201
|
Morningstar-Wright L, Czinn SJ, Piazuelo MB, Banerjee A, Godlewska R, Blanchard TG. The TNF-Alpha Inducing Protein is Associated With Gastric Inflammation and Hyperplasia in a Murine Model of Helicobacter pylori Infection. Front Pharmacol 2022; 13:817237. [PMID: 35237167 PMCID: PMC8883333 DOI: 10.3389/fphar.2022.817237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach leading to the development of chronic gastritis, peptic ulcers and gastric adenocarcinoma. A combination of host, environment and bacterial virulence factors contribute to disease development. The H. pylori TNFα inducing protein (Tipɑ) is a virulence factor shown to induce multiple pro-inflammatory cytokines in addition to TNFα in vitro. The goal of the present study was to elucidate the role of Tipα in promoting inflammation in vivo and to identify the molecular pathways associated with Tipα associated virulence. Mice were infected with wild-type Sydney strain (SS1) or a tipα mutant (Δtipα) for 1 month and 4 months. We also completed a second 4 months infection including a 1:1 SS1 to Δtipα co-infected group in addition to SS1 and Δtipα infected groups. The expression of TNFα, and KC were significantly higher in the SS1 infected group compared to both uninfected control (naïve) and Δtipα groups. Mice infected with Tipα expressing SS1 induced more severe histological gastritis and developed hyperplasia compared to Δtipα infected mice. Microarray analysis of gastric epithelial cells co-cultured with recombinant Tipα (rTipα) demonstrates up-regulation of the NFκB pathway. This data suggest Tipα plays an important role in H. pylori induced inflammation.
Collapse
Affiliation(s)
- Lindsay Morningstar-Wright
- GeneDx, Gaithersburg, MD, United States.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Steven J Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - M Blanca Piazuelo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Renata Godlewska
- Department of Bacterial Genetics, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
202
|
Wen J, Wu S, Ma X, Zhao Y. Zuojin Pill attenuates Helicobacter pylori-induced chronic atrophic gastritis in rats and improves gastric epithelial cells function in GES-1 cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114855. [PMID: 34808298 DOI: 10.1016/j.jep.2021.114855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuojin pill (ZJP), a classical Chinese medicine formula, has been widely applied in Chinese clinical practice for the treatment of gastric injury such as acute gastric lesion, acute gastric mucosal injury, chronic unpredictable mild stress, gastroesophageal reflux disease, etc, thereby exerting anti-chronic atrophic gastritis (CAG) effects in traditional Chinese herbal medicine. AIM OF THE STUDY This study was aimed to explore the therapeutic effects and molecular mechanisms of ZJP on Helicobacter pylori (H. pylori)-induced CAG based on the comprehensive approaches. MATERIALS AND METHODS Sprague-Dawley rats were infected with H. pylori for 8 weeks to establish CAG model. Then, rats in the ZJP groups received doses of 0.63, 1.26, and 2.52 g/kg ZJP for 4 weeks. Therapeutic effects of ZJP on serum indices and the histopathology of the gastric were analyzed in vivo. Moreover, GES-1 cells were infected with H. pylori to establish gastric epithelial cell injury model in vitro. Cell viability and gastric epithelial cell morphology were detected by a high-content screening (HCS) assay. Furthermore, the relative mRNA and protein expression of JMJD2B/COX-2/VEGF axis and HMGB1/NF-κB signaling pathway in vivo and in vitro were determined by RT-PCR and Western Blotting, respectively. RESULTS The results showed that the therapeutic effects of ZJP on CAG rats were presented in down-regulation serum biochemical indices and alleviating histological damage of gastric tissue. ZJP could dose-dependently decrease the serum IL-6, MCP-1, PGE2, TNF-α, and VEGF level and significantly improved gastric tissue inflammatory lesions. Besides, ZJP has an effect on increasing cell proliferation of GES-1 cells, ameliorating H. pylori-induced gastric epithelial cell damage. It was found that ZJP has a down-regulating effect on inflammatory reaction and could inhibit the relative mRNA and protein expression of JMJD2B/COX-2/VEGF axis and HMGB1/NF-κB signaling pathway in vivo and in vitro, including JMJD2B, COX-2, VEGF, VEGFR1, and VEGFR2, which in turn reduced the damage of gastric mucosal cells. CONCLUSIONS The results suggested that ZJP exerts therapeutic effects on H. pylori-induced CAG by inhibiting the JMJD2B/COX-2/VEGF axis and HMGB1/NF-κB signaling pathway. These findings deeply explained why ZJP could be used to treat CAG clinically and clarified its pharmacological effect and potential mechanism in the treatment of CAG.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China; Department of Pharmacy, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Shihua Wu
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiao Ma
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, 100039, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
203
|
Broad learning system stacking with multi-scale attention for the diagnosis of gastric intestinal metaplasia. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
204
|
Kawamura M, Uedo N, Koike T, Kanesaka T, Hatta W, Ogata Y, Oikawa T, Iwai W, Yokosawa S, Honda J, Asonuma S, Okata H, Ohyauchi M, Ito H, Abe Y, Ara N, Kayaba S, Shinkai H, Shimokawa T. Kyoto classification risk scoring system and endoscopic grading of gastric intestinal metaplasia for gastric cancer: Multicenter observation study in Japan. Dig Endosc 2022; 34:508-516. [PMID: 34415621 DOI: 10.1111/den.14114] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The usefulness of endoscopic and histological risk assessment for gastric cancer (GC) has not been fully investigated in Japanese clinical practice. METHODS In this multicenter observation study, GC and non-GC patients were prospectively enrolled in 10 Japanese facilities. The Kyoto classification risk scoring system, the Kimura-Takemoto endoscopic atrophy classification, the endoscopic grading of gastric intestinal metaplasia (EGGIM), the operative link on gastritis assessment (OLGA) and the operative link on gastric intestinal metaplasia assessment (OLGIM) were applied to all patients. The strength of an association with GC risk was compared. In addition, important endoscopic findings in the Kyoto classification were identified. RESULTS Overall, 115 GC and 265 non-GC patients were analyzed. Each risk stratification method had a significant association with GC risk in univariate analysis. In multivariate analysis, OLGIM stage III/IV (odds ratio [OR] 2.8 [95% CI 1.5-5.3]), high EGGIM score (OR 1.8 [1.0-3.1]) and opened-type Kimura-Takemoto (OR 2.5 [1.4-4.5]) had significant associations with GC risk. In the Kyoto classification, opened-type endoscopic atrophy, invisible regular arrangement of collecting venules (RAC), extensive (>30%) intestinal metaplasia in the corpus in image-enhanced endoscopy, and map-like redness in the corpus were independent high-risk endoscopic findings. The modified Kyoto classification risk scoring system using these four findings demonstrated a better area under the receiver operating characteristic curve value (0.750, P = 0.052) than that of the original Kyoto classification (0.706). CONCLUSIONS The OLGIM stage III/IV, high EGGIM score and open-typed Kimura-Takemoto had strong association with GC risk in Japanese patients. The modified Kyoto classification risk scoring system may be useful for GC risk assessment, which warrants further validation. (UMIN000027023).
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Gastroenterology, Sendai City Hospital, Miyagi, Japan
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takashi Kanesaka
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tomoyuki Oikawa
- Department of Gastroenterology, Miyagi Cancer Center, Miyagi, Japan
| | - Wataru Iwai
- Department of Gastroenterology, Miyagi Cancer Center, Miyagi, Japan
| | - Satoshi Yokosawa
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Junya Honda
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Sho Asonuma
- Department of Gastroenterology, South Miyagi Medical Center, Miyagi, Japan
| | - Hideki Okata
- Department of Gastroenterology, South Miyagi Medical Center, Miyagi, Japan
| | - Motoki Ohyauchi
- Department of Gastroenterology, Osaki Citizen Hospital, Miyagi, Japan
| | - Hirotaka Ito
- Department of Gastroenterology, Osaki Citizen Hospital, Miyagi, Japan
| | - Yasuhiko Abe
- Division of Endoscopy, Yamagata University Hospital, Yamagata, Japan
| | - Nobuyuki Ara
- Department of Gastroenterology, National Hospital Organization Sendai Medical Center, Miyagi, Japan
| | - Shoichi Kayaba
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Hirohiko Shinkai
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Toshio Shimokawa
- Clinical Study Support Center, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
205
|
Sáenz JB, Vargas N, Cho CJ, Mills JC. Regulation of the double-stranded RNA response through ADAR1 licenses metaplastic reprogramming in gastric epithelium. JCI Insight 2022; 7:153511. [PMID: 35132959 PMCID: PMC8855806 DOI: 10.1172/jci.insight.153511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/15/2021] [Indexed: 01/17/2023] Open
Abstract
Cells recognize both foreign and host-derived double-stranded RNA (dsRNA) via a signaling pathway that is usually studied in the context of viral infection. It has become increasingly clear that the sensing and handling of endogenous dsRNA is also critical for cellular differentiation and development. The adenosine RNA deaminase, ADAR1, has been implicated as a central regulator of the dsRNA response, but how regulation of the dsRNA response might mediate cell fate during injury and whether such signaling is cell intrinsic remain unclear. Here, we show that the ADAR1-mediated response to dsRNA was dramatically induced in 2 distinct injury models of gastric metaplasia. Mouse organoid and in vivo genetic models showed that ADAR1 coordinated a cell-intrinsic, epithelium-autonomous, and interferon signaling–independent dsRNA response. In addition, dsRNA accumulated within a differentiated epithelial population (chief cells) in mouse and human stomachs as these cells reprogrammed to a proliferative, reparative (metaplastic) state. Finally, chief cells required ADAR1 to reenter the cell cycle during metaplasia. Thus, cell-intrinsic ADAR1 signaling is critical for the induction of metaplasia. Because metaplasia increases cancer risk, these findings support roles for ADAR1 and the response to dsRNA in oncogenesis.
