1
|
Endoscopic findings in the upper gastrointestinal tract in patients with Crohn's disease are common, highly specific, and associated with chronic gastritis. Sci Rep 2023; 13:703. [PMID: 36639398 PMCID: PMC9839771 DOI: 10.1038/s41598-022-21630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/29/2022] [Indexed: 01/14/2023] Open
Abstract
Crohn's disease (CD) may affect the entire gastrointestinal tract including its upper part. However, this aspect is poorly addressed in scientific literature and considered a rare finding. Here we aimed to prospectively investigate the prevalence, characteristics and clinical significance of upper gastrointestinal tract involvement in patients with CD, with particular focus on stomach bamboo joint-like appearance (BJA), Helicobacter pylori status and presence of microscopic changes. 375 prospectively recruited patients were included. In CD patients the prevalence of gastric and duodenal, but not esophageal, mucosal lesions, such as gastric mucosal inflammation, duodenal edema, ulcerations, and duodenal bulb deformation was significantly higher (at least p < 0.01 for all). Similar results were found when only H. pylori negative individuals were analyzed. Moreover, BJA of the stomach and in case of H. pylori negative patients also duodenal bulb deformation were detected exclusively in CD patients. Presence of BJA lesion was not significantly associated with neither duration of the disease nor use/history of biologic treatment. Despite absence of H. pylori infection microscopic features of chronic gastritis were found in almost all (93.5%) patients, and in 31% of controls (p < 0.00001). Our analysis outlines that upper gastrointestinal tract involvement in CD is a very common event and frequently manifests with a highly specific BJA lesion. Furthermore, our study reveals that in almost all CD patients features of H. pylori negative gastritis are present.
Collapse
|
2
|
Cancelliere R, Di Tinno A, Di Lellis AM, Contini G, Micheli L, Signori E. Cost-effective and disposable label-free voltammetric immunosensor for sensitive detection of interleukin-6. Biosens Bioelectron 2022; 213:114467. [PMID: 35760020 DOI: 10.1016/j.bios.2022.114467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
IL-6 detection is highly desirable since can monitor many diseases in humans and assess the response to treatments. Herein, two novel label-free voltammetric immunosensors for rapid and accurate interleukin-6 (IL-6) detection in human serum are presented. The immunosensors are fabricated by immobilising two different IL-6 antibodies, identified as mAb-IL-6 clone-5 and clone-7, on in-house produced screen-printed electrodes modified with inexpensive recycling biochar (Bio-SPEs). To ensure high structural fidelity and performance, an in-depth electrochemical characterization of the layer-by-layer assembly of the immunosensor was conducted by cyclic voltammetry (CV) and sensing was performed using square wave voltammetry (SWV). The two immunosensors showed good analytical performances in human serum, exhibiting a wide linear range (LR) between 26-125 and 30-138 pg/mL, a good limit of detection (LOD) of 4.8 and 5.4 pg/mL and selectivity for IL-6 over other common cytokines, including IL-1β and TNF-α. Performance comparison of IL-6 immunosensors with those of a commercial spectrophotometric ELISA kit (LOD of 20 pg/mL, RSD% of 15%) denotes a better sensitivity and reproducibility of the proposed label-free devices, associated with a reduced detection time (30 min instead of more than 3 h for ELISA test). Furthermore, the proposed immunosensors were successfully applied in blood samples (with only a dilution of 1:100 v/v in PBS and without additional treatments) with good sensitivity (LOD of 14.3 pg/mL) and reproducibility (RSD% < 11%), thus paving the way for their application as viable diagnostic and therapeutic point-of-care tools alternative to the IL-6 detection techniques routinely used (ELISA and Western Blot).
Collapse
Affiliation(s)
- Rocco Cancelliere
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy
| | - Alessio Di Tinno
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy; Department of Electrical and Information Engineering, University of Cassino and Southern Lazio, 03043, Cassino, FR, Italy
| | | | - Giorgio Contini
- Istituto di Struttura della Materia-CNR (ISM-CNR), Via Fosso del Cavaliere 100, 00133, Roma, Italy; Department of Physics, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy.
| | - Laura Micheli
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy.
| | - Emanuela Signori
- Istituto di Farmacologia Traslazionale-CNR (IFT-CNR), Via Fosso del Cavaliere 100, 00133, Roma, Italy.
