of new biomarkers useful in the early diagnosis of GC is mandatory. AIM To evaluate the potential of COL10A1 as a circulating biomarker for the diagnosis and prognosis of gastric adenocarcinoma patients. METHODS Plasma and tissue obtained from 49 patients with gastric adenocarcinoma have been used in exploring the expression of COL10A1. Real-time PCR and western blot techniques were used to evaluate COL10A1 level in gastric tumor tissue compared to normal adjacent tissue. The circulating level of COL10A1 was also evaluated by ELISA in plasma of gastric adenocarcinoma patients. Survival analysis was made in order to evaluate the potential of COL10A1 as a biomarker for the diagnosis and prognosis of gastric adenocarcinoma patients. RESULTS Our results showed a significant increase in COL10A1 gene expression and protein levels in gastric tumor tissue compared to adjacent normal tissue (P < 0.05). COL10A1 seems to show an elevated expression from the beginning of carcinogenesis, in the early stages, and its increased level remains elevated during cancer progression. A significant increase of COL10A1 plasma level in gastric adenocarcinoma patients was also identified. Moreover, increased COL10A1 plasma level was associated with poor survival of the patients. Plasma COL10A1 performed a diagnostic value in GC with area under the receiver operating characteristic curve (AUC) of 0.9171 (P = 0.0002), sensitivity of 87.76%, and specificity of 100.0%. Furthermore, this study demonstrated the potential role of plasma COL10A1 in the early detection of GC, as in the early stage, we obtained an AUC of 0.8789 (P = 0.0030), sensitivity of 81.25%, and specificity of 100.0%. CONCLUSION Circulating expression level of COL10A1 is significantly increased in gastric adenocarcinoma patients being associated with poor survival and is a potential biomarker for early detection of GC.
Collapse
Affiliation(s)
- Laura Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
- Titu Maiorescu University, Faculty of Medicine, Bucharest 040441, Romania
| | - Lilia Matei
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Denisa Dragu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Ioana Pitica
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Ana Iulia Neagu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Simona Dima
- Fundeni Clinical Institute, Bucharest 022328, Romania
| | | | - Carmen C Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Mihaela Chivu-Economescu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| |
Collapse
|
279
|
The Correlation Between Helicobacter pylori Infection and Lnc-OC1 Expression in Gastric Cancer Tissues in an Iranian Population. J Gastrointest Cancer 2020; 52:600-605. [DOI: 10.1007/s12029-020-00438-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
280
|
Liu M, Li J, Huang Z, Li Y. Gastric cancer risk-scoring system based on analysis of a competing endogenous RNA network. Transl Cancer Res 2020; 9:3889-3902. [PMID: 35117756 PMCID: PMC8798172 DOI: 10.21037/tcr-19-2977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/17/2020] [Indexed: 12/24/2022]
Abstract
Background Long noncoding RNAs (lncRNAs) can play vital roles in tumor initiation, progression, invasion, and metastasis. However, the functional role of the lncRNA-based competing endogenous RNA (ceRNA) networks in gastric cancer (GC) is still unclear. We aimed to identify novel lncRNAs and their association with GC prognosis. Methods The lncRNA, miRNA, and mRNA expression profiles of GC patients data were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) were identified using the edge-R package. Then, the relationship among lncRNAs-miRNAs-mRNAs was integrated into a constructed ceRNA network with Cytoscape software. Using Cox regression analysis, a risk score system based on DEGs associated with patient prognosis in GC was established. Finally, a nomogram was founded to predict the prognosis of GC patients. Results A total of 971 differentially expressed lncRNAs (DElncRNAs), 144 differentially expressed miRNAs (DEmiRNAs) and 2,789 differentially expressed mRNAs (DEmRNAs) were identified and found to be associated with GC risk. Using the bioinformatics method, a ceRNA network involving 62 DElncRNAs, 21 DEmiRNAs and 59 DEmRNAs was constructed. Based on the results of the Cox regression analysis, a risk-scoring system involving 3 lncRNAs (i.e., ADAMTS9-AS1, C15orf54, and AL391152.1) was set up for the survival analysis of GC patients. The area under the receiver operating characteristic (ROC) curve for the risk-scoring system was 0.674, with a C-index of 0.64 [95% confidence interval (CI): 0.59–0.69, P=2.806485e−08]. Univariate and multivariate Cox regression analyses demonstrated that the risk-scoring system was an independent prognostic factor for GC. The risk-scoring system is positively associated with advanced tumor grade. The expression of these 3 lncRNAs were validated in GEPIA database. A nomogram based on these 3 lncRNAs was created to predict the prognosis of GC patients. Conclusions Our study established a novel lncRNA-expression-based ceRNA network and an ADAMTS9-AS1-C15orf54-AL391152.1-based risk-scoring system, which can be used to predict the prognosis of GC patients.