Collapse
Affiliation(s)
- José B Sáenz
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Nancy Vargas
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Charles J Cho
- Section of Gastroenterology and Hepatology, Department of Medicine
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine.,Department of Pathology and Immunology; and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
206
|
Abstract
Gastric cancer (GC) is a leading contributor to global cancer incidence and mortality. Pioneering genomic studies, focusing largely on primary GCs, revealed driver alterations in genes such as ERBB2, FGFR2, TP53 and ARID1A as well as multiple molecular subtypes. However, clinical efforts targeting these alterations have produced variable results, hampered by complex co-alteration patterns in molecular profiles and intra-patient genomic heterogeneity. In this Review, we highlight foundational and translational advances in dissecting the genomic cartography of GC, including non-coding variants, epigenomic aberrations and transcriptomic alterations, and describe how these alterations interplay with environmental influences, germline factors and the tumour microenvironment. Mapping of these alterations over the GC life cycle in normal gastric tissues, metaplasia, primary carcinoma and distant metastasis will improve our understanding of biological mechanisms driving GC development and promoting cancer hallmarks. On the translational front, integrative genomic approaches are identifying diverse mechanisms of GC therapy resistance and emerging preclinical targets, enabled by technologies such as single-cell sequencing and liquid biopsies. Validating these insights will require specifically designed GC cohorts, converging multi-modal genomic data with longitudinal data on therapeutic challenges and patient outcomes. Genomic findings from these studies will facilitate 'next-generation' clinical initiatives in GC precision oncology and prevention.
Collapse
Affiliation(s)
- Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Patrick Tan
- Singapore Gastric Cancer Consortium, Singapore, Singapore.
- Cancer and Stem Cell Biology, Duke-NUS Medical School Singapore, Singapore, Singapore.
- Genome Institute of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
207
|
Salazar BE, Pérez-Cala T, Gomez-Villegas SI, Cardona-Zapata L, Pazos-Bastidas S, Cardona-Estepa A, Vélez-Gómez DE, Justinico-Castro JA, Bernal-Cobo A, Dávila-Giraldo HA, Benítez-Guerra JC, Valencia-Cárdenas JT, Ospina EDJ, Castaño-Llano R, Bravo MM, Cataño-Correa JC, Zabaleta J, Trespalacios-Rangel AA, Cock-Botero AM, Roldán-Pérez MI, Martínez A. The OLGA-OLGIM staging and the interobserver agreement for gastritis and preneoplastic lesion screening: a cross-sectional study. Virchows Arch 2022; 480:759-769. [PMID: 35089403 DOI: 10.1007/s00428-022-03286-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/04/2022] [Accepted: 01/21/2022] [Indexed: 12/20/2022]
Abstract
Stomach cancer (SC) incidence and mortality are relevant public health issues worldwide. In Colombia, screening for preneoplastic lesions (PNL) and the presence of H. pylori is not routinely performed. Therefore, the aim of this study was to evaluate OLGA-OLGIM staging and the interobserver agreement in gastritis and preneoplastic lesions in patients with gastroduodenal symptoms from Colombia. A cross-sectional study was conducted in 272 patients with gastroduodenal symptoms. Gastric biopsies were taken following the Updated Sydney System with the OLGA-OLGIM classification, and the results were evaluated by two pathologists. Chronic gastritis and PNL were reported in 76% and 24% of the patients, respectively. Furthermore, 25% of the patients with PNL displayed gastric atrophy (GA) and 75% intestinal metaplasia (IM). Agreement in the histopathological reading for IM was good, whereas for OLGA was variable, and for the H. pylori quantity was poor. OLGA-OLGIM stages 0-II were the most frequent (96%), while stage III (4%) and SC (4%) were the least frequent. Age and coffee consumption were associated with a higher prevalence of PNL. This work determined that 4% of the population is at high risk of developing SC and would benefit from follow-up studies. Reinforcement of training programs to improve the agreement in histopathology readings is required.
Collapse
Affiliation(s)
- Beatriz E Salazar
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia.
| | - Tania Pérez-Cala
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Sara Isabel Gomez-Villegas
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Laura Cardona-Zapata
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Sebastián Pazos-Bastidas
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Alejandra Cardona-Estepa
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Diego Enrique Vélez-Gómez
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| | | | - Andrés Bernal-Cobo
- Department of Pathology, School of Medicine, University of Antioquia, Medellín, Colombia
| | | | - Juan Carlos Benítez-Guerra
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia.,Promedan IPS, Medellín, Colombia
| | | | | | | | | | | | - Jovanny Zabaleta
- Department of Integrative Oncology and Department of Pediatrics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | | | - Miguel Ignacio Roldán-Pérez
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia.,Department of Pathology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Alonso Martínez
- Bacteria & Cancer Group, Department of Microbiology, School of Medicine, University of Antioquia, Medellín, Colombia
| |
Collapse
|
208
|
Zan X, Chen Z, Guo Q, Wang Y, Zhang Z, Ji R, Zheng Y, Zhang J, Wu Z, Li M, Wang X, Ye Y, Li X, An F, Xu C, Lu L, Fan P, Zhang J, Guan Q, Li Q, Liu M, Ren Q, Hu X, Lu H, Wang Y, Zhang H, Zhao Y, Gou X, Shu X, Wang J, Hu Z, Liu R, Yuan H, Liu J, Qiao L, Zhou Y. The Association of Trefoil Factors with Gastric Cancer and Premalignant Lesions: A Cross-sectional Population-based Cohort Study. Cancer Epidemiol Biomarkers Prev 2022; 31:625-632. [PMID: 35027436 DOI: 10.1158/1055-9965.epi-21-0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/18/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A lack of research on the association of trefoil factors (TFFs) with gastric cancer (GC) and premalignant lesions (PMLs) in the general populations is an important obstacle to the application of TFFs for GC screening. We aimed to analyze the association of TFFs with GC and PMLs in a general population. METHODS We evaluated 3,986 adults residing in Wuwei, China. We collected baseline characteristics and GC risk factors, including TFFs, endoscopic diagnosis, and pathological information. Three logistic regression models were generated to analyze the association between TFFs and GC, as well as PMLs. Adjusted odds ratio (OR) and 95% confidence intervals (95% CI) were calculated to determine the strength of association. RESULTS Compared with pepsinogen (PG) and anti-Helicobacter pylori immunoglobulin G antibody (Hp-IgG), TFFs had significant association with GC and PMLs after adjusting for biomarkers and risk factors (P < 0.05). The ORs [95% CI] for TFF1 (1.67 [1.27-2.20]), TFF2 (2.66 [2.01-3.51]), and TFF3 (1.32 [1.00-1.74]) were larger than the ORs for PGI (0.79 [0.61-1.03]), PGI/II (1.00 [0.76-1.31]) and Hp-IgG (0.99 [0.73-1.35]) in the GC group. In intestinal metaplasia (IM) group, not only the TFF3 serum level was the highest, but also the OR (1.92 [1.64-2.25]) was the highest. CONCLUSIONS Trefoil factor were associated with risk of GC and PMLs. IMPACT Serum TFFs can improve the screening of high-risk populations for GC.
Collapse
Affiliation(s)
- Xiangyi Zan
- Department of Gastroenterology, The First Affiliated Hospital of Lanzhou University
| | - Zhaofeng Chen
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qinghong Guo
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuping Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital
| | - Rui Ji
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Ya Zheng
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | - Min Li
- School of Basic Medical Sciences, Lanzhou University
| | - Xiang Wang
- Department of Gastroenterology, Lanzhou University Second Hospital
| | - Yuwei Ye
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | | | | | | | - Jun Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Quanlin Guan
- First Hospital of Lanzhou University, Lanzhou University
| | - Qiang Li
- The First Affiliated Hospital of Lanzhou University
| | - Min Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qian Ren
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaobin Hu
- Lanzhou University School of Public Health
| | - Hong Lu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuling Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hongling Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yue Zhao
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xi Gou
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaochuang Shu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jun Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zenan Hu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Rong Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hao Yuan
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jiankang Liu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Liang Qiao
- Storr Liver Centre, the Westmead Institute for Medical Research, the University of Sydney at the Westmead
| | - Yongning Zhou
- Department of Gastroenterology, First Hospital of Lanzhou University
| |
Collapse
|
209
|
Development and Application of Magnetically Controlled Capsule Endoscopy in Detecting Gastric Lesions. Gastroenterol Res Pract 2022; 2021:2716559. [PMID: 35003252 PMCID: PMC8739542 DOI: 10.1155/2021/2716559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
In the past 20 years, several magnetically controlled capsule endoscopes (MCCE) have been developed for the evaluation of gastric lesions, including NaviCam (ANKON), MiroCam-Navi (Intromedic), Endocapsule MGCE (Olympus and Siemens), SMCE (JIFU), and FAMCE (Jinshan). Although limited to observing esophageal and duodenal lesions and lacking the ability of biopsy, MCCE has the advantages of comfort, safety, no anesthesia, no risk of cross-infection, and high acceptability. Several high-quality RCTs showed that the diagnostic accuracy of MCCE is comparable to the traditional gastroscopy. Due to the nonnecessity of anesthesia, MCCE may be more suitable for the elderly with obvious comorbidities as well as children. With more evidences accumulated and more innovative technologies developed, MCCE is expected to be an important tool for screening of early gastric cancer or the diagnosis of gastric diseases.