| |
Collapse
|
3
|
Blogowski W, Dolegowska K, Deskur A, Dolegowska B, Starzynska T. Lipoxins and Resolvins in Patients With Pancreatic Cancer: A Preliminary Report. Front Oncol 2022; 11:757073. [PMID: 35087747 PMCID: PMC8787076 DOI: 10.3389/fonc.2021.757073] [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: 08/11/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Eicosanoids are bioactive lipids derived from arachidonic acid, which have emerged as key regulators of a wide variety of pathophysiological processes in recent times and are implicated as mediators of gastrointestinal cancer. In this study, we investigated the systemic levels of lipoxygenase (LOX)-derived lipoxin A4 and B4, together with resolvin D1 and D2 in patients with pancreatic adenocarcinoma (n = 68), as well as in healthy individuals (n = 32). Systemic concentrations of the aforementioned immunoresolvents were measured using an enzyme-linked immunosorbent assay (ELISA). In this study, we observed that compared with concentrations in healthy individuals, the peripheral concentrations of the aforementioned eicosanoids were significantly elevated (2- to 10-fold) in patients with pancreatic cancer (in all cases p<0.00001). No significant association was observed between eicosanoid levels and the TNM clinical staging. Furthermore, we observed no significant differences in concentrations of the analyzed bioactive lipids between patients diagnosed with early-stage (TNM stage I-II) and more advanced disease (TNM stage III-IV). Receiver operating characteristic (ROC) curve analysis of each aforementioned immunoresolvent showed area under the curve values ranging between 0.79 and 1.00. Sensitivity, specificity, as well as positive and negative predictive values of the eicosanoids involved in the detection/differentiation of pancreatic adenocarcinoma ranged between 56.8% and 100%. In summary, our research is the first study that provides clinical evidence to support a systemic imbalance in LOX-derived lipoxins and resolvins as the mechanism underlying the pathogenesis of pancreatic adenocarcinoma. This phenomenon occurs regardless of the clinical TNM stage of the disease. Furthermore, our study is the first to preliminarily highlight the role of peripheral levels of immunoresolvents, particularly resolvin D1, as potential novel biomarkers of pancreatic cancer in humans.
Collapse
Affiliation(s)
- Wojciech Blogowski
- Institute of Medical Sciences, University of Zielona Gora, Zielona Gora, Poland
| | - Katarzyna Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Anna Deskur
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Barbara Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Teresa Starzynska
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
4
|
Yang E, Chua W, Ng W, Roberts TL. Peripheral Cytokine Levels as a Prognostic Indicator in Gastric Cancer: A Review of Existing Literature. Biomedicines 2021; 9:1916. [PMID: 34944729 PMCID: PMC8698340 DOI: 10.3390/biomedicines9121916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Although strong connections exist between the carcinogenesis of gastric cancer and chronic inflammation, gastric cancer is unique in that the chronic gastritis which frequently precedes carcinogenesis is strongly associated with H. pylori infection. The interplay between H. pylori virulence factors and host immune cells is complex but culminates in the activation of inflammatory pathways and transcription factors such as NF-κB, STAT3, and AP-1, all of which upregulate cytokine production. Due to the key role of cytokines in modulating the immune response against tumour cells as well as possibly stimulating tumour growth and proliferation, different patterns of cytokine secretion may be associated with varying patient outcomes. In relation to gastric cancer, interleukin-6, 8, 10, 17A, TNF, and IFN-γ may have pro-tumour properties, although interleukin-10, TNF, and IFN-γ may have anti-tumour effects. However, due to the lack of studies investigating patient outcomes, only a link between higher interleukin-6 levels and poorer prognosis has been demonstrated. Further investigations which link peripheral cytokine levels to patient prognosis may elucidate important pathological mechanisms in gastric cancer which adversely impact patient survival and allow treatments targeting these processes to be developed.
Collapse
Affiliation(s)
- Elton Yang
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Weng Ng
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| |
Collapse
|
5
|
Yin HH, Xu MQ, Liu BZ, Tao L, Ma YJ, Li F, Zhang WJ. Combination of preoperative CA19-9 levels, cell differentiation, and age predicts survival for patients with gastric cancer before surgery. Medicine (Baltimore) 2021; 100:e28017. [PMID: 34889247 PMCID: PMC8663841 DOI: 10.1097/md.0000000000028017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 11/11/2021] [Indexed: 01/05/2023] Open
Abstract
Gastric cancer (GC) is very common in China, posing a threat to public health, with high morbidity and mortality ranks. Tumor-node-metastasis (TNM) staging system is routinely used to predict prognosis for patients with GC but only available after surgery. Therefore, searching for markers that can predict prognosis of GC patients before surgery is desirable to assist management decisions preoperatively. Among 322 GC patients followed-up for 128 months, the tumor markers alpha fetoprotein, carcinoembryonic antigen, carbohydrate antigen 19-9 (CA19-9), carbohydrate antigen 15-3 and carbohydrate antigen 72-4 of 168 patients were detected before surgery, and their impact on survival was analyzed. Four major findings were revealed: (1) Preoperative examined CA19-9 levels and cell differentiation using endoscopic biopsies were positively correlated with lymphatic metastases and TNM stages obtained after surgery. (2) Kaplan-Meier analyses demonstrated that poor survival of patients with GC was associated with higher CA19-9 levels, poor cell differentiation, and older age. (3) Cox multi-factorial regression analyses indicated that, in terms of predicting overall survival for GC patients, preoperative CA19-9 level, cell differentiation and age were independent factors, respectively, comparable to postoperative TNM staging system. (4) Using receiver operating characteristic curve analysis, we first revealed that preoperative CA19-9 levels and cell differentiation had the impact weights (IW) on survival comparable to postoperative TNM components. These findings suggest that preoperative CA19-9 levels, cell differentiation and age are useful prognostic related markers for GC patients, superior to postoperative TNM system in terms of timing for management. We propose that, assisted by clinical imaging, a comprehensive utilization of these preoperative survival-predictors may help formulate individualized medical management for GC patients such as surgical strategy, optimal chemotherapy and radiotherapy, and appropriate follow-up intervals after surgery.