Collapse
Affiliation(s)
- Min Liu
- Department of Respiratory Medicine, The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha 41006, China
| | - Jing Li
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Zhengkai Huang
- College of Integrated Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuejun Li
- Department of Oncology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou 412000, China.,Department of Oncology, The First Affiliated Hospital of Hunan College of Chinese Medicine, Zhuzhou 412000, China
| |
Collapse
|
281
|
Katz H, Biglow L, Alsharedi M. Immune Checkpoint Inhibitors in Locally Advanced, Unresectable, and Metastatic Upper Gastrointestinal Malignancies. J Gastrointest Cancer 2020; 51:611-619. [PMID: 31028537 DOI: 10.1007/s12029-019-00243-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Upper gastrointestinal (UGI) malignancies including esophageal, gastroesophageal junction (GEJ), and gastric cancers have a poor prognosis in the metastatic setting. Treatment with cytotoxic chemotherapy remains the treatment of choice in the first-line setting with the addition of trastuzumab, a monoclonal antibody against HER-2, if the tumor is HER2-positive. Before the era of checkpoint inhibitors, there were only few treatment options after failure of the first-line systemic therapy. METHODS We extensively searched the English written literature for peer-reviewed manuscripts regarding the use of checkpoint inhibitors in advanced stage and metastatic UGI cancer. We also searched and reviewed ongoing clinical trials from Clinicaltrials.gov. RESULTS Checkpoint inhibition is a promising therapeutic option in UGI cancers, which have overexpression of PD-L1, high mutation burden, or microsatellite instability. Checkpoint inhibitors that are being investigated or are approved in advanced UGI malignancies include PD-1 antibodies, nivolumab and pembrolizumab, PD-L1 antibody, avelumab, and CTLA-4 inhibitors, ipilimumab and tremelimumab. CONCLUSIONS Based on recent and ongoing studies, eligible patients who have progressed beyond the first-line cytotoxic chemotherapy may benefit from immunotherapy. This review outlines the checkpoint inhibitors that are currently or previously being investigated for patients with metastatic UGI cancers.
Collapse
Affiliation(s)
- Heather Katz
- Department of Hematology/Oncology, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Dr, Huntington, WV, 25701, USA
| | - Layana Biglow
- Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25701, USA
| | - Mohamed Alsharedi
- Department of Hematology/Oncology, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Dr, Huntington, WV, 25701, USA.
- Division of Hematology and Oncology, Joan C. Edwards School of Medicine, Marshall University, Edwards Comprehensive Cancer Center at Cabell Huntington Hospital, 1400 Hal Greer Blvd., Huntington, WV, 25701, USA.
| |
Collapse
|
282
|
Amini M, Ghorban K, Mokhtarzadeh A, Dadmanesh M, Baradaran B. CD40 DNA hypermethylation in primary gastric tumors; as a novel diagnostic biomarker. Life Sci 2020; 254:117774. [PMID: 32407843 DOI: 10.1016/j.lfs.2020.117774] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
AIMS Gastric cancer (GC) remains one of the deadliest malignancies worldwide due to its poor prognosis. DNA methylation changes, as an early event during tumor progression, constitute attractive markers for cancer diagnostics. In the current study, CD40 DNA methylation was investigated in GC as a novel epigenetic biomarker. MAIN METHODS We first analyzed DNA methylation microarrays from the Gene Expression Omnibus database on GC samples to evaluate the potential diagnostic value of CD40 methylation. Moreover, using q-MSP, in a set of internal samples including GC primary tumors and adjacent normal specimens, CD40 DNA methylation levels were determined. The Cancer Genome Atlas (TCGA) data on GC was also analyzed for further validation. KEY FINDINGS Our results illustrated significant CD40 hypermethylation in GC samples compared to normal specimens which was significantly correlated with the clinical stage of malignancy. Besides, the high accuracy of CD40 methylation as a diagnostic biomarker in GC was confirmed using the ROC curve analysis with an AUC value of 0.9089. Also, gene set enrichment analysis showed that CD40 is mainly involved in biological processes regulating immune response activation in GC. Further analysis of other prevalent cancer entities in TCGA showed that CD40 hypermethylation is a common event during tumor progression and could be considered as a potential biomarker for the detection of breast, colorectal, and prostate cancers as well. SIGNIFICANCE The finding of this study suggests that CD40 methylation as a potential pan biomarker could be a valuable target for liquid biopsy application of human cancers.
Collapse
Affiliation(s)
- Mohammad Amini
- Department of Immunology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Khodayar Ghorban
- Department of Immunology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Dadmanesh
- Department of Infectious Diseases, School of Medicine, Aja University of Medical Sciences, Tehran, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
283
|
Tan B, Li Y, Zhao Q, Fan L, Zhang M. The impact of Harmine hydrochloride on growth, apoptosis and migration, invasion of gastric cancer cells. Pathol Res Pract 2020; 216:152995. [PMID: 32402536 DOI: 10.1016/j.prp.2020.152995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 12/29/2022]
Abstract
It has been reported that Harmine hydrochloride (HH) has an inhibitory effect on tumor cells, but the effect and mechanism of HH on gastric cancer cells remains unclear. The aim of this study was to investigate the effect and mechanism of HH on human gastric cancer cell line. In present study, results showed that HH could inhibit AGS, SGC7901 and HGC-27 cells in a time-dose-dependent manner (P < 0.01). Furthermore, this study demonstrated that more cells were arrested in G0/G1 phase, and apoptosis rate of AGS cells was significantly increased after HH treatment (P < 0.01). In addition, the study results showed that the mRNA and proteins of CyclinE, CyclinD1, PCNA declined dramatically, while p27, p21 increased significantly (P < 0.01). The results in this research also showed that the mRNA and proteins of Survivin and Bcl-2 decreased, while the expression of Bax, caspase-3, Bad increased significantly (P < 0.01). Also, the results of this study showed that invasion and migration of AGS cells decreased significantly after HH treatment (P < 0.01), with the expression of MMP-2, HIF-1 α and PRDX1 decreasing on observation after HH treatment (P < 0.01). In conclusion, HH has the property to inhibit GC cells via regulating GC cells' proliferation, apoptosis, invasion and migration.