Collapse
|
210
|
Park SY, Jeong SH, Jung EJ, Ju YT, Jeong CY, Kim JY, Park T, Park J, Kim TH, Park M, Yang JW, Lee YJ. PHLPP1 Overexpression was Associated With a Good Prognosis With Decreased AKT Activity in Gastric Cancer. Technol Cancer Res Treat 2022; 21:15330338211067063. [PMID: 34982011 PMCID: PMC8733352 DOI: 10.1177/15330338211067063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: The aim of this study was to perform a clinicopathologic analysis of PHLPP1 expression in gastric cancer patients and analyze AKT activity with chemotherapy drug treatment in cancer subtypes. Materials and Methods: Surgically resected gastric cancer tissue specimens were obtained from 309 patients who underwent gastrectomy, and PHLPP1 expression was validated by tissue microarray analysis with immunohistochemistry. We assessed whether PHLPP1 selectively dephosphorylates Ser473 of AKT in an in-vitro study. Results: We found that the PHLPP1 overexpression (OE) group showed significantly greater proportions of differentiated subtype samples and early T stage samples, lower lymph node metastasis, and lower TNM stage than the PHLPP1 underexpression (UE) group. The overall survival of the PHLPP1-OE group was significantly higher (53.39 ± 0.96 months) than that of the PHLPP1-UE group (47.82 ± 2.57 months) (P = .01). In vitro analysis, we found that the PHLPP1-OE group showed a significant decrease in relative AKT S-473 levels in both cell lines (MKN-74 and KATO-III). We found that treatment with chemotherapy drugs decreased the activity of Ser473 in the MKN-74 cell line with PHLPP1 OE, but it did not affect the activity of Ser473 in KATO-III cells. Conclusion: We found that patients who overexpressed PHLPP1 showed low recurrence and good prognosis. PHLPP1 was found to work by lowering the activity of AKT Ser473 in gastric cancer. Additionally, we found a clue regarding the mechanism of chemotherapeutic drug resistance in a cell line of signet ring cell origin and will uncover this mechanism in the future.
Collapse
Affiliation(s)
- Sun Yi Park
- 26720Gyeongsang National University, Jinju, South Korea
| | - Sang-Ho Jeong
- 26720Gyeongsang National University, Jinju, South Korea.,553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Eun-Jung Jung
- 26720Gyeongsang National University, Jinju, South Korea.,553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Young-Tae Ju
- 26720Gyeongsang National University, Jinju, South Korea
| | | | - Ju-Yeon Kim
- 26720Gyeongsang National University, Jinju, South Korea
| | - Taejin Park
- 553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Jiho Park
- 26720Gyeongsang National University, Jinju, South Korea
| | - Tae-Han Kim
- 553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Miyeong Park
- 553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Jung Wook Yang
- Gyeongsang National University Hospital, Jinju, South Korea
| | - Young-Joon Lee
- 26720Gyeongsang National University, Jinju, South Korea.,553954Gyeongsang National University Changwon Hospital, Changwon, South Korea
| |
Collapse
|
211
|
Zhuang H, Bao A, Tan Y, Wang H, Xie Q, Qiu M, Xiong W, Liao F. Application and prospect of artificial intelligence in digestive endoscopy. Expert Rev Gastroenterol Hepatol 2022; 16:21-31. [PMID: 34937459 DOI: 10.1080/17474124.2022.2020646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the progress of science and technology, artificial intelligence represented by deep learning has gradually begun to be applied in the medical field. Artificial intelligence has been applied to benign gastrointestinal lesions, tumors, early cancer, inflammatory bowel disease, gallbladder, pancreas, and other diseases. This review summarizes the latest research results on artificial intelligence in digestive endoscopy and discusses the prospect of artificial intelligence in digestive system diseases. AREAS COVERED We retrieved relevant documents on artificial intelligence in digestive tract diseases from PubMed and Medline. This review elaborates on the knowledge of computer-aided diagnosis in digestive endoscopy. EXPERT OPINION Artificial intelligence significantly improves diagnostic accuracy, reduces physicians' workload, and provides a shred of evidence for clinical diagnosis and treatment. Shortly, artificial intelligence will have high application value in the field of medicine.
Collapse
Affiliation(s)
- Huangming Zhuang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Anyu Bao
- Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yulin Tan
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hanyu Wang
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingfang Xie
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Meiqi Qiu
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanli Xiong
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fei Liao
- Gastroenterology Department, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
212
|
Fukuda K, Kodama M, Mizukami K, Okamoto K, Ogawa R, Hirashita Y, Fukuda M, Togo K, Matsunari O, Okimoto T, Murakami K. Analysis of long-term serological and histological changes after eradication of <i>Helicobacter pylori</i>. J Clin Biochem Nutr 2022; 71:151-157. [PMID: 36213784 PMCID: PMC9519420 DOI: 10.3164/jcbn.21-164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022] Open
Abstract
Stratification of gastric cancer risk by measuring serological biomarkers is useful for screening of gastric cancer. However, this method has problem such as overlooking past infected patients. We aimed to evaluate the association between Helicobacter pylori infection status and serological biomarkers. We divided 5,268 patients according to Helicobacter pylori infection status and past infected patients were divided into 12 groups according to time elapsed since eradication. We analyzed mean serum H. pylori immunoglobulin G antibody, pepsinogen titers, histological and endoscopic atrophy score of each group. Mean H. pylori immunoglobulin G antibody showed a decreasing tendency, there was no significant difference from the uninfected group at 11 years after eradication (p = 0.19). PGI, PGII decreased in short term after eradication. However, both PGI and PGII gradually increased as long-term changes after eradication, became comparable to those in the uninfected group (p = 0.41, p = 0.37, respectively). Histological atrophy improved gradually, became equivalent to uninfected group. Endoscopic atrophy score did not improve for long term after eradication. In conclusion, patients with long term after eradication reach the uninfected condition serologically, histologically. Endoscopic assessment of gastric mucosal atrophy may be useful for accurate assessment of gastric cancer risk.
Collapse
Affiliation(s)
- Kensuke Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Masaaki Kodama
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Ryo Ogawa
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Yuka Hirashita
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Masahide Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Kazumi Togo
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Osamu Matsunari
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Tadayoshi Okimoto
- Department of Gastroenterology, Faculty of Medicine, Oita University
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University
| |
Collapse
|
213
|
Kathi PR, Babaria R, Banerjee B. Impact of helicobacter pylori on human physiology and digestive disorders. NUTRITION AND FUNCTIONAL FOODS IN BOOSTING DIGESTION, METABOLISM AND IMMUNE HEALTH 2022:193-205. [DOI: 10.1016/b978-0-12-821232-5.00021-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
214
|
Association between triglyceride-glucose index and gastric carcinogenesis: a health checkup cohort study. Gastric Cancer 2022; 25:33-41. [PMID: 34355281 DOI: 10.1007/s10120-021-01222-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND We aimed to investigate the association between the triglyceride-glucose (TyG) index and gastric carcinogenesis, including precancerous conditions such as dysplasia, atrophic gastritis, and intestinal metaplasia. METHODS Patients who received an upper endoscopic assessment at a medical center were included. The enrolled patients were divided into four categories according to their TyG index quartile (Q). To evaluate the relationship between increase of TyG index and gastric cancer, we analyzed the patients who received a health checkup twice. Moreover, receiver-operating characteristic curve analysis was used to establish cut-off value of the TyG index for gastric cancer. RESULTS Of 127,564 enrolled patients, 43,525 (34.1%) and 186 (0.1%) were diagnosed with precancerous conditions and gastric cancer, respectively. The odds ratios (ORs) of precancerous conditions given TyG index progressively increased across quartiles: using Q1 as the reference: Q2 (OR = 1.403, P < 0.001), Q3 (OR = 1.646, P < 0.001), and Q4 (OR = 1.656, P < 0.001). The ORs of gastric cancer also increased according to the quartiles: Q2 (OR = 1.619, P = 0.045), Q3 (OR = 2.180, P = 0.004), and Q4 (OR = 2.363, P = 0.001). Moreover, the increase in TyG index between baseline and follow-up tests was more significant in gastric cancer group than in control group (P = 0.001). The optimal cut-off value for predicting gastric cancer was 9.73. CONCLUSIONS The TyG index may be a novel predictive biomarker for gastric carcinogenesis. Notably, increase in the TyG index is significantly associated with gastric cancer.
Collapse
|
215
|
Relationship of prognostic factors in stomach cancer with Helicobacter pylori: A retrospective study. Acta Gastroenterol Belg 2022; 85:35-45. [PMID: 35304992 DOI: 10.51821/85.1.7352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background and study aims The prognostic value of H. pylori, which infects more than half of the human population living in the world and plays a role in gastric cancer pathogenesis, is controversial. Our aim is to investigate the relationship between H. pylori and prognostic factors in gastric cancer. Patients and methods The data of 110 patients (38 females and 72 males) that underwent surgeries due to gastric cancer between 2014 and 2017 were retrospectively analyzed. The relationships between survival (disease-free and overall) and factors such as p53, HER2/neu, Ki-67, neutrophil and platelet lymphocyte ratio (NLR / PLR), histopathological and demographic characteristics were examined. In addition, the results of H. pylori positive and negative groups were compared. Results Sixty-one (55%) patients were H. pylori negative and 49 (45%) were positive. In multivariate analysis, TNM stage, lymph node capsule invasion and NLR were determined as independent prognostic factors in both disease-free and overall survival. Age>62 and PLR>14.3 were determined as independent predictive factors of poor prognosis in overall survival. In univariate analysis, tumor diameter of >4.3 cm, lymphovascular and perineural invasion, and diffuse p53 expression were determined as predictive factors of poor prognosis in disease-free and overall survival. The effectiveness of these markers in prognosis was not different between H. pylori negative and positive groups. Conclusion While age, tumor diameter, TNM stage, lymph node capsule invasion, perineural and lymphovascular invasion, diffuse p53, PLR, and NLR were determined as prognostic factors in gastric cancer, these factors were not affected by the presence of H. pylori.