Collapse
Affiliation(s)
- Hui Hui Yin
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Wuxi Branch of Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine/Xinrui Hospital of Xinwu District, Wuxi, Jiangsu, China
| | - Meng Qing Xu
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Gastroenterology, Jinling Hospital, Nanjing, Jiangsu, China
| | - Bin Zheng Liu
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lin Tao
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Ya Jing Ma
- Department of Clinical Laboratories, the First Affiliated University Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Feng Li
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Department of Pathology, Beijing Chaoyang Hospital, the Capital Medical University, Beijing, China
| | - Wen Jie Zhang
- Department of Pathology, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| |
Collapse
|
6
|
Wang X, Li J, Liu W, Zhang X, Xue L. The diagnostic value of interleukin 6 as a biomarker for gastric cancer: A meta-analysis and systematic review. Medicine (Baltimore) 2021; 100:e27945. [PMID: 34964773 PMCID: PMC8615365 DOI: 10.1097/md.0000000000027945] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer is one of the most common cancers and a main cause of global cancer death. The expression of interleukin 6 is associated with the risk of gastric cancer. But the diagnostic accuracy of interleukin 6 remains unclear. This study was designed to assess the diagnostic performance of interleukin 6 in gastric cancer diagnosis. METHODS The related data was obtained from Oncomine and studied using bioinformatics analysis. The PubMed, Embase, Cochrane Library, Web of science databases were searched for related studies published from inception to July 14, 2020. Measuring tools of diagnostic performance including sensitivity, specificity, and diagnostic odds ratio were pooled using bivariate mixed-effects meta-analysis model. The summery receiver operator characteristic curves were plotted. RESULTS The result from Oncomine showed that the expression of interleukin 6 in gastric cancer (GC) patients was higher than the normal groups (P < .05). Furthermore, a total of 4 eligible articles were enrolled, containing 390 cases and 404 controls. The diagnostic results were as follows: a sensitivity of 0.80 (95% confidence interval [CI] 0.57-0.92), a specificity of 0.86 (95% CI 0.74-0.93), a positive likelihood ratio of 5.76 (95% CI 3.49-9.49), a negative likelihood ratio of 0.23 (95% CI 0.11-0.51) and a diagnostic odds ratio of 24.58 (95% CI 14.14-42.73). The summary area under the receiver operating characteristic curves was 0.90 (95% CI 0.87-0.93). CONCLUSION Higher interleukin 6 expression was detected in GC patients, and interleukin 6 could be a helpful indicator of diagnosis of gastric cancer. Further large-scale prospective studies are required for identifying the diagnostic value of interleukin 6 in gastric cancer.
Collapse
Affiliation(s)
- Xiaozi Wang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Jie Li
- Department of Hematology, Hebei General Hospital, Shijiazhuang, China
| | - Wenjing Liu
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Liying Xue
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
7
|
Park HS, Kim YM, Kim S, Lee WS, Kong SJ, Yang H, Kang B, Cheon J, Shin SJ, Kim C, Chon HJ. High endothelial venule is a surrogate biomarker for T-cell inflamed tumor microenvironment and prognosis in gastric cancer. J Immunother Cancer 2021; 9:jitc-2021-003353. [PMID: 34670828 PMCID: PMC8529985 DOI: 10.1136/jitc-2021-003353] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Background High endothelial venule (HEV) is a specialized vasculature for lymphocyte trafficking. While HEVs are frequently observed within gastric cancer (GC), the vascular–immune interaction between HEV and tumor-infiltrating lymphocytes (TILs) has not been well elucidated. In this study, we aimed to unveil the potential value of HEVs as a surrogate marker for T-cell inflamed immune microenvironment in GC using a large number of prospectively collected surgical specimens of GC. Methods We included 460 patients with GC who underwent surgical resection. Nanostring PanCancer immune profiling was performed to evaluate the immunological phenotype of GCs. HEV density and three distinct patterns of TILs (Crohn-like lymphoid reaction, peritumoral lymphoid reaction, and intratumoral lymphoid reaction) were analyzed for their relationship and evaluated as prognostic factors for relapse-free survival (RFS) and overall survival (OS). Results HEV-high GC revealed increased infiltration by immune cell subsets, including dendritic cells, CD8+ cytotoxic T cells, and CD4+ helper T cells. In addition, HEV-high GC demonstrated increased immune-modulating chemokines, type I or II interferon pathway, and immune checkpoints, all of which indicate the inflamed tumor microenvironment (TME). All three distinct patterns of TILs were associated with HEV density. In survival analysis, patients with HEV-high GC displayed significantly longer RFS and OS than those with HEV-low GC (p<0.001 for RFS, p<0.001 for OS). Multivariate analysis demonstrated that HEV was the most significant immunological prognostic factor for RFS (patients with high HEV compared with those with low HEV; HR 0.412, 95% CI 0.241 to 0.705, p=0.001) and OS (HR 0.547, 95% CI 0.329 to 0.909, p=0.02) after adjustment for age, stage, and TIL. Conclusion HEV is the most significant immunological prognosticator for RFS and OS in resected GC, indicating inflamed TME.