Collapse
Affiliation(s)
- Bibo Tan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| | - Qun Zhao
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Liqiao Fan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Mingyue Zhang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| |
Collapse
|
284
|
Namikawa T, Yokota K, Yamaguchi S, Iwabu J, Munekage M, Uemura S, Tsujii S, Maeda H, Kitagawa H, Kumon M, Kobayashi M, Hanazaki K. Evaluation of Systemic Inflammatory Response and Nutritional Biomarkers as Predictive Factors in Patients with Recurrent Gastric Cancer. Oncology 2020; 98:452-459. [PMID: 32182616 DOI: 10.1159/000505973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND The present study sought to evaluate host-related factors as predictors in patients receiving chemotherapy for recurrent advanced gastric cancer. METHODS Sixty-three patients were enrolled in the study and received chemotherapy for recurrent gastric cancer at the Kochi Medical School from 2008 to 2015. Clinicopathological information and systemic inflammatory response data were obtained retrospectively to investigate associations between baseline cancer-related prognostic variables and survival outcomes. RESULTS The median survival time was significantly higher for patients with a Glasgow prognostic score (GPS) of 0 compared to a GPS of 1 or 2 (18.2 vs. 7.1 months; p = 0.006), and for patients in the normal range for carbohydrate antigen-125 (CA125) compared to higher levels (17.9 vs. 4.1 months; p = 0.003). There was no significant influence on overall survival by age, gender, disease status, metastatic site, time to recurrence, carcinoembryonic antigen level, CA19-9 level, prognostic nutrition index, or neutrophil to lymphocyte ratio according to the results of the univariate log-rank tests. Multivariate survival analysis identified a GPS of 1 or 2 (hazard ratio, 3.520; 95% confidence interval, 1.343-9.227; p = 0.010) and a high CA125 level (hazard ratio, 3.135; 95% confidence interval, 1.276-7.697; p = 0.013) as significant independent predictors associated with a poorer prognosis in the studied group of cancer patients. CONCLUSIONS A GPS of 1 or 2 and a high level of CA125 are independent predictors of a poorer prognosis in patients receiving chemotherapy for recurrent gastric cancer.
Collapse
Affiliation(s)
| | | | | | - Jun Iwabu
- Department of Surgery, Kochi Medical School, Kochi, Japan
| | | | - Sunao Uemura
- Department of Surgery, Kochi Medical School, Kochi, Japan
| | | | | | | | - Masamitsu Kumon
- Department of Surgery, Noichi Central Hospital, Kochi, Japan
| | - Michiya Kobayashi
- Department of Human Health and Medical Sciences, Kochi Medical School, Kochi, Japan
| | | |
Collapse
|
285
|
Jeong S, Oh MJ, Kim U, Lee J, Kim JH, An HJ. Glycosylation of serum haptoglobin as a marker of gastric cancer: an overview for clinicians. Expert Rev Proteomics 2020; 17:109-117. [DOI: 10.1080/14789450.2020.1740091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Seunghyup Jeong
- Asia-pacific Glycomics Reference Site, Chungnam National University, Daejeon, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Myung Jin Oh
- Asia-pacific Glycomics Reference Site, Chungnam National University, Daejeon, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Unyong Kim
- Biocomplete Inc, Seoul, Republic of Korea
| | - Jua Lee
- Asia-pacific Glycomics Reference Site, Chungnam National University, Daejeon, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Jae-Han Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Joo An
- Asia-pacific Glycomics Reference Site, Chungnam National University, Daejeon, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
286
|
Wang J, Lv W, Lin Z, Wang X, Bu J, Su Y. Hsa_circ_0003159 inhibits gastric cancer progression by regulating miR-223-3p/NDRG1 axis. Cancer Cell Int 2020; 20:57. [PMID: 32099530 PMCID: PMC7031989 DOI: 10.1186/s12935-020-1119-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Background Abnormally expressed circular RNAs (circRNAs) are implicated in the development and treatment of gastric cancer (GC). Previous study has reported that hsa_circ_0003159 is expressed in GC. However, the role and mechanism of hsa_circ_0003159 in GC progression remain unclear. Methods GC tissues and normal tissues were harvested from 55 patients in this study. The levels of hsa_circ_0003159, microRNA (miR)-223-3p and N-myc downstream regulated gene 1 (NDRG1) were measured by quantitative real-time polymerase chain reaction or western blot. Cell proliferation, migration, invasion and apoptosis were determined by cell counting kit (CCK)-8, transwell assay, flow cytometry and western blot, respectively. The target association of miR-223-3p-hsa_circ_0003159 and miR-223-3p-NDRG1 was explored by dual-luciferase reporter assay. Xenograft model was established to assess the roles of hsa_circ_0003159 in GC in vivo. Results Hsa_circ_0003159 was lowly expressed in GC tissues and cells and mainly presented in the cytoplasm. Low expression of hsa_circ_0003159 was associated with lower overall survival and disease-free survival. Hsa_circ_0003159 overexpression inhibited proliferation, migration and invasion but induced apoptosis in GC cells. MiR-223-3p was a target of hsa_circ_0003159 and abated the effect of hsa_circ_0003159 on proliferation, migration, invasion and apoptosis in GC cells. Hsa_circ_0003159 promoted NDRG1 expression by competitively sponging miR-223-3p. Knockdown of NDRG1 reversed the suppressive effect of hsa_circ_0003159 on GC progression. Besides, hsa_circ_0003159 decreased GC cell xenograft tumor growth by regulating miR-223-3p and NDRG1. Conclusion Hsa_circ_0003159 suppressed proliferation, migration, invasion and xenograft tumor growth but promoted apoptosis by decreasing miR-223-3p and increasing NDRG1 in GC, indicating a novel target for treatment of GC.