Collapse
|
216
|
Lin Y, Wang C, Kikuchi S, Akita T, Tanaka J, Abe SK, Hirabayashi M, Saito E, Hori M, Katanoda K, Matsuda T, Collaborators Manami Inoue; the Cancer PAF Japan. Burden of cancer attributable to infection in Japan in 2015. GHM OPEN 2021; 1:63-69. [PMID: 40145072 PMCID: PMC11933937 DOI: 10.35772/ghmo.2021.01016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/09/2021] [Accepted: 12/04/2021] [Indexed: 03/28/2025]
Abstract
Population attributable fraction (PAF) offers a means to quantify cancer burden that is attributable to a specific etiological factor. To better characterize the current cancer burden due to infection in the Japanese population, we estimated the PAF for cancer incidence and mortality in 2015 that could be attributable to infectious agents, including Helicobacter pylori (H. pylori), Hepatitis B and C (HBV/HCV), Human papillomavirus virus (HPV), Epstein- Barr virus, and human T-lymphotropic virus type 1. We estimated the PAFs for each infectious agent on the basis of representative data on prevalence and risk-outcome associations assuming a latency period of 10 years. Overall, 16.6% of cancer cases in 2015 in Japan were attributable to the infectious agents included in this analysis. The estimated PAF was slightly higher in men (18.1%) than in women (14.7%). The highest proportion of cancer deaths attributable to infectious agents was observed for H. pylori infection, followed by HBV/HCV, and HPV infection. Our findings corroborated with previous estimates that H. pylori and HBV/HCV infections were the two most important infectious agents in the Japanese population. Strategies focusing on eradication of infectious agents among infected individuals or primary prevention through vaccination could decrease the burden of infection-related cancers.
Collapse
Affiliation(s)
- Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Chaochen Wang
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tomoyuki Akita
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sarah Krull Abe
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Mayo Hirabayashi
- Division of Prevention, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Eiko Saito
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Megumi Hori
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Kota Katanoda
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Tomohiro Matsuda
- National Cancer Registry Section Center for Cancer Registries Center for Cancer Control and Information Services/Office of International Affairs, Strategic Planning Bureau National Cancer Center, National Cancer Center, Tokyo, Japan
| | | |
Collapse
|
217
|
Ray AK, Luis PB, Mishra SK, Barry DP, Asim M, Pandey A, Chaturvedi M, Gupta J, Gupta S, Mahant S, Das R, Kumar P, Shalimar, Wilson KT, Schneider C, Chaturvedi R. Curcumin Oxidation Is Required for Inhibition of Helicobacter pylori Growth, Translocation and Phosphorylation of Cag A. Front Cell Infect Microbiol 2021; 11:765842. [PMID: 35004346 PMCID: PMC8740292 DOI: 10.3389/fcimb.2021.765842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/02/2021] [Indexed: 01/22/2023] Open
Abstract
Curcumin is a potential natural remedy for preventing Helicobacter pylori-associated gastric inflammation and cancer. Here, we analyzed the effect of a phospholipid formulation of curcumin on H. pylori growth, translocation and phosphorylation of the virulence factor CagA and host protein kinase Src in vitro and in an in vivo mouse model of H. pylori infection. Growth of H. pylori was inhibited dose-dependently by curcumin in vitro. H. pylori was unable to metabolically reduce curcumin, whereas two enterobacteria, E. coli and Citrobacter rodentium, which efficiently reduced curcumin to the tetra- and hexahydro metabolites, evaded growth inhibition. Oxidative metabolism of curcumin was required for the growth inhibition of H. pylori and the translocation and phosphorylation of CagA and cSrc, since acetal- and diacetal-curcumin that do not undergo oxidative transformation were ineffective. Curcumin attenuated mRNA expression of the H. pylori virulence genes cagE and cagF in a dose-dependent manner and inhibited translocation and phosphorylation of CagA in gastric epithelial cells. H. pylori strains isolated from dietary curcumin-treated mice showed attenuated ability to induce cSrc phosphorylation and the mRNA expression of the gene encoding for IL-8, suggesting long-lasting effects of curcumin on the virulence of H. pylori. Our work provides mechanistic evidence that encourages testing of curcumin as a dietary approach to inhibit the virulence of CagA.
Collapse
Affiliation(s)
- Ashwini Kumar Ray
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Department of Microbiology, Saheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Paula B. Luis
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | | | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Achyut Pandey
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Maya Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Jyoti Gupta
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Shilpi Gupta
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Shweta Mahant
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| | - Rajashree Das
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| | - Pramod Kumar
- Department of Chemistry, Sri Aurobindo College, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Claus Schneider
- Department of Pharmacology and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
218
|
Rojas A, Lindner C, Schneider I, Gonzàlez I, Araya H, Morales E, Gómez M, Urdaneta N, Araya P, Morales MA. Diabetes mellitus contribution to the remodeling of the tumor microenvironment in gastric cancer. World J Gastrointest Oncol 2021; 13:1997-2012. [PMID: 35070037 PMCID: PMC8713306 DOI: 10.4251/wjgo.v13.i12.1997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/10/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Compelling pieces of evidence derived from both clinical and experimental research has demonstrated the crucial contribution of diabetes mellitus (DM) as a risk factor associated with increased cancer incidence and mortality in many human neoplasms, including gastric cancer (GC). DM is considered a systemic inflammatory disease and therefore, this inflammatory status may have profound effects on the tumor microenvironment (TME), particularly by driving many molecular mechanisms to generate a more aggressive TME. DM is an active driver in the modification of the behavior of many cell components of the TME as well as altering the mechanical properties of the extracellular matrix (ECM), leading to an increased ECM stiffening. Additionally, DM can alter many cellular signaling mechanisms and thus favoring tumor growth, invasion, and metastatic potential, as well as key elements in regulating cellular functions and cross-talks, such as the microRNAs network, the production, and cargo of exosomes, the metabolism of cell stroma and resistance to hypoxia. In the present review, we intend to highlight the mechanistic contributions of DM to the remodeling of TME in GC.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Cristian Lindner
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Iván Schneider
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Ileana Gonzàlez
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Hernan Araya
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Erik Morales
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Anatomía Patologica, Hospital Regional de Talca, Talca 34600000, Chile
| | - Milibeth Gómez
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Nelson Urdaneta
- Department of Clinical Sciences, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
- Servicio de Oncología, Hospital Regional de Talca, Talca 34600000, Chile
| | - Paulina Araya
- Biomedical Research Lab., Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Miguel Angel Morales
- Department of Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago 8320000, Chile
| |
Collapse
|
219
|
Rodriguez AM, Urrea DA, Prada CF. Helicobacter pylori virulence factors: relationship between genetic variability and phylogeographic origin. PeerJ 2021; 9:e12272. [PMID: 34900406 PMCID: PMC8628625 DOI: 10.7717/peerj.12272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/17/2021] [Indexed: 01/18/2023] Open
Abstract
Background Helicobacter pylori is a pathogenic bacteria that colonize the gastrointestinal tract from human stomachs and causes diseases including gastritis, peptic ulcers, gastric lymphoma (MALT), and gastric cancer, with a higher prevalence in developing countries. Its high genetic diversity among strains is caused by a high mutation rate, observing virulence factors (VFs) variations in different geographic lineages. This study aimed to postulate the genetic variability associated with virulence factors present in the Helicobacter pylori strains, to identify the relationship of these genes with their phylogeographic origin. Methods The complete genomes of 135 strains available in NCBI, from different population origins, were analyzed using bioinformatics tools, identifying a high rate; as well as reorganization events in 87 virulence factor genes, divided into seven functional groups, to determine changes in position, number of copies, nucleotide identity and size, contrasting them with their geographical lineage and pathogenic phenotype. Results Bioinformatics analyses show a high rate of gene annotation errors in VF. Analysis of genetic variability of VFs shown that there is not a direct relationship between the reorganization and geographic lineage. However, regarding the pathogenic phenotype demonstrated in the analysis of many copies, size, and similarity when dividing the strains that possess and not the cag pathogenicity island (cagPAI), having a higher risk of developing gastritis and peptic ulcer was evidenced. Our data has shown that the analysis of the overall genetic variability of all VFs present in each strain of H. pylori is key information in understanding its pathogenic behavior.
Collapse
Affiliation(s)
- Aura M Rodriguez
- Grupo de Investigación de Biología y Ecología de Artrópodos. Facultad de Ciencias, Universidad del Tolima, Ibague, Tolima, Colombia
| | - Daniel A Urrea
- Laboratorio de Investigaciones en Parasitología Tropical. Facultad de Ciencias, Universidad del Tolima, Ibague, Tolima, Colombia
| | - Carlos F Prada
- Grupo de Investigación de Biología y Ecología de Artrópodos. Facultad de Ciencias, Universidad del Tolima, Ibague, Tolima, Colombia
| |
Collapse
|
220
|
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
|
221
|
Sáenz JB. Follow the Metaplasia: Characteristics and Oncogenic Implications of Metaplasia's Pattern of Spread Throughout the Stomach. Front Cell Dev Biol 2021; 9:741574. [PMID: 34869328 PMCID: PMC8633114 DOI: 10.3389/fcell.2021.741574] [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] [Received: 07/14/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
The human stomach functions as both a digestive and innate immune organ. Its main product, acid, rapidly breaks down ingested products and equally serves as a highly effective microbial filter. The gastric epithelium has evolved mechanisms to appropriately handle the myriad of injurious substances, both exogenous and endogenous, to maintain the epithelial barrier and restore homeostasis. The most significant chronic insult that the stomach must face is Helicobacter pylori (Hp), a stomach-adapted bacterium that can colonize the stomach and induce chronic inflammatory and pre-neoplastic changes. The progression from chronic inflammation to dysplasia relies on the decades-long interplay between this oncobacterium and its gastric host. This review summarizes the functional and molecular regionalization of the stomach at homeostasis and details how chronic inflammation can lead to characteristic alterations in these developmental demarcations, both at the topographic and glandular levels. More importantly, this review illustrates our current understanding of the epithelial mechanisms that underlie the pre-malignant gastric landscape, how Hp adapts to and exploits these changes, and the clinical implications of identifying these changes in order to stratify patients at risk of developing gastric cancer, a leading cause of cancer-related deaths worldwide.