Collapse
Affiliation(s)
- Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea (the Republic of)
| | - Yoo Min Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Sewha Kim
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Beodeul Kang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Jaekyung Cheon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| |
Collapse
|
8
|
Song X, Wang D, Ben B, Xiao C, Bai L, Xiao H, Zhang W, Li W, Jia J, Qi Y. Association between interleukin gene polymorphisms and susceptibility to gastric cancer in the Qinghai population. J Int Med Res 2021; 49:3000605211004755. [PMID: 33942631 PMCID: PMC8113958 DOI: 10.1177/03000605211004755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate the associations between interleukin (IL) gene polymorphisms and susceptibility to gastric cancer in the Qinghai population, China. METHODS Patients with gastric cancer and cancer-free controls were enrolled into the study from Qinghai Provincial People's Hospital between September 2016 and September 2018. Single nucleotide polymorphisms (SNPs) were genotyped with the Sequenom MassARRAY® SNP genotype system. The Hardy-Weinberg equilibrium in allele and genotype frequencies, and general characteristics between patients with gastric cancer and cancer-free controls, were evaluated using χ2-test. Potential associations between interleukin gene variants and the risk of gastric cancer were analysed by logistic regression. RESULTS Among eight candidate SNPs, the allele and genotype frequency distribution of IL-1B rs1143634 polymorphism was significantly different between patients with gastric cancer (n = 190) and cancer-free controls (n = 186). The IL-1B rs1143634 GA genotype and IL-1B rs1143634 GA + AA genotype were associated with a reduced risk of gastric cancer, however, the remaining SNPs were not statistically associated with gastric cancer risk in the Qinghai population. CONCLUSION The IL-1B rs1143634 polymorphism might be associated with a decreased risk of gastric cancer, and may be a protective factor against gastric cancer.
Collapse
Affiliation(s)
- Xiaoyan Song
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| | - Dongmei Wang
- Department of Oncology, Affiliated Hospital of Heze Medical College, Heze, China
| | - Baji Ben
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| | - Chenghua Xiao
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| | - Liyan Bai
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| | - Han Xiao
- Department of Medical Oncology, Qinghai University Affiliated Hospital, Xining, China
| | - Wenyan Zhang
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| | - Wanchao Li
- Department of Oncology, Affiliated Hospital of Heze Medical College, Heze, China
| | - Jingying Jia
- Department of Oncology, Affiliated Hospital of Heze Medical College, Heze, China
| | - Yujuan Qi
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, China
| |
Collapse
|
9
|
Genomic analysis and clinical implications of immune cell infiltration in gastric cancer. Biosci Rep 2021; 40:222774. [PMID: 32338286 PMCID: PMC7240200 DOI: 10.1042/bsr20193308] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 03/20/2020] [Accepted: 04/16/2020] [Indexed: 12/24/2022] Open
Abstract
The immune infiltration of patients with gastric cancer (GC) is closely associated with clinical prognosis. However, previous studies failed to explain the different subsets of immune cells involved in immune responses and diverse functions. The present study aimed to uncover the differences in immunophenotypes in a tumor microenvironment (TME) between adjacent and tumor tissues and to explore their therapeutic targets. In our study, the relative proportion of immune cells in 229 GC tumor samples and 22 paired matched tissues was evaluated with a Cell type Identification By Estimating Relative Subsets Of known RNA Transcripts (CIBERSORT) algorithm. The correlation between immune cell infiltration and clinical information was analyzed. The proportion of 22 immune cell subsets was assessed to determine the correlation between each immune cell type and clinical features. Three molecular subtypes were identified with ‘CancerSubtypes’ R-package. Functional enrichment was analyzed in each subtype. The profiles of immune infiltration in the GC cohort from The Cancer Genome Atlas (TCGA) varied significantly between the 22 paired tissues. TNM stage was associated with M1 macrophages and eosinophils. Follicular helper T cells were activated at the late stage. Monocytes were associated with radiation therapy. Three clustering processes were obtained via the ‘CancerSubtypes’ R-package. Each cancer subtype had a specific molecular classification and subtype-specific characterization. These findings showed that the CIBERSOFT algorithm could be used to detect differences in the composition of immune-infiltrating cells in GC samples, and these differences might be an important driver of GC progression and treatment response.