Collapse
Affiliation(s)
- Jingyu Wang
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Weize Lv
- 2Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zhidong Lin
- 3Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xiao Wang
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Juyuan Bu
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| | - Yonghui Su
- 1Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, No. 52, East Meihua Road, Zhuhai, 519000 Guangdong China
| |
Collapse
|
287
|
Ruan Y, Li Z, Shen Y, Li T, Zhang H, Guo J. Functions of circular RNAs and their potential applications in gastric cancer. Expert Rev Gastroenterol Hepatol 2020; 14:85-92. [PMID: 31922886 DOI: 10.1080/17474124.2020.1715211] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: In recent years, circular RNAs (circRNAs) have emerged in the field of RNA research and their biological functions are now being gradually identified. circRNAs are divided into three categories: exonic circular RNAs (ecircRNAs), exon-intron circular RNAs (EIciRNAs), and intronic circular RNAs (ciRNAs). The circular structure of circRNAs confers unique biological characteristics upon them, such as enhanced stability over linear RNAs.Areas covered: circRNAs function to competitively bind with microRNAs (miRNAs) and proteins, participate in protein coding, regulate transcription, and form pseudogenes after reverse transcription. In gastric cancer, the circRNA-miRNA-mRNA axis is the most studied mechanisms underlying gastric cancer occurrence and development. Some specific and sensitive circRNAs, such as hsa_circ_102958, hsa_circ_0000520, and hsa_circ_0001017 may have potential diagnostic potential in early-stage gastric cancer. Abnormal expression of some circRNAs, including circ-LMTK2, circ-PSMC3, and circ-DLST are associated with the development of gastric cancer. Other circRNAs, such as hsa_circ_0001368, circ-ZFR, and circ-ERBB2, may also play important roles in gastric cancer treatment.Expert opinion: Exploring the roles of circRNAs in gastric cancer occurrence and development will help us to elucidate the functions of circRNAs and develop potential tools for early diagnosis and effective treatment of gastric cancer.
Collapse
Affiliation(s)
- Yao Ruan
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Zhe Li
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Yijing Shen
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Tianwen Li
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Haiyan Zhang
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| | - Junming Guo
- Department of Biochemistry and Molecular Biology, And Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
288
|
Matsuoka T, Yashiro M. Precision medicine for gastrointestinal cancer: Recent progress and future perspective. World J Gastrointest Oncol 2020; 12:1-20. [PMID: 31966910 PMCID: PMC6960076 DOI: 10.4251/wjgo.v12.i1.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 10/12/2019] [Accepted: 11/04/2019] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal (GI) cancer has a high tumor incidence and mortality rate worldwide. Despite significant improvements in radiotherapy, chemotherapy, and targeted therapy for GI cancer over the last decade, GI cancer is characterized by high recurrence rates and a dismal prognosis. There is an urgent need for new diagnostic and therapeutic approaches. Recent technological advances and the accumulation of clinical data are moving toward the use of precision medicine in GI cancer. Here we review the application and status of precision medicine in GI cancer. Analyses of liquid biopsy specimens provide comprehensive real-time data of the tumor-associated changes in an individual GI cancer patient with malignancy. With the introduction of gene panels including next-generation sequencing, it has become possible to identify a variety of mutations and genetic biomarkers in GI cancer. Although the genomic aberration of GI cancer is apparently less actionable compared to other solid tumors, novel informative analyses derived from comprehensive gene profiling may lead to the discovery of precise molecular targeted drugs. These progressions will make it feasible to incorporate clinical, genome-based, and phenotype-based diagnostic and therapeutic approaches and apply them to individual GI cancer patients for precision medicine.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka 5458585, Japan
- Oncology Institute of Geriatrics and Medical Science, Osaka City University Graduate School of Medicine, Osaka 5458585, Japan
| |
Collapse
|
289
|
Ghweil AA, Osman HA, Hassan MH, Sabry AM, Mahdy RE, Ahmed AR, Okasha A, Khodeary A, Ameen HH. Validity of serum amyloid A and HMGB1 as biomarkers for early diagnosis of gastric cancer. Cancer Manag Res 2020; 12:117-126. [PMID: 32021428 PMCID: PMC6958557 DOI: 10.2147/cmar.s207934] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Background and aim Gastric carcinomais a frequent neoplasm with poor outcome, and its early detection would improve prognosis. This study was designed to evaluate the possible use of new biomarkers, namely SAA and HMGB1, for early diagnosis of gastric cancer. Methods A total of 100 patients presenting with gastric symptoms were included. All patients underwent upper endoscopic evaluation, histopathological diagnosis and serum CEA, SAA, and HMGB1 measurements. Results Patients were classed endoscopically with neoplastic, inflammatory, and normal-appearing gastric mucosa: 50, 25, and 25 patients, respectively. Histologically, half the patients had chronic gastritis and the remaining cases gastric carcinoma of diffuse (n=28) or intestinal (n=22) type. SAA at cutoff of 18.5 mg/L had the best validity to differentiate gastritis from gastric carcinoma, with AUC, sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV) of 0.99, 98%, 100%, 100%, and 98%, respectively, followed by HMGB1 at cutoff of 14.5 pg/μL, with AUC, sensitivity, specificity, PPV, and NPV of 0.91, 70%, 96%, 94.6%, and 76.2%, respectively. Sensitivity, specificity, PPV, and NPV of serum CEA at cutoff of 2.9 ng/mL to differentiate gastritis from gastric carcinoma were 42%, 72%, 60%, and 55.4%, respectively, with AUC of 0.53. Nonetheless, higher serum levels of both SAA and HMGB1 reflected higher tumor grade (P=0.027 and P=0.016, respectively) and advanced tumor stage (P-OBrk-0.001 for both). Conclusion Serum levels of both SAA and HMGB1 could be of great value for early diagnosis of gastric carcinoma, comparable to the diagnostic role of serum CEA, which is not valid for early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Ali A Ghweil