Collapse
Affiliation(s)
- José B Sáenz
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
222
|
Cho JH, Jin SY, Park S. Scoring model for discriminating gastric cancer risk in patients with negative serum pepsinogen and anti-Helicobacter pylori antibody results. J Gastroenterol Hepatol 2021; 36:3345-3353. [PMID: 34328237 DOI: 10.1111/jgh.15630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/07/2021] [Accepted: 07/16/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND The ABC test measures serum pepsinogen and anti-Helicobacter pylori IgG antibody levels to predict precancerous conditions in the stomach and gastric cancer. However, a limitation of this test is that the gastric cancer risk is not negligible in patients with a negative result. METHODS Based on their ABC results, 1157 patients were classified into Groups A (n = 392), B (n = 479), C (n = 247), and D (n = 39). In Group A, 24.2% of patients had atrophic gastritis and/or intestinal metaplasia and had thus been incorrectly assigned to Group A. Patients in Group A were then assigned to derivation (n = 236) and validation (n = 156) cohorts by 3:2 random sampling. Logistic regression analyses were performed to identify the factors discriminating between a correct (true) and incorrect (false) Group A classification. RESULTS A 4-point discriminative model was constructed based on a high-negative H. pylori IgG antibody titer and the patient's age (50-64 and ≥65 years). The areas under the receiver operating characteristic curve for the derivation and validation cohorts were 0.868 and 0.894, respectively. In the validation cohort, the addition of a discriminative model score ≥2 to the ABC method showed a similar accuracy for predicting gastric cancer risk compared with the ABC method alone (93.8% vs. 92.4%). CONCLUSION The 4-point discriminative model may help identify patients with a normal serological test who are nonetheless at risk of developing gastric cancer.
Collapse
Affiliation(s)
- Jun-Hyung Cho
- Digestive Disease Center, Soonchunhyang University Hospital, Seoul, South Korea
| | - So-Young Jin
- Department of Pathology, Soonchunhyang University Hospital, Seoul, South Korea
| | - Suyeon Park
- Department of Medical Biostatistics, Soonchunhyang University Hospital, Seoul, South Korea
| |
Collapse
|
223
|
Koustas E, Trifylli EM, Sarantis P, Kontolatis NI, Damaskos C, Garmpis N, Vallilas C, Garmpi A, Papavassiliou AG, Karamouzis MV. The Implication of Autophagy in Gastric Cancer Progression. Life (Basel) 2021; 11:1304. [PMID: 34947835 PMCID: PMC8705750 DOI: 10.3390/life11121304] [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: 10/30/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer is the fifth most common malignancy and the third leading cause of cancer-related death worldwide. The three entirely variable entities have distinct epidemiology, molecular characteristics, prognosis, and strategies for clinical management. However, many gastric tumors appear to be resistant to current chemotherapeutic agents. Moreover, a significant number of gastric cancer patients, with a lack of optimal treatment strategies, have reduced survival. In recent years, multiple research data have highlighted the importance of autophagy, an essential catabolic process of cytoplasmic component digestion, in cancer. The role of autophagy as a tumor suppressor or tumor promoter mechanism remains controversial. The multistep nature of the autophagy process offers a wide array of targetable points for designing novel chemotherapeutic strategies. The purpose of this review is to summarize the current knowledge regarding the interplay between gastric cancer development and the autophagy process and decipher the role of autophagy in this kind of cancer. A plethora of different agents that direct or indirect target autophagy may be a novel therapeutic approach for gastric cancer patients.
Collapse
Affiliation(s)
- Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Eleni-Myrto Trifylli
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Nikolaos I. Kontolatis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Christos Damaskos
- Renal Transplantation Unit, ‘Laiko’ General Hospital, 11527 Athens, Greece;
- ‘N.S. Christeas’ Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Garmpis
- ‘N.S. Christeas’ Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Second Department of Propedeutic Surgery, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Vallilas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Anna Garmpi
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-M.T.); (P.S.); (N.I.K.); (C.V.); (A.G.P.); (M.V.K.)
| |
Collapse
|
224
|
Duan S, Rico K, Merchant JL. Gastrin: From Physiology to Gastrointestinal Malignancies. FUNCTION (OXFORD, ENGLAND) 2021; 3:zqab062. [PMID: 35330921 PMCID: PMC8788842 DOI: 10.1093/function/zqab062] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023]
Abstract
Abetted by widespread usage of acid-suppressing proton pump inhibitors (PPIs), the mitogenic actions of the peptide hormone gastrin are being revisited as a recurring theme in various gastrointestinal (GI) malignancies. While pathological gastrin levels are intricately linked to hyperplasia of enterochromaffin-like cells leading to carcinoid development, the signaling effects exerted by gastrin on distinct cell types of the gastric mucosa are more nuanced. Indeed, mounting evidence suggests dichotomous roles for gastrin in both promoting and suppressing tumorigenesis. Here, we review the major upstream mediators of gastrin gene regulation, including inflammation secondary to Helicobacter pylori infection and the use of PPIs. We further explore the molecular biology of gastrin in GI malignancies, with particular emphasis on the regulation of gastrin in neuroendocrine neoplasms. Finally, we highlight tissue-specific transcriptional targets as an avenue for targetable therapeutics.
Collapse
Affiliation(s)
- Suzann Duan
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Karen Rico
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | | |
Collapse
|
225
|
Al Hennawi H, Khedr A, Maan RK, Albarazi M, Atluri P. Gastric Intestinal Metaplasia and Its Rapid Progression Toward Gastric Adenocarcinoma: A Call for Clear Patient Management and Awareness. Cureus 2021; 13:e18751. [PMID: 34796052 PMCID: PMC8589342 DOI: 10.7759/cureus.18751] [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] [Accepted: 10/12/2021] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related death worldwide. Helicobacter pylori (H. pylori) infection is known to cause gastric adenocarcinoma in a stepwise fashion. Gastric intestinal metaplasia is a known premalignant stage. We report a case of a 70-year-old male patient with active chronic H. pylori-associated gastritis and focal intestinal metaplasia on the initial presentation, who rapidly developed diffuse, poorly differentiated gastric adenocarcinoma 20 months after the loss to follow-up. Our case highlights the premalignant nature of gastric intestinal metaplasia (GIM) and the extreme importance of early eradication of H. pylori. We also address the lack of definitive GIM surveillance guidelines.
Collapse
Affiliation(s)
- Hussam Al Hennawi
- Internal Medicine, Alfaisal University College of Medicine, Riyadh, SAU
| | - Anwar Khedr
- Internal Medicine, Tanta University Faculty of Medicine, Tanta, EGY
| | | | | | - Purna Atluri
- Gastroenterology, University Hospital of Brooklyn, State University of New York Downstate Medical Center, Brooklyn, USA
| |
Collapse
|
226
|
Yu J, Song H, Ekheden I, Löhr M, Ploner A, Ye W. Gastric Mucosal Abnormality and Risk of Pancreatic Cancer: A Population-Based Gastric Biopsy Cohort Study in Sweden. Cancer Epidemiol Biomarkers Prev 2021; 30:2088-2095. [PMID: 34497088 PMCID: PMC9398138 DOI: 10.1158/1055-9965.epi-21-0580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/07/2021] [Accepted: 08/25/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND It remains open whether gastric precancerous lesions are associated with an elevated risk of pancreatic cancer. Our aim was to investigate the association between gastric mucosal status and pancreatic cancer risk. METHODS Patients with gastric biopsies [normal, minor changes, superficial gastritis, and atrophic gastritis/intestinal metaplasia/dysplasia (AG/IM/Dys)] from the Swedish histopathology registers during 1979 to 2011 were included. Cross-linkages with several nationwide registries allowed complete follow-up and identification of pancreatic cancer cases until 2014. Standardized incidence ratios (SIR) and HRs were estimated. RESULTS During 3,438,248 person-years of follow-up with 318,653 participants, 3,540 cases of pancreatic cancer were identified. The same pattern of excess risk of pancreatic cancer compared with the general population was observed across all groups: a peak of 12- to 21-fold excess risk in the first year after biopsy [e.g., normal: SIR = 17.4; 95% confidence interval (CI), 15.7-19.3; AG/IM/Dys: SIR = 11.5; 95% CI, 9.9-13.4], which dropped dramatically during the second and third years, followed by 20% to 30% increased risk after the third year (e.g., normal: SIR = 1.2; 95% CI, 1.1-1.4; AG/IM/Dys: SIR = 1.3; 95% CI, 1.1-1.5). However, no significant excess risk was observed with the normal gastric mucosa as reference. CONCLUSIONS This unique, large pathologic cohort study did not find evidence that abnormal gastric mucosal status is causally associated with a long-term pancreatic cancer risk. However, a highly increased short-term risk was observed for people undergoing gastroscopy with biopsy sampling compared with the general population. IMPACT Further studies for a long-term risk of pancreatic cancer in patients with gastric biopsies are needed, with further adjustments.
Collapse
Affiliation(s)
- Jingru Yu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Corresponding Authors: Jingru Yu, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm, Stockholm 17177, Sweden. E-mail: ; and Weimin Ye, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm, Stockholm 17177, Sweden. E-mail:
| | - Huan Song
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.,Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Isabella Ekheden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Upper Gastrointestinal Unit, Cancer Division, Karolinska University Hospital, Stockholm, Sweden
| | - Alexander Ploner
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Corresponding Authors: Jingru Yu, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm, Stockholm 17177, Sweden. E-mail: ; and Weimin Ye, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm, Stockholm 17177, Sweden. E-mail:
| |
Collapse
|
227
|
Jiang X, Zheng J, Liu L, Jiang K, Wen Y, Yan Y, Liu Y, Zhong L, Huang Y, Yao Z, Nie K, Zheng Z, Pan J, Liu P, Zhuang K, Liu F, Xu S, Li P. CXCR4 is a Novel Biomarker Correlated With Malignant Transformation and Immune Infiltrates in Gastric Precancerous Lesions. Front Mol Biosci 2021; 8:697993. [PMID: 34676245 PMCID: PMC8523893 DOI: 10.3389/fmolb.2021.697993] [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: 04/20/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background: As early gastric cancer (EGC) has a far better prognosis than advanced gastric cancer (GC), early diagnosis and treatment are essential. However, understanding the mechanism of the process from gastric precancerous lesion (GPL) becoming EGC has made little advances. Besides, biomarkers that can monitor the progression of GPL-to-GC are still much insufficient. Methods: Key gene modules associated with GPL progression to EGC were identified by integrating two GPL-related data sets, GSE55696 and GSE130823, using the WGCNA method. Combining with the TCGA-STAD cohort, hub genes were identified. Immunofluorescence was conducted to validate the expression. To explore the implication of hub genes in GPL malignant transformation, a correlation test was conducted to identify their co-expression genes, co-expression cytokines, and co-expression immune cells. Least absolute shrinkage and selection operator (LASSO) Cox regression was applied to shrink CXCR4-related predictors and construct a prognostic model. Functional enrichment was applied for exploring the potential mechanism. Results: The green module in GSE55696 and the yellow module in GSE130823 were regarded as key gene modules associated with GPL progression to EGC, and 219 intersection genes from them were mainly enriched in critical immune biological processes. Combining with the TCGA-STAD cohort, CXCR4 was identified as a novel biomarker correlated with the malignant transformation of GPL, the positive rate of which was increased with GPL progression according to immunofluorescence. CXCR4 co-expression genes were found mainly involved in regulation of actin. CXCR4 co-expression cytokines were enriched in regulation of chemotaxis, cell chemotaxis, mononuclear cell migration, leukocyte chemotaxis, etc. As for co-expression immune cells, the expression level of CXCR4 was positively correlated with the abundance of macrophages but negatively correlated with that of effector memory T cells and NKT cells during GPL malignant transformation. In addition, the CXCR4-related prognostic model was able to predict the prognosis of GC and serve as an independent predictor for overall survival (OS). Conclusions: CXCR4 was a novel biomarker correlated with malignant transformation of GPL and played a vital role in the control of tumor immunity. CXCR4 is possible to serve as a therapeutic target for malignant transformation of GPL.