Collapse
|
10
|
TNF genetic polymorphism (rs1799964) may modify the effect of the dietary inflammatory index on gastric cancer in a case-control study. Sci Rep 2020; 10:14590. [PMID: 32883994 PMCID: PMC7471946 DOI: 10.1038/s41598-020-71433-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
The inflammatory process is known to increase the risk of gastric carcinogenesis, and both genetic and dietary factors are associated with inflammation. In the present study of 1,125 participants (373 cases and 752 controls), we determined whether the dietary inflammatory index (DII) is associated with the risk of gastric cancer (GC) and investigated whether a TNF polymorphism (rs1799964) modifies this association. Semi-quantitative food frequency questionnaire derived data were used to calculate the DII scores. Odds ratios (OR) and 95% confidence intervals (CI) were calculated using multivariable logistic models adjusted for confounders. When we stratified the data by sex, the association between GC and the DII was significant only among the women (OR, 2.27; 95% CI 1.25-4.19), and the DII effect on the risk of GC differed depending on the TNF genotype (OR, 2.30; 95% CI 1.27-4.24 in TT genotype; OR, 0.78; 95% CI 0.37-1.65 in CC + CT, p for interaction = 0.035). Furthermore, the association between the DII and GC was significant in the Helicobacter pylori-positive group; similarly, the effect differed based on the TNF genotype (OR, 1.76; 95% CI 1.13-2.73 in TT genotype; OR,0.98; 95% CI 0.54-1.77 in CT + CC, p for interaction = 0.034). In conclusion, rs1799964 may modify the effect of the DII on GC.
Collapse
|
11
|
Park H, Cho D, Huang E, Seo JY, Kim WG, Todorov SD, Ji Y, Holzapfel WH. Amelioration of Alcohol Induced Gastric Ulcers Through the Administration of Lactobacillus plantarum APSulloc 331261 Isolated From Green Tea. Front Microbiol 2020; 11:420. [PMID: 32256476 PMCID: PMC7090068 DOI: 10.3389/fmicb.2020.00420] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric inflammation is an indication of gastric ulcers and possible other underlying gastric malignancies. Epidemiological studies have revealed that several Asian countries, including South Korea, suffer from a high incidence of gastric diseases derived from high levels of stress, alcoholic consumption, pyloric infection and usage of non-steroidal anti-inflammatory drugs (NSAIDs). Clinical treatments of gastric ulcers are generally limited to proton pump inhibitors that neutralize the stomach acid, and the application of antibiotics for Helicobacter pylori eradication, both of which are known to have a negative effect on the gut microbiota. The potential of probiotics for alleviating gastrointestinal diseases such as intestinal bowel syndrome and intestinal bowel disease receives increasing scientific interest. Probiotics may support the amelioration of disease-related symptoms through modulation of the gut microbiota without causing dysbiosis. In this study the potential of Lactobacillus plantarum APSulloc 331261 (GTB1TM), isolated from green tea, was investigated for alleviating gastric inflammation in an alcohol induced gastric ulcer murine model (positive control). Treatment with the test strain significantly influenced the expression of pro-inflammatory and anti-inflammatory biomarkers, interleukin 6 (IL6) and interleukin 10 (IL10), of which the former was down- and the latter up-regulated when the alcohol induced mice were treated with the test strain. This positive effect was also indicated by less severe gastric morphological changes and the histological score of the gastric tissues. A significant increase in the abundance of Akkermansia within the GTB1TM treated group compared to the positive control group also correlated with a decrease in the ratio of acetate over propionate. The increased levels of propionate in the GTB1TM group appear to result from the impact of the test strain on the microbial population and the resulting metabolic activities. Moreover, there was a significant increase in beta-diversity in the group that received GTB1TM over that of the alcohol induced control group.