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Heba A Osman
- Tropical Medicine and Gastroenterology Department, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Mohammed H Hassan
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Abeer Mm Sabry
- Internal Medicine and Gastroenterology Department, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Reem E Mahdy
- Internal Medicine Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Rh Ahmed
- Pathology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Ahmed Okasha
- Radiology Department, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Ashraf Khodeary
- Clinical Pathology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Hesham H Ameen
- Clinical Pathology Department, Faculty of Medicine, Al-Azhar University (Assiut Branch), Assiut, Egypt
| |
Collapse
|
290
|
Moundir C, Chehab F, Senhaji N, Boufettal R, Idouz K, Erguibi D, Nadifi S. Association of the IL-17A rs2275913 and MIF rs755622 polymorphisms with the risk of gastric and colorectal cancer. Meta Gene 2019. [DOI: 10.1016/j.mgene.2019.100605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
291
|
Zhou X, Liu J, Meng A, Zhang L, Wang M, Fan H, Peng W, Lu J. Gastric juice piR-1245: A promising prognostic biomarker for gastric cancer. J Clin Lab Anal 2019; 34:e23131. [PMID: 31777102 PMCID: PMC7171314 DOI: 10.1002/jcla.23131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
Background Emerging reports demonstrated that PIWI‐interacting RNAs (piRNAs) played an indispensable role in tumorigenesis. However, it still remains elusive whether piR‐1245 in gastric juice specific in stomach could be employed as a biomarker for gastric cancer (GC). The present work is aiming at exploring the possibility of piR‐1245 in gastric juice as a potential marker to judge for diagnosis and prognosis of gastric cancer. Methods Gastric juice was collected from 66 GC patients and 66 healthy individuals. Quantitative real‐time reverse transcriptase polymerase chain reaction (qRT‐PCR) was employed to measure the levels of piR‐1245 expression. Then, the pattern of piR‐1245 expression in gastric juice was determined between GC patients and healthy individuals. A receiver operating characteristic (ROC) curve was constructed for distinguishing GC from healthy individuals. Results Gastric juice piR‐1245 levels in GC were higher than those of controls (P < .0001). The value of area under ROC (AUC) was 0.885 (sensitivity, 90.9%; specificity, 74.2%; 95% confidence interval, 0.8286 to 0.9414). High gastric juice piR‐1245 expression was signally correlated with tumor size (P = .013) and TNM stage (P = .001). GC patients with high piR‐1245 expression in gastric juice exerted a poorer overall survival (OS) (P = .0152) and progression‐free survival (PFS) (P = .013). COX regression analysis verified that gastric juice piR‐1245 expression was an independent prognostic risk variable for OS (P < .05). Conclusions The current study suggested that piR‐1245 in gastric juice had the potential to be a useful biomarker for GC detection and prognosis prediction.
Collapse
Affiliation(s)
- Xiaorong Zhou
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Jianhong Liu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Aifeng Meng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Lihong Zhang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Min Wang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Hong Fan
- Department of Gastroenterology, First People's Hospital of Yunnan Province, Kunming, China
| | - Wei Peng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Jianwei Lu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| |
Collapse
|
292
|
Lu H, Feng Y, Hu Y, Guo Y, Liu Y, Mao Q, Xue W. Spondin 2 promotes the proliferation, migration and invasion of gastric cancer cells. J Cell Mol Med 2019; 24:98-113. [PMID: 31691494 PMCID: PMC6933360 DOI: 10.1111/jcmm.14618] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 12/24/2022] Open
Abstract
Spondin 2 (SPON2), a member of the Mindin F‐Spondin family, identifies pathogens, activates congenital immunity and promotes the growth and adhesion of neurons as well as binding to their receptors, but its role in promoting or inhibiting tumour metastasis is controversial. Here, we investigated its expression levels and mechanism of action in gastric cancer (GC). Western blotting and GC tissue arrays were used to determine the expression levels of SPON2. ELISAs were performed to measure the serum levels of SPON2 in patients with GC. Two GC cell lines expressing low levels of SPON2 were used to analyse the effects of regulating SPON2 expression on proliferation, migration, invasion, the cell cycle and apoptosis. The results revealed that SPON2 was highly expressed in GC tissues from patients with relapse or metastasis. The levels of SPON2 in sera of patients with GC were significantly higher compared with those of healthy individuals and patients with atrophic gastritis. Knockdown of SPON2 expression significantly inhibited the proliferation, migration and invasion of GC cells in vitro and in vivo. Down‐regulation of SPON2 arrested the cell cycle in G1/S, accelerated apoptosis through the mitochondrial pathway and inhibited the epithelial‐mesenchymal transition by blocking activation of the ERK1/2 pathway. In summary, this study suggests that SPON2 acts as an oncogene in the development of GC and may serve as a marker for the diagnosing GC as well as a new therapeutic target for GC.