Collapse
Affiliation(s)
- Xiaotao Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junhui Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanxing Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kailin Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Wen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhua Yan
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yufeng Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yuancheng Huang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhengyang Yao
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kechao Nie
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhihua Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinglin Pan
- Department of Gastroenterology, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, China
| | - Peng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kunhai Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengbin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shijie Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiwu Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
228
|
Fiuza F, Maluf-Filho F, Ide E, Furuya Jr CK, Fylyk SN, Ruas JN, Stabach L, Araujo GA, Matuguma SE, Uemura RS, Sakai CM, Yamazaki K, Ueda SS, Sakai P, Martins BC. Association between mucosal surface pattern under near focus technology and Helicobacter pylori infection. World J Gastrointest Endosc 2021; 13:518-528. [PMID: 34733412 PMCID: PMC8546563 DOI: 10.4253/wjge.v13.i10.518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/21/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Many studies evaluated magnification endoscopy (ME) to correlate changes on the gastric mucosal surface with Helicobacter pylori (H. pylori) infection. However, few studies validated these concepts with high-definition endoscopy without ME.
AIM To access the association between mucosal surface pattern under near focus technology and H. pylori infection status in a western population.
METHODS Cross-sectional study including all patients referred to routine upper endoscopy. Endoscopic exams were performed using standard high definition (S-HD) followed by near focus (NF-HD) examination. Presence of erythema, erosion, atrophy, and nodularity were recorded during S-HD, and surface mucosal pattern was classified using NF-HD in the gastric body. Biopsies were taken for rapid urease test and histology.
RESULTS One hundred and eighty-seven patients were analyzed from August to November 2019. Of those, 47 (25.1%) were H. pylori+, and 42 (22.5%) had a previous H. pylori treatment. In the examination with S-HD, erythema had the best sensitivity for H. pylori detection (80.9%). Exudate (99.3%), nodularity (97.1%), and atrophy (95.7%) demonstrated better specificity values, but with low sensitivity (6.4%-19.1%). On the other hand, the absence of erythema was strongly associated with H. pylori- (negative predictive value = 92%). With NF-HD, 56.2% of patients presented type 1 pattern (regular arrangement of collecting venules, RAC), and only 5.7% of RAC+ patients were H. pylori+. The loss of RAC presented 87.2% sensitivity for H. pylori detection, 70.7% specificity, 50% positive predictive value, and 94.3% negative predictive value, indicating that loss of RAC was suboptimal to confirm H. pylori infection, but when RAC was seen, H. pylori infection was unlikely.
CONCLUSION The presence of RAC at the NF-HD exam and the absence of erythema at S-HD were highly predictive of H. pylori negative status. On the other hand, the loss of RAC had a suboptimal correlation with the presence of H. pylori.
Collapse
Affiliation(s)
- Felipe Fiuza
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | - Fauze Maluf-Filho
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | - Edson Ide
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | | | - Sonia Nadia Fylyk
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | | | - Luciana Stabach
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | | | - Sergio Eiji Matuguma
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | - Ricardo Sato Uemura
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | | | - Kendi Yamazaki
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | | | - Paulo Sakai
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| | - Bruno Costa Martins
- Department of Endoscopy, Hospital Alemão Oswaldo Cruz, São Paulo 01323-020, Brazil
| |
Collapse
|
229
|
Cullin N, Azevedo Antunes C, Straussman R, Stein-Thoeringer CK, Elinav E. Microbiome and cancer. Cancer Cell 2021; 39:1317-1341. [PMID: 34506740 DOI: 10.1016/j.ccell.2021.08.006] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022]
Abstract
The human microbiome constitutes a complex multikingdom community that symbiotically interacts with the host across multiple body sites. Host-microbiome interactions impact multiple physiological processes and a variety of multifactorial disease conditions. In the past decade, microbiome communities have been suggested to influence the development, progression, metastasis formation, and treatment response of multiple cancer types. While causal evidence of microbial impacts on cancer biology is only beginning to be unraveled, enhanced molecular understanding of such cancer-modulating interactions and impacts on cancer treatment are considered of major scientific importance and clinical relevance. In this review, we describe the molecular pathogenic mechanisms shared throughout microbial niches that contribute to the initiation and progression of cancer. We highlight advances, limitations, challenges, and prospects in understanding how the microbiome may causally impact cancer and its treatment responsiveness, and how microorganisms or their secreted bioactive metabolites may be potentially harnessed and targeted as precision cancer therapeutics.
Collapse
Affiliation(s)
- Nyssa Cullin
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Camila Azevedo Antunes
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, 7610001 Rehovot, Israel
| | - Christoph K Stein-Thoeringer
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Immunology, Weizmann Institute of Science, 234 Herzl Street, 7610001 Rehovot, Israel.
| |
Collapse
|
230
|
Relationship of prognostic factors in stomach cancer with helicobacter pylori: a retrospective study. Acta Gastroenterol Belg 2021; 84:607-617. [PMID: 34965043 DOI: 10.51821/84.4.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND STUDY AIMS The prognostic value of H. pylori, which infects more than half of the human population living in the world and plays a role in gastric cancer pathogenesis, is controversial. Our aim is to investigate the relationship between H. pylori and prognostic factors in gastric cancer. PATIENTS AND METHODS The data of 110 patients (38 females and 72 males) that underwent surgeries due to gastric cancer between 2014 and 2017 were retrospectively analyzed. The relationships between survival (disease-free and overall) and factors such as p53, HER2/neu, Ki-67, neutrophil and platelet lymphocyte ratio (NLR / PLR), histopathological and demographic characteristics were examined. In addition, the results of H. pylori positive and negative groups were compared. RESULTS Sixty-one (55%) patients were H. pylori negative and 49 (45%) were positive. In multivariate analysis, TNM stage, lymph node capsule invasion and NLR were determined as independent prognostic factors in both disease-free and overall survival. Age>62 and PLR>14.3 were determined as independent predictive factors of poor prognosis in overall survival. In univariate analysis, tumor diameter of >4.3 cm, lymphovascular and perineural invasion, and diffuse p53 expression were determined as predictive factors of poor prognosis in disease-free and overall survival. The effectiveness of these markers in prognosis was not different between H. pylori negative and positive groups. CONCLUSION While age, tumor diameter, TNM stage, lymph node capsule invasion, perineural and lymphovascular invasion, diffuse p53, PLR, and NLR were determined as prognostic factors in gastric cancer, these factors were not affected by the presence of H. pylori.
Collapse
|
231
|
Molecular Landscapes of Gastric Pre-Neoplastic and Pre-Invasive Lesions. Int J Mol Sci 2021; 22:ijms22189950. [PMID: 34576114 PMCID: PMC8468646 DOI: 10.3390/ijms22189950] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) represents one of the most common and most lethal malignancies worldwide. The histopathological characterization of GC precursor lesions has provided great knowledge about gastric carcinogenesis, with the consequent introduction of effective strategies of primary and secondary prevention. In recent years, a large amount of data about the molecular events in GC development is emerging, flanking the histomorphological descriptions. In this review, we describe the landscape of molecular alterations in gastric pre-invasive lesions with a glance at their potential use in the diagnostic and therapeutic decision-making process.
Collapse
|
232
|
Gastric Microbiota in a Low-Helicobacter pylori Prevalence General Population and Their Associations With Gastric Lesions. Clin Transl Gastroenterol 2021; 11:e00191. [PMID: 32764211 PMCID: PMC7431247 DOI: 10.14309/ctg.0000000000000191] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION: Non–Helicobacter pylori microbiota might account for some cases with unexplained chronic gastritis that may in a minority eventually progress to gastric cancer through the Correa cascade. We characterized gastric microbiota by describing the normal stomach, compared it with early precancerous lesions and other disease states, and assessed whether H. pylori status affects bacterial diversity. METHODS: In a population-based study of those with and without gastrointestinal symptoms, cytology brush samples were collected during endoscopy from 316 individuals. Mucosal status was classified as normal mucosa (171), nonatrophic H. pylori gastritis (33), atrophic gastritis (12), or antral chemical gastritis (61). The 16S rRNA gene sequencing and analysis were performed to characterize the microbiota. RESULTS: Microbiota in atrophic gastritis and nonatrophic H. pylori gastritis stomachs were dysbiotic and differed from those in the normal stomach (P = 0.001). The normal stomach had the highest microbial diversity, followed by antral chemical gastritis. The atrophic gastritis and chronic H. pylori gastritis groups had the lowest diversity, a difference that was statistically significant (P = 0.01). Besides H. pylori, non–H. pylori bacteria accounted for group differences. Microbial network analysis showed that the normal group network was most highly connected, whereas the H. pylori gastritis group had the lowest connection. We found an increasing positive co-occurrence of oral bacteria in the stomach because samples deviated from the normal network, some of which were pathogens. The H. pylori–negative group had the highest microbial diversity (Shannon index) compared with the H. pylori–positive group (P = 0.001). DISCUSSION: In this low–H. pylori prevalence general population, the gastric mucosal microbiota of the normal stomach differed significantly from those with nonatrophic or atrophic gastritis. There was an increasing abundance of pathogenic bacteria from the normal state to early precancerous states.