Collapse
Affiliation(s)
- Haryung Park
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
| | - Donghyun Cho
- Vital Beautie Research Division, Amore Pacific R&D Unit, Gyeonggi-do, South Korea
| | - Eunchong Huang
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
| | - Ju Yeon Seo
- Vital Beautie Research Division, Amore Pacific R&D Unit, Gyeonggi-do, South Korea
| | - Wan Gi Kim
- Vital Beautie Research Division, Amore Pacific R&D Unit, Gyeonggi-do, South Korea
| | | | - Yosep Ji
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
| | | |
Collapse
|
12
|
Kędzierska L, Madej-Michniewicz A, Marczuk N, Dołęgowska B, Starzyńska T, Błogowski W. Clinical significance of various growth factors in patients with different gastric neoplasms. Am J Transl Res 2020; 12:118-129. [PMID: 32051741 PMCID: PMC7013217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/20/2019] [Indexed: 06/10/2023]
Abstract
Growth factors represent a family of important biological molecules that can also be critical in the pathogenesis of various gastrointestinal cancers. In this study, we conducted a comprehensive analysis of the systemic levels of selected growth factors - hepatocyte, vascular-endothelial, fibroblast, and insulin-like 1 growth factors (HGF, VEGF, FGF, and IGF-1, respectively), as well as granulocyte-colony stimulating factor (G-CSF) in 75 patients with different gastric neoplasms (carcinomas, gastrointestinal stromal tumors - GISTs, neuroendocrine neoplasms - NENs, and lymphomas) and 40 healthy volunteers. Patients with gastric carcinoma or other types of gastric neoplasms had higher HGF and IGF-1 levels than healthy individuals (P < 0.05 in all cases). In comparison to healthy control subjects, systemic VEGF concentrations were elevated in patients with gastric carcinoma (P < 0.05), but not in individuals with other types of gastric malignancies. No statistically significant differences were observed between the analyzed groups in terms of FGF and G-CSF levels. When patients with gastric carcinoma were subdivided according to the Japanese classification system, significantly elevated levels of HGF, VEGF, and IGF-1 concentrations were observed in patients with advanced gastric carcinoma (extending beyond the submucosal layer of the stomach). Only the systemic levels of HGF were associated with tumor node metastasis - TNM staging, the absolute numbers of bone marrow-derived mesenchymal cells, and very small embryonic/epiblast-like stem cells circulating in patients with gastric carcinoma. ROC curves analyses demonstrated that AUC values of systemic levels of examined growth factors ranged from 0.40-0.65 (P > 0.06 in all cases). In conclusion, patients with gastric malignancies showed a systemic biochemical imbalance in multiple growth factors, which appears to be associated with clinical presentation of these neoplasms in humans. However, none of the growth factors examined here seem to be suitable diagnostic biomarkers for detecting or differentiating different types of gastric malignancies in humans.
Collapse
Affiliation(s)
- Lidia Kędzierska
- Department of Gastroenterology, Pomeranian Medical University in SzczecinPoland
| | | | - Natalia Marczuk
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in SzczecinPoland
| | - Barbara Dołęgowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in SzczecinPoland
| | - Teresa Starzyńska
- Department of Gastroenterology, Pomeranian Medical University in SzczecinPoland
| | | |
Collapse
|
13
|
Benítez J, Marra R, Reyes J, Calvete O. A genetic origin for acid-base imbalance triggers the mitochondrial damage that explains the autoimmune response and drives to gastric neuroendocrine tumours. Gastric Cancer 2020; 23:52-63. [PMID: 31250150 DOI: 10.1007/s10120-019-00982-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Type I gastric neuroendocrine tumors (gNETs) arise from hypergastrinemia in patients with autoimmune chronic atrophic gastritis. According to the classical model, the gastric H+/K+ ATPase was the causative autoantigen recognized by CD4+ T cells in chronic autoimmune scenario that secretes IL-17 and correlates with parietal cell (PC) atrophy, which drives to gastric achlorhydria and increases the risk for gastric neoplasms. However, the mechanism by which the inflammatory response correlates with PC atrophy is not clearly defined. METHODS Recently, we found that the ATP4Ap.R703C mutation impaired PC function and gastric acidification, which drove familial gNET. Our group constructed a knock-in mouse model for the ATP4A mutation, which has served us to better understand the relation between impaired capability to export protons across the plasma membrane of PCs and tumor progression. RESULTS The ATP4Ap.R703C mutation drives gastric achlorhydria, but also deregulates the acid-base balance within PCs, affecting mitochondrial biogenesis. Mitochondrial malfunction activates ROS signaling, which triggers caspase-3-mediated apoptosis of parietal cells. In addition, when gastric euchlorhydria was restored, mitochondrial function is recovered. Infection by H. pylori promotes destabilization of the mitochondria of the PCs by a mechanism similar to that described for APT4Ap.R703C carriers. CONCLUSIONS A genetic origin that drives mitochondria alteration would initiate the gastric chronic inflammation instead of the classical IL-17 secretion-mediated mechanism explanation. Gastric euchlorhydria restoration is suggested to be indicated for mitochondrial recover. Our results open a new window to understand gastric neoplasms formation but also the inflammatory mechanisms and autoimmune disorders conducted by genetic origin that composes a premalignant scenario.