Collapse
Affiliation(s)
- Haoming Lu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| | - Ying Feng
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Yilin Hu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Yibing Guo
- Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| | - Yifei Liu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, China
| | - Qinsheng Mao
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Wanjiang Xue
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| |
Collapse
|
293
|
Bu X, Zhang A, Chen Z, Zhang X, Zhang R, Yin C, Zhang J, Zhang Y, Yan Y. Migration of gastric cancer is suppressed by recombinant Newcastle disease virus (rL-RVG) via regulating α7-nicotinic acetylcholine receptors/ERK- EMT. BMC Cancer 2019; 19:976. [PMID: 31640627 PMCID: PMC6805660 DOI: 10.1186/s12885-019-6225-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nicotinic acetylcholine receptors (nAChRs) have been reported to be overexpressed in malignancies in humans and is associated with tumorigenesis and cell migration. In previous studies of gastric cancer, alpha7 nicotinic acetylcholine receptor (α7-nAChR) overexpression leads to epithelial-mesenchymal transition (EMT) and promotes the migration of gastric cancer cells. Recombinant avirulent LaSota strain of Newcastle disease virus (NDV) expressing the rabies virus glycoprotein (rL-RVG) may promote apoptosis of gastric cancer cells and reduces the migration of lung cancer metastasis. However, whether rL-RVG inhibits migration of gastric cancer cells and what the underlying functional mechanism is remains unknown. METHODS The gastric cancer cell lines BGC and SGC were randomly divided into 3 groups: rL-RVG, NDV and Phosphate Buffered Solution (PBS) control groups. Furthermore,we adopted ACB and MLA,α7nAChR-siRNA for the overexpression and silencing of α7-nAChR.Corynoxenine was used for inhibiting the MEK-ERK pathway. Western blot, Immunofluoresce,cell proliferation assays,cell migration analyses through wound-healing assays and Transwell assays were used to explore the underlying mechanisms. A mouse xenograft model was used to investigate the effects of rL-RVG,NDV on tumor growth. RESULTS In this study, our findings demonstrate that rL-RVG suppressed the migration of gastric cancer cells and reduced EMT via α7-nAChR in vitro. Furthermore rL-RVG decreased the phosphorylation levels of the MEK/ERK signaling pathway such as down-regulating the expression of P-MEK and P-ERK. Additionally, rL-RVG also reduced the expression level of mesenchymal markers N-cadherin and Vimentin and enhanced the expression of the epithelial marker E-cadherin. Lastly, rL-RVG inhibited nicotinic acetylcholine receptors (nAChRs) to suppress cell migration and epithelial to mesenchymal transition (EMT) in gastric cell. We also found that rL-RVG suppresses the growth of gastric cancer subcutaneous tumor cells in vivo. CONCLUSION rL-RVG inhibits α7-nAChR-MEK/ERK-EMT to suppress migration of gastric cancer cells.
Collapse
Affiliation(s)
- Xuefeng Bu
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, DianLi Road No.8, Zhenjiang, 212002, JiangSu, China
| | - Anwei Zhang
- Department of Liver and gallbladder surgery, First People's Hospital of Kunshan, QianJin West Road No.91, Kunshan, 215300, JiangSu, China
- Clinical Medicine College of Jiangsu University, XueFu Road No.301, Zhenjiang, 212013, JiangSu, China
| | - Zhengwei Chen
- Clinical Medicine College of Jiangsu University, XueFu Road No.301, Zhenjiang, 212013, JiangSu, China
| | - Xuanfeng Zhang
- Clinical Medicine College of Jiangsu University, XueFu Road No.301, Zhenjiang, 212013, JiangSu, China
| | - Riting Zhang
- Clinical Medicine College of Jiangsu University, XueFu Road No.301, Zhenjiang, 212013, JiangSu, China
| | - Chaoyun Yin
- Department of General Surgery, Affiliated Hospital of Jiangsu University, JieFang Road No.438, Zhenjiang, 212001, JiangSu, China
| | - Jie Zhang
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, DianLi Road No.8, Zhenjiang, 212002, JiangSu, China
| | - Yao Zhang
- Clinical Medicine College of Jiangsu University, XueFu Road No.301, Zhenjiang, 212013, JiangSu, China
| | - Yulan Yan
- Department of Respiratory Medicine, Affiliated People's Hospital of Jiangsu University, DianLi Road No.8, Zhenjiang, 212002, JiangSu, China.
| |
Collapse
|
294
|
Dong Z, Sun X, Xu J, Han X, Xing Z, Wang D, Ge J, Meng L, Xu X. Serum Membrane Type 1-Matrix Metalloproteinase (MT1-MMP) mRNA Protected by Exosomes as a Potential Biomarker for Gastric Cancer. Med Sci Monit 2019; 25:7770-7783. [PMID: 31619663 PMCID: PMC6820360 DOI: 10.12659/msm.918486] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Our previous research revealed that membrane type 1-matrix metalloproteinase (MT1-MMP) is overexpressed and plays a crucial role in gastric cancer (GC) progression. Exosomes are important for GC carcinogenesis, and the exosomal contents are ideal biomarkers. However, the expression of exosomal MT1-MMP mRNA in serum and its potential significance in GC remains unclear. Material/Methods The expression of exosomal MT1-MMP mRNA in serum of patients with GC, chronic gastritis, or atypical hyperplasia, and healthy controls was detected using real-time quantitative RT-PCR. Serum CEA, CA19-9, and CA72-4 were also measured by electrochemiluminescence assay. Results Exosomes were isolated and identified in serum, and serum exosomal MT1-MMP mRNA was found to be higher in patients with GC compared with healthy controls and patients with chronic gastritis or atypical hyperplasia (all P<0.05). Serum exosomal MT1-MMP mRNA was significantly correlated with tumor diameter, differentiation, Borrmann type, invasion depth, lymphatic metastasis, distal metastasis, and TNM stage. The AUC of exosomal MT1-MMP mRNA was 0.788 (95% CI: 0.714–0.850) with 63.9% sensitivity and 87.1% specificity, and was higher than that of CEA (0.655 (95% CI: 0.573–0.730)). The combination of 2 markers gave an AUC of 0.821 (95% CI: 0.750–0.878), which was better than with the individual marker. The sensitivity, specificity, and positive and negative predictive values were 75.6%, 83.9%, 94.7%, and 47.3%, respectively. Moreover, the multiple logistic regression model showed that tumor diameter, differentiation, invasion depth, and exosomal MT1-MMP mRNA were the risk factors for lymphatic metastasis in GC. Conclusions Our results characterized serum exosomal MT1-MMP mRNA in GC, providing a foundation for discovering serum exosomes-targeted biomarkers for GC diagnosis.