Collapse
|
233
|
Park JM, Han YM, Hahm KB. Rejuvenation of Helicobacter pylori-Associated Atrophic Gastritis Through Concerted Actions of Placenta-Derived Mesenchymal Stem Cells Prevented Gastric Cancer. Front Pharmacol 2021; 12:675443. [PMID: 34483897 PMCID: PMC8416416 DOI: 10.3389/fphar.2021.675443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/22/2021] [Indexed: 01/06/2023] Open
Abstract
Chronic Helicobacter pylori infection causes gastric cancer via the progression of precancerous chronic atrophic gastritis (CAG). Therefore, repairing gastric atrophy could be a useful strategy in preventing H. pylori-associated gastric carcinogenesis. Although eradication of the bacterial pathogen offers one solution to this association, this study was designed to evaluate an alternative approach using mesenchymal stem cells to treat CAG and prevent carcinogenesis. Here, we used human placenta-derived mesenchymal stem cells (PD-MSCs) and their conditioned medium (CM) to treat H. pylori-associated CAG in a mice/cell model to explore their therapeutic effects and elucidate their molecular mechanisms. We compared the changes in the fecal microbiomes in response to PD-MSC treatments, and chronic H. pylori-infected mice were given ten treatments with PD-MSCs before being sacrificed for end point assays at around 36 weeks of age. These animals presented with significant reductions in the mean body weights of the control group, which were eradicated following PD-MSC treatment (p < 0.01). Significant changes in various pathological parameters including inflammation, gastric atrophy, erosions/ulcers, and dysplastic changes were noted in the control group (p < 0.01), but these were all significantly reduced in the PD-MSC/CM-treated groups. Lgr5+, Ki-67, H+/K+-ATPase, and Musashi-1 expressions were all significantly increased in the treated animals, while inflammatory mediators, MMP, and apoptotic executors were significantly decreased in the PD-MSC group compared to the control group (p < 0.001). Our model showed that H. pylori-initiated, high-salt diet-promoted gastric atrophic gastritis resulted in significant changes in the fecal microbiome at the phylum/genus level and that PD-MSC/CM interventions facilitated a return to more normal microbial communities. In conclusion, administration of PD-MSCs or their conditioned medium may present a novel rejuvenating agent in preventing the progression of H. pylori-associated premalignant lesions.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daejeon, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Seoul, Korea.,CHA Cancer Preventive Research Center, CHA Bio Complex, Seongnam, Korea
| |
Collapse
|
234
|
Danis N, Ozkan AE, Karatas F, Cakici C, Yigitbasi T, Sevencan NO, Kayhan B. The importance of antioxidant status in gastric intestinal metaplasia. J Med Biochem 2021; 40:378-383. [PMID: 34616227 PMCID: PMC8451221 DOI: 10.5937/jomb0-29349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Oxidative stress status in different cancer types was investigated before, but not studied in gastric intestinal metaplasia to the best of our knowledge. Purpose of this study is to examine whether there is a difference between oxidative stress status in patients with intestinal metaplasia (IM) compared to individuals without IM, we compared the serum levels of disulfide (SS), total thiol (TT) and native thiol (NT). METHODS This was a prospective, non-randomized casecontrol study including 67 patients with histopathologically confirmed IM and 60 individuals demographically matched in terms of age, gender, BMI, smoking status, and chronic diseases as control group. RESULTS The mean NT, TT and NT to TT (NT/TT) ratios were statistically significantly higher in IM group compared to controls ((351.71 ± 81.9 mol/L vs. 271.82 ± 54.13 mol/L, p=0.000), (391.5 ± 92.69 mol/L vs. 308.59 ± 55.53 mol/L, p=0.000) and (0.89 ± 0.6 vs. 0.87 ± 0.29, p=0.022), respectively). The mean SS to TT (SS/TT) ratio was significantly lower in IM group than control group (0.050 ± 0.31 vs. 0.060 ± 0.014, P=0.022). Median SS and mean SS/NT ratio was similar in both groups (16.3 (3.3-78) vs. 18.3 (10-32.7), p=0.271 and 0.055 ± 0.041 vs. 0.070 ± 0.019, p=0.068, respectively). In ROC analysis, cut off value of SS/NT for IM was found 0.062, in regression analysis, SS/NT <0.062 was found as an independently prognostic marker for IM (OR, 2.38; 95%CI: 1.168-4.865, P=0.017). CONCLUSIONS SS/NT ratio lower than 0.062 was found as an independently prognostic marker for IM. This ratio could help to distinguish which patients should be followed closely for gastric cancer.
Collapse
Affiliation(s)
- Nilay Danis
- Karabuk University Education and Research Hospital, Gastroenterology, Karabuk, Turkey
| | - Aysegul Ertinmaz Ozkan
- Karabuk University Education and Research Hospital, Department of Internal Madicine, Karabuk, Turkey
| | - Fatih Karatas
- Karabuk University Education and Research Hospital, Medical Oncology, Karabuk, Turkey
| | - Cagri Cakici
- Medipol University, Regenerative and Restorative Medicine Research Center, Istanbul, Turkey
| | - Turkan Yigitbasi
- Medipol University, Regenerative and Restorative Medicine Research Center, Istanbul, Turkey
| | - Nurhayat Ozkan Sevencan
- Karabuk University Education and Research Hospital, Department of Internal Madicine, Karabuk, Turkey
| | - Burcak Kayhan
- Karabuk University Education and Research Hospital, Department of Internal Madicine, Karabuk, Turkey
| |
Collapse
|
235
|
Wang S, Ye F, Sheng Y, Yu W, Liu Y, Liu D, Zhang K. Development and Validation of Nomograms to Predict Operative Link for Gastritis Assessment Any-Stage and Stages III-IV in the Chinese High-Risk Gastric Cancer Population. Front Med (Lausanne) 2021; 8:724566. [PMID: 34447771 PMCID: PMC8383045 DOI: 10.3389/fmed.2021.724566] [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: 06/13/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose: It is very essential to diagnose gastric atrophy in the area with high prevalence of gastric cancer. Operative link for gastritis assessment (OLGA) was developed to detect the severity of gastric atrophy. The aim of this study was to develop and validate nomograms for predicting OLGA any-stage and stages III-IV in the Chinese high-risk gastric cancer population. Methods: We retrospectively analyzed 7,945 participants obtained by a multicenter cross-sectional study. We randomly selected 55% individuals (4,370 participants, training cohort) to analyze and generate the prediction models and validated the models on the remaining individuals (3,575 participants, validation cohort). A multivariate logistic regression model was used to select variables in the training cohort. The corresponding nomograms were developed to predict OLGA any-stage and stages III-IV, respectively. The area under the receiver operating characteristic curves and the GiViTI calibration belts were used to estimate the discrimination and calibration of the prediction models. Results: There were 1,226 (28.05%) participants in the training sample and 970 (27.13%) in the validation sample who were diagnosed with gastric atrophy. The nomogram predicting OLGA any-stage had an area under the curve (AUC) of 0.610 for the training sample and 0.615 for the validation sample, with favorable calibrations in the overall population. Similarly, the nomogram predicting OLGA stages III-IV had an AUC of 0.702 and 0.714 for the training and validation samples, respectively, with favorable calibrations in the overall population. Conclusions: The prediction model can early identify the occurrence of gastric atrophy and the severity stage of gastric atrophy to some extent.
Collapse
Affiliation(s)
- Song Wang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fei Ye
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuan Sheng
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenyong Yu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yingling Liu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dehua Liu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaiguang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
236
|
Mirbagheri SZ, Bakhtiari R, Fakhre Yaseri H, Rahimi Foroushani A, Eshraghi SS, Alebouyeh M. Transcriptional alteration of genes linked to gastritis concerning Helicobacter pylori infection status and its virulence factors. Mol Biol Rep 2021; 48:6481-6489. [PMID: 34427890 DOI: 10.1007/s11033-021-06654-w] [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: 04/24/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Helicobacter pylori infection and heterogeneity in its pathogenesis could describe diversity in the expression of inflammatory genes in the gastric tissue. We aimed to investigate transcriptional alteration of genes linked to gastritis concerning the H. pylori infection status and its virulence factors. METHODS AND RESULTS Biopsy samples of 12 infected and 12 non-infected patients with H. pylori that showed moderate chronic gastritis were selected for transcriptional analysis. Genotyping of H. pylori strains was done using PCR and relative expression of inflammatory genes was compared between the infected and non-infected patients using relative quantitative real-time PCR. Positive correlations between transcriptional changes of IL8 with TNF-α and Noxo1 in the infected and TNF-α with Noxo1, MMP7, and Atp4A in the non-infected patients were detected. Six distinct genotypes of H. pylori were detected that showed no correlation with gender, ethnicity, age, endoscopic findings, and transcriptional levels of host genes. Irrespective of the characterized genotypes, our results showed overexpression of TNF-α, MMP7, Noxo1, and ATP4A in the infected and IL-8, Noxo1, and ATP4A in the non-infected patients. CONCLUSIONS A complexity in transcription of genes respective to the characterized H. pylori genotypes in the infected patients was detected in our study. The observed difference in co-regulation of genes linked to gastritis in the infected and non-infected patients proposed involvement of different regulatory pathways in the inflammation of the gastric tissue in the studied groups.