Collapse
Affiliation(s)
- Javier Benítez
- Human Genetics Group, Spanish National Cancer Research Center (CNIO), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
- Network of Research on Rare Diseases (CIBERER), 28029, Madrid, Spain
| | - Roberta Marra
- Human Genetics Group, Spanish National Cancer Research Center (CNIO), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, CEINGE-Biotecnologie Avanzate, Napoli, Italia
| | - José Reyes
- Department of Gastroenterology, Hospital Comarcal de Inca, Balearic Islands Health Investigation Institute (IDISBA), 07300, Majorca, Spain
| | - Oriol Calvete
- Human Genetics Group, Spanish National Cancer Research Center (CNIO), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
- Network of Research on Rare Diseases (CIBERER), 28029, Madrid, Spain.
| |
Collapse
|
14
|
Kouroupis D, Bowles AC, Willman MA, Perucca Orfei C, Colombini A, Best TM, Kaplan LD, Correa D. Infrapatellar fat pad-derived MSC response to inflammation and fibrosis induces an immunomodulatory phenotype involving CD10-mediated Substance P degradation. Sci Rep 2019; 9:10864. [PMID: 31350444 PMCID: PMC6659713 DOI: 10.1038/s41598-019-47391-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
The infrapatellar fat pad (IFP) serves as a reservoir of Mesenchymal Stem Cells (MSC), and with adjacent synovium plays key roles in joint disease including the production of Substance P (SP) affecting local inflammatory responses and transmitting nociceptive signals. Here, we interrogate human IFP-derived MSC (IFP-MSC) reaction to inflammatory and pro-fibrotic environments (cell priming by TNFα/IFNγ and TNFα/IFNγ/CTGF exposure respectively), compared with bone marrow-derived MSC (BM-MSC). Naïve IFP-MSC exhibit increased clonogenicity and chondrogenic potential compared with BM-MSC. Primed cells experienced dramatic phenotypic changes, including a sharp increase in CD10, upregulation of key immunomodulatory transcripts, and secreted growth factors/cytokines affecting key pathways (IL-10, TNF-α, MAPK, Ras and PI3K-Akt). Naïve, and more so primed MSC (both) induced SP degradation in vitro, reproduced with their supernatants and abrogated with thiorphan, a CD10 inhibitor. These findings were reproduced in vivo in a rat model of acute synovitis, where transiently engrafted human IFP-MSC induced local SP reduction. Functionally, primed IFP-MSC demonstrated sustained antagonism of activated human peripheral blood mononuclear cells (PBMC) proliferation, significantly outperforming a declining dose-dependent effect with naïve cohorts. Collectively, our in vitro and in vivo data supports cell priming as a way to enhance the immunoregulatory properties of IFP-MSC, which selectively engraft in areas of active synovitis/IFP fibrosis inducing SP degradation, resulting in a cell-based product alternative to BM-MSC to potentially treat degenerative/inflammatory joint diseases.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Annie C Bowles
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami College of Engineering, Miami, FL, USA
| | - Melissa A Willman
- Diabetes Research Institute & Cell Transplant Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Carlotta Perucca Orfei
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all'Ortopedia, Milan, Italy
| | - Alessandra Colombini
- IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all'Ortopedia, Milan, Italy
| | - Thomas M Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Lee D Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Diego Correa
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL, USA.
- Diabetes Research Institute & Cell Transplant Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
15
|
Bodnarczuk T, Deskur A, Dolegowska K, Dolegowska B, Starzynska T, Blogowski W. Hydroxyeicosatetraenoic acids in patients with pancreatic cancer: a preliminary report. Am J Cancer Res 2018; 8:1865-1872. [PMID: 30323978 PMCID: PMC6176181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 06/08/2023] Open
Abstract
Previous experimental reports have demonstrated that lipoxygenase (LOX) derivatives of arachidonic acid (AA), such as hydroxyeicosatetraenoic acids (HETEs), may be of significance in the pathogenesis of pancreatic cancer. However, these observations have not been confirmed in clinical studies. In the current study, we comprehensively evaluated the systemic levels of selected LOX-derived HETEs such as 5-, 12- and 15-HETE in patients with pancreatic adenocarcinoma (n=36), chronic pancreatitis (n=39), and in healthy individuals (n=35). Compared to healthy individuals, patients with pancreatic adenocarcinoma showed 3-8-fold higher levels of 5-, 12- and 15-HETE (at least P<0.003). Similar results were observed in patients with chronic pancreatitis, who had elevated concentrations of all examined HETE acids compared to healthy volunteers (in all cases at least P<0.03). Interestingly, the levels of the examined HETEs were not significantly associated with the TNM stage of pancreatic cancer in our patients. Finally, analyses of receiver operating characteristic curves demonstrated that all HETEs examined had relatively low area under the curve values for discriminating pancreatic adenocarcinoma from non-cancerous conditions (0.49-0.61; P>0.05 in each case). Our study provides first preliminary clinical evidence for the significance of the examined HETEs in the clinical pathogenesis of pancreatic cancer and other pancreatic diseases in humans. Moreover, our data demonstrate that the HETEs examined here do not show sufficient clinical potential to be used as independent (bio)markers for differentiating pancreatic adenocarcinoma from other non-cancerous conditions in humans.