Collapse
Affiliation(s)
- Zhaogang Dong
- Department of Clinical Laboratory Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoyan Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Jingjing Xu
- School of Microelectronics, Shandong University, Jinan, Shandong, China (mainland)
| | - Xia Han
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Zhaoquan Xing
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Ding Wang
- Department of Clinical Laboratory Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jian Ge
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Liwei Meng
- Department of Urology Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaofei Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
295
|
Comment on "circSMARCA5 Functions as a Diagnostic and Prognostic Biomarker for Gastric Cancer". DISEASE MARKERS 2019; 2019:2790379. [PMID: 31612069 PMCID: PMC6757250 DOI: 10.1155/2019/2790379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/10/2019] [Indexed: 11/17/2022]
|
296
|
Huang Y, Yang L, Lin Y, Chang X, Wu H, Chen Y. Prognostic value of non-invasive serum Cytokeratin 18 detection in gastrointestinal cancer: a meta-analysis. J Cancer 2019; 10:4814-4823. [PMID: 31598152 PMCID: PMC6775513 DOI: 10.7150/jca.31408] [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: 11/11/2018] [Accepted: 06/26/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Gastrointestinal cancer is one of the most common neoplasms. Cytokeratin 18(CK18) is widely expressed in many different organs and cancers. Emerging data suggested conflicting results about the role of CK18 during carcinogenesis. The aim of this study is to systematically review the prognostic value of circulating CK18 (M65) and caspase-Cleaved CK18 (M30) in digestive cancers. Materials and Methods: We searched major database for manuscripts reporting the effect of pretreatment CK18 on survival of digestive cancer patients. Revman5.3 and R were the software used for analysis. Pooled multivariable-adjusted hazard ratios (HR) for overall survival (OS) were calculated in all patients and many different subgroup analyses by stratifying on tumor type, metastasis stage, and ethnicity. Results: 11 original studies were included for analysis. A low level of M30 and M65 were shown to be a protective factor for all cancer patients (HR 0.49, 95%CI 0.33-0.73, P=0.0003; HR 0.48, 95% CI 0.32-0.70, P =0.0001, respectively). The low M30 remained to be a protective factor for metastasized cancer patients while M65 had no statistically significant correlation with prognosis. Conclusions: Non-invasive total and cleaved CK18 level detection by ELISA could be potentially a useful predictor of prognosis of digestive cancer patients. Further studies are warranted to investigate the molecular mechanisms of CK18.
Collapse
Affiliation(s)
- Yuejuan Huang
- Department of Chemotherapy, the People's Hospital of Baise City, No 8 Chengxiang Road, Baise, Guangxi 533000, People's Republic of China
| | - Ling Yang
- Affiliated Tumor Hospital of Guangxi Medical University, No 71 Hedi Road, Nanning, Guangxi 530021, People's Republic of China
| | - Yan Lin
- Affiliated Tumor Hospital of Guangxi Medical University, No 71 Hedi Road, Nanning, Guangxi 530021, People's Republic of China
| | - Xin Chang
- Affiliated Tumor Hospital of Guangxi Medical University, No 71 Hedi Road, Nanning, Guangxi 530021, People's Republic of China
| | - Huini Wu
- Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1St Ave, Maywood, IL 60153, USA
| | - Ying Chen
- Affiliated Tumor Hospital of Guangxi Medical University, No 71 Hedi Road, Nanning, Guangxi 530021, People's Republic of China
| |
Collapse
|
297
|
Choi RSY, Lai WYX, Lee LTC, Wong WLC, Pei XM, Tsang HF, Leung JJ, Cho WCS, Chu MKM, Wong EYL, Wong SCC. Current and future molecular diagnostics of gastric cancer. Expert Rev Mol Diagn 2019; 19:863-874. [PMID: 31448971 DOI: 10.1080/14737159.2019.1660645] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
Abstract
Introduction: Gastric cancer (GC) is the fifth most common cancer and confers the second-highest mortality among other cancers. Improving the survival rates of GC patients requires prompt and accurate diagnosis and effective treatment which is often preceded by the poorly understood pathogenic mechanisms. Area covered: This literature review aims to summarize current understanding of genetic and molecular alterations that promote carcinogenesis including (1) activation of oncogenes, (2) overexpression of growth factors, receptors and matrix metalloproteinases, (3) inactivation of tumor suppressor genes, DNA repair genes, and cell adhesion molecules and (4) alterations of cell-cycle regulators that regulate biological characteristics of cancer cells. Moreover, the significance of molecular biomarkers such as micro-RNAs (miRNAs) and long non-coding RNAs (lncRNAs) and advanced molecular techniques including droplet digital polymerase chain reaction (ddPCR), quantitative PCR (qPCR) and next-generation sequencing (NGS) are also discussed. Expert opinion: A GC-specific panel of biomarkers based on the NGS or ddPCR has the potential for diagnosis, prognosis, and monitoring treatment response in GC patients. Despite the requirements for validation in larger population in clinical studies, race-specific differences in the gene panel have also to be examined by performing the clinical trials in subjects with different races.
Collapse
Affiliation(s)
- Rachel Sin-Yu Choi
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Wing Yin Xenia Lai
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Lok Ting Claire Lee
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Wing Lam Christa Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Xiao Meng Pei
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Hin Fung Tsang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Joel Johnson Leung
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital , Kowloon , Hong Kong Special Administrative Region, China
| | - Man Kee Maggie Chu
- Division of Life Science, The Hong Kong University of Science and Technology , Clear Water Bay , Hong Kong Special Administrative Region, China
| | - Elaine Yue Ling Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| | - Sze Chuen Cesar Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University , Hong Kong , Hong Kong Special Administrative Region, China
| |
Collapse
|
298
|
Cai J, Chen Z, Zuo X. Response to: Comment on "circSMARCA5 Functions as a Diagnostic and Prognostic Biomarker for Gastric Cancer". DISEASE MARKERS 2019; 2019:9356804. [PMID: 31662805 PMCID: PMC6778885 DOI: 10.1155/2019/9356804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Juan Cai
- Department of Oncology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, Anhui, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu, China
| | - Xueliang Zuo
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, Anhui, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
299
|
Cao SQ, Zheng H, Sun BC, Wang ZL, Liu T, Guo DH, Shen ZY. Long non-coding RNA highly up-regulated in liver cancer promotes exosome secretion. World J Gastroenterol 2019; 25:5283-5299. [PMID: 31558873 PMCID: PMC6761235 DOI: 10.3748/wjg.v25.i35.5283] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/07/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Highly upregulated in liver cancer (HULC) is a long non-coding RNA (lncRNA) which has recently been identified as a key regulator in hepatocellular carcinoma (HCC) progression. However, its role in the secretion of exosomes from HCC cells remains unknown. AIM To explore the mechanism by which HULC promotes the secretion of exosomes from HCC cells. METHODS Serum and liver tissue samples were collected from 30 patients with HCC who had not received chemotherapy, radiotherapy, or immunotherapy before surgery. HULC expression in serum exosomes and liver cancer tissues of patients was measured, and compared with the data obtained from healthy controls and tumor adjacent tissues. The effect of HULC upregulation in HCC cell lines and the relationship between HULC and other RNAs were studied using qPCR and dual-luciferase reporter assays. Nanoparticle tracking analysis was performed to detect the quantity of exosomes. RESULTS HULC expression in serum exosomes of patients with HCC was higher than that in serum exosomes of healthy controls, and HULC levels were higher in liver cancer tissues than in tumor adjacent tissues. The expression of HULC in serum exosomes and liver cancer tissues correlated with the tumor-node-metastasis (TNM) classification, and HULC expression in tissues correlated with that in serum exosomes. Upregulation of HULC promoted HCC cell growth and invasion and repressed apoptosis. Notably, it also facilitated the secretion of exosomes from HCC cells. Moreover, qPCR assays showed that HULC repressed microRNA-372-3p (miR-372-3p) expression. We also identified Rab11a as a downstream target of miR-372-3p. Dual-luciferase reporter assays suggested that miR-372-3p could directly bind both HULC and Rab11a. CONCLUSION Our findings illustrate the importance of the HULC/miR-372-3p/Rab11a axis in HCC and provide new insights into the molecular mechanism regulating the secretion of exosomes from HCC cells.
Collapse
Affiliation(s)
- Shun-Qi Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, China
| | - Hong Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, China
| | - Bao-Cun Sun
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Zheng-Lu Wang
- Department of Pathology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Tao Liu
- NHC Key Laboratory of Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300192, China
| | - Dong-Hui Guo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, China
| | - Zhong-Yang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, China
| |
Collapse
|
300
|
Fu Z, Lin L, Liu S, Qin M, He S, Zhu L, Huang J. Ginkgo Biloba Extract Inhibits Metastasis and ERK/Nuclear Factor kappa B (NF-κB) Signaling Pathway in Gastric Cancer. Med Sci Monit 2019; 25:6836-6845. [PMID: 31509521 PMCID: PMC6753842 DOI: 10.12659/msm.915146] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Ginkgo biloba extract (EGb761), a standard extract of the Chinese traditional medicine Ginkgo biloba, plays an anti-tumor role in various cancers. However, whether EGb761 is involved in the invasion and metastasis of gastric cancer remains unclear. Material/Methods In the current study, cell viability assay, Western blotting, wound-healing assay, Transwell invasion assay, and orthotopic transplantation model were performed to explore the effects of EGb761 on gastric cancer. Results In vitro, the results showed that EGb761 suppressed the proliferation of gastric cancer cells in a dose-dependent manner. Furthermore, the migration and invasiveness were weakened and the protein levels of p-ERK1/2, NF-κB P65, NF-κB p-P65, and MMP2 were decreased by EGb761 or U0126 (an inhibitor of ERK signaling pathway) exposure in gastric cancer cells. Moreover, the combined treatment with EGb761 and U0126 significantly inhibited ERK, NF-κB signaling pathway, and the expression of MMP2 than those of single drug. In vivo, EGb761 inhibited the tumor growth and hepatic metastasis of gastric cancer in the mouse model. Results of immunohistochemistry indicated that the expression of ERK1/2, NF-κB P65 and MMP2 were decreased by EGb761 in the tumor tissues. Conclusions EGb761 plays a vital role in the suppression of metastasis and ERK/NF-κB signaling pathway in gastric cancer.
Collapse
Affiliation(s)
- Zhenhua Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Lan Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Shiquan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Mengbin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Siwei He
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Liye Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|