Collapse
Affiliation(s)
- Tomasz Bodnarczuk
- Department of Gastroenterology, Pomeranian Medical UniversitySzczecin, Poland
| | - Anna Deskur
- Department of Gastroenterology, Pomeranian Medical UniversitySzczecin, Poland
| | - Katarzyna Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical UniversitySzczecin, Poland
| | - Barbara Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical UniversitySzczecin, Poland
| | - Teresa Starzynska
- Department of Gastroenterology, Pomeranian Medical UniversitySzczecin, Poland
| | - Wojciech Blogowski
- Department of Internal Medicine, University of Zielona GoraZielona Gora, Poland
| |
Collapse
|
16
|
Interleukins 17 and 23 in patients with gastric neoplasms. Sci Rep 2016; 6:37451. [PMID: 27869179 PMCID: PMC5116626 DOI: 10.1038/srep37451] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
Recently there has been heightened interest in the potential significance of interleukin (IL)-17 and IL-23 in the development/progression of human malignancies. Here, we analyzed the systemic levels of these cytokines in 75 patients with different types of gastric neoplasms (carcinoma, gastrointestinal stromal tumors, neuroendocrine neoplasms, and lymphomas) and 42 healthy volunteers. We found that patients with all types of gastric neoplasms have significantly lower IL-23 levels. However, in comparison to the levels in healthy individuals, IL-17 concentrations were lower only in patients with types of gastric neoplasms other than carcinoma. Interestingly, IL-17 levels significantly differed between patients with early and advanced gastric carcinoma. No significant associations were detected between the systemic levels of examined interleukins and TNM staging. However, peripheral levels of IL-23 were correlated with the absolute numbers of circulating populations of bone marrow-derived mesenchymal and very small embryonic/epiblast-like stem cells in patients with gastric carcinoma. ROC curve analyses demonstrated that systemic levels of IL-17 seem to meet basic criteria for consideration as a helpful diagnostic marker in the detection of gastric carcinoma. In conclusion, our study provides translational evidence confirming the clinical significance of IL-17 and IL-23 in the pathogenesis of different types of gastric neoplasms in humans.
Collapse
|
17
|
Shen W, Yuan Y, Zhao M, Li J, Xu J, Lou G, Zheng J, Bu S, Guo J, Xi Y. Novel long non-coding RNA GACAT3 promotes gastric cancer cell proliferation through the IL-6/STAT3 signaling pathway. Tumour Biol 2016; 37:14895-14902. [PMID: 27644247 DOI: 10.1007/s13277-016-5372-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play an important role in cancer occurrence and development. We previously demonstrated that lncRNA gastric cancer-associated transcript 3 (GACAT3) was positively correlated with TNM stages, tumor size, and distant metastasis of patients with gastric cancer. However, the role of GACAT3 in gastric cancer remains unclear. In this study, to investigate its function, we synthesized small interference RNAs (siRNAs) against GACTA3 and developed a GACAT3 overexpression vector (pcDNA3-GACAT3), respectively. The siRNA-mediated knockdown of GACAT3 significantly decreased cell proliferation of the gastric cancer HGC-27 cells, in which GACAT3 is overexpressed. Furthermore, GACAT3 overexpression in gastric cancer SGC-7901 cells promoted cell growth. Moreover, GACAT3 expression in HGC-27 cells was greatly upregulated by IL-6 treatment in a concentration-dependent manner. In contrast, siRNA-mediated knockdown of STAT3 decreased GACAT3 expression even in the presence of IL-6. These results demonstrated that as a downstream target of the IL6/STAT3 signaling, lncRNA GACAT3 promotes gastric cancer cell growth suggesting that GACAT3 is an inflammatory response gene and may be served as a valuable potential target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Wanjing Shen
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Yanyan Yuan
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Ming Zhao
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jiahui Li
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jin Xu
- Department of Otorhinolaryngology, Ningbo No.7 Hospital, Ningbo, 315211, China
| | - Guoying Lou
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Jiachen Zheng
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Shizhong Bu
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Junming Guo
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China
| | - Yang Xi
- Diabetes Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road, Jiangbei District, Ningbo, 315211, China.
| |
Collapse
|