Collapse
Affiliation(s)
- Seyedeh Zohre Mirbagheri
- Department of Pathobiology, School of Public Health and Institute Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Ronak Bakhtiari
- Department of Pathobiology, School of Public Health and Institute Health Research, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hashem Fakhre Yaseri
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.,Gastroenterology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Rahimi Foroushani
- Department of Epidemiology and Biostatistics, School of Public Health and Institute Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Saeed Eshraghi
- Department of Pathobiology, School of Public Health and Institute Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Alebouyeh
- Pediatric Infections Research Centre, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
237
|
Pennelli G, Grillo F, Galuppini F, Ingravallo G, Pilozzi E, Rugge M, Fiocca R, Fassan M, Mastracci L. Gastritis: update on etiological features and histological practical approach. Pathologica 2021; 112:153-165. [PMID: 33179619 PMCID: PMC7931571 DOI: 10.32074/1591-951x-163] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric biopsies represent one of the most frequent specimens that the pathologist faces in routine activity. In the last decade or so, the landscape of gastric pathology has been changing with a significant and constant decline of H. pylori-related pathologies in Western countries coupled with the expansion of iatrogenic lesions due to the use of next-generation drugs in the oncological setting. This overview will focus on the description of the elementary lesions observed in gastric biopsies and on the most recent published recommendations, guidelines and expert opinions.
Collapse
Affiliation(s)
- Gianmaria Pennelli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| | - Federica Grillo
- Anatomic Pathology, San Martino IRCCS Hospital, Genova, Italy.,Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Organ Transplantation, Section of Pathological Anatomy, University of Bari Aldo Moro, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, UOC Anatomic Pathology, Sant'Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Massimo Rugge
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy.,Veneto Tumor Registry (RTV), Veneto Regional Authority, Padua, Italy
| | - Roberto Fiocca
- Anatomic Pathology, San Martino IRCCS Hospital, Genova, Italy.,Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Italy
| | - Luca Mastracci
- Anatomic Pathology, San Martino IRCCS Hospital, Genova, Italy.,Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| |
Collapse
|
238
|
Chen G, Xu R, Yue B, Jia M, Li P, Ji M, Zhang S. A Parallel Comparison Method of Early Gastric Cancer: The Light Transmission-Assisted Pathological Examination of Specimens of Endoscopic Submucosal Dissection. Front Oncol 2021; 11:705418. [PMID: 34414114 PMCID: PMC8370090 DOI: 10.3389/fonc.2021.705418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022] Open
Abstract
Objective It is always challenging to diagnose and characterize early gastric cancer surrounded by non-cancerous mucosa, including the malignant diagnosis and extent and depth of the lesions. Therefore, we developed a light transmission-assisted pathological examination to diagnose and characterize early gastric cancer. Here, we performed a parallel comparison between the light transmission-assisted pathological examination under endoscopy and the histological examination for the diagnosis of early gastric cancer. Methods First, the endoscopic submucosal dissection (ESD) specimen was first placed on the surface of the light-emitting diode lamp to observe the mucosal surface structure and blood vessels. Second, the sliced and embedded tissue strips were cut into 3-µm sections for hematoxylin and eosin staining. Third, the histopathology of each section was projected onto a macroscopic image. Finally, the macroscopic and microscopic changes in the ESD specimens observed under endoscopy were compared. Seventy cases of early gastric adenocarcinoma were diagnosed and characterized using this new method. Results Using the conventional pathological method, the demarcation line of the lesions was seen in 40 of 70 (57.1%) cases. Furthermore, no surface structure or microvascular changes were observed in any of the cases. Based on the light transmission-assisted pathological examination, 58 of 70 (82.9%) cases presented clear edges of neoplastic and non-neoplastic epithelia, with a classifiable surface structure (88.6%) and microvascular type (78.8%). Conclusions This pilot method provided a practical bridge between endoscopic and pathological examinations. Compared to the histological examination, the light transmission-assisted pathological examination was an easier and more precise way to match the in vivo endoscopic observation and in vitro pathological examination.
Collapse
Affiliation(s)
- Guangyong Chen
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rui Xu
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bing Yue
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mei Jia
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Peng Li
- Department of Gastroenterology, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ming Ji
- Department of Gastroenterology, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shutian Zhang
- Department of Gastroenterology, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
239
|
Young E, Philpott H, Singh R. Endoscopic diagnosis and treatment of gastric dysplasia and early cancer: Current evidence and what the future may hold. World J Gastroenterol 2021; 27:5126-5151. [PMID: 34497440 PMCID: PMC8384753 DOI: 10.3748/wjg.v27.i31.5126] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/07/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer accounts for a significant proportion of worldwide cancer-related morbidity and mortality. The well documented precancerous cascade provides an opportunity for clinicians to detect and treat gastric cancers at an endoscopically curable stage. In high prevalence regions such as Japan and Korea, this has led to the implementation of population screening programs. However, guidelines remain ambiguous in lower prevalence regions. In recent years, there have been many advances in the endoscopic diagnosis and treatment of early gastric cancer and precancerous lesions. More advanced endoscopic imaging has led to improved detection and characterization of gastric lesions as well as superior accuracy for delineation of margins prior to resection. In addition, promising early data on artificial intelligence in gastroscopy suggests a future role for this technology in maximizing the yield of advanced endoscopic imaging. Data on endoscopic resection (ER) are particularly robust in Japan and Korea, with high rates of curative ER and markedly reduced procedural morbidity. However, there is a shortage of data in other regions to support the applicability of protocols from these high prevalence countries. Future advances in endoscopic therapeutics will likely lead to further expansion of the current indications for ER, as both technology and proceduralist expertise continue to grow.
Collapse
Affiliation(s)
- Edward Young
- Department of Gastroenterology, Lyell McEwin Hospital, Elizabeth Vale 5112, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, SA, Australia
| | - Hamish Philpott
- Department of Gastroenterology, Lyell McEwin Hospital, Elizabeth Vale 5112, SA, Australia
| | - Rajvinder Singh
- Department of Gastroenterology, Lyell McEwin Hospital, Elizabeth Vale 5112, SA, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, SA, Australia
| |
Collapse
|
240
|
Varon C, Azzi-Martin L, Khalid S, Seeneevassen L, Ménard A, Spuul P. Helicobacters and cancer, not only gastric cancer? Semin Cancer Biol 2021; 86:1138-1154. [PMID: 34425210 DOI: 10.1016/j.semcancer.2021.08.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The Helicobacter genus actually comprises 46 validly published species divided into two main clades: gastric and enterohepatic Helicobacters. These bacteria colonize alternative sites of the digestive system in animals and humans, and contribute to inflammation and cancers. In humans, Helicobacter infection is mainly related to H. pylori, a gastric pathogen infecting more than half of the world's population, leading to chronic inflammation of the gastric mucosa that can evolve into two types of gastric cancers: gastric adenocarcinomas and gastric MALT lymphoma. In addition, H. pylori but also non-H. pylori Helicobacter infection has been associated with many extra-gastric malignancies. This review focuses on H. pylori and its role in gastric cancers and extra-gastric diseases, as well as malignancies induced by non-H. pylori Helicobacters. Their different virulence factors and their involvement in carcinogenesis is discussed. This review highlights the importance of both gastric and enterohepatic Helicobacters in gastrointestinal and liver cancers.
Collapse
Affiliation(s)
- Christine Varon
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Lamia Azzi-Martin
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France; Univ. Bordeaux, UFR des Sciences Médicales, Bordeaux, France
| | - Sadia Khalid
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia
| | - Lornella Seeneevassen
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Armelle Ménard
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Pirjo Spuul
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia.
| |
Collapse
|
241
|
Gu Z, Ling J, Cong J, Li D. A Review of Therapeutic Effects and the Pharmacological Molecular Mechanisms of Chinese Medicine Weifuchun in Treating Precancerous Gastric Conditions. Integr Cancer Ther 2021; 19:1534735420953215. [PMID: 32865036 PMCID: PMC7466872 DOI: 10.1177/1534735420953215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Patients with precancerous gastric conditions are at a high risk for gastric carcinoma. The Chinese medicine Weifuchun (WFC) is used in treating chronic superficial gastritis and in postoperative adjuvant treatment of gastric cancer. Both monotherapy and combination therapy of WFC with other drugs can result in a favorable therapeutic outcome. WFC can dramatically improve clinical outcomes in patients with gastric precancerous lesions by targeting multiple pathways including pathways involved in the pharmacological action of Radix Ginseng Rubra (red ginseng), Rabdosia amethystoides, and fried Fructus Aurantii, including regulation of NF-κB, RUNX3/TGF-beta/Smad, Hedgehog (Hh) and Wnt signaling pathways, modulation of the expression of oncogenes and tumor suppressor genes, and indirect inhibition of Helicobacter pylori (Hp) by maintaining gastric microbial ecosystem. In this review, we will discuss the clinical efficacy and therapeutic regimen of WFC for gastric precancerous lesions and the molecular mechanisms involved. This review will highlight WFC-based therapeutic strategies in disrupting progress to gastric cancer and provide more information on the pharmacological mechanisms of WFC and its clinical application for the treatment of precancerous gastric lesions.
Collapse
Affiliation(s)
- Zhijian Gu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghong Ling
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Cong
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Li
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
242
|
Hao Y, Zhang R, Morris R, Cheng F, Zhu Z, Xu Y, Wang Y. Metabolome and microbiome alterations in tongue coating of gastric precancerous lesion patients. Expert Rev Gastroenterol Hepatol 2021; 15:949-963. [PMID: 33252275 DOI: 10.1080/17474124.2021.1850259] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: This paper seeks to provide mechanistic insight into the pathological transition through the analysis of metabolites and microorganisms in the tongue coating of gastric precancerous lesions (GPL) patients.Methods: GC-TOF-MS and UHPLC-QE-MS metabolomics, combined with 16S rRNA microbiome techniques, were performed to explore the changes in metabolites and microorganisms in the tongue coating of GPL patients.Results: When compared with 15 controls, 133 metabolites were found to be differentially expressed in 60 GPL cases, of which could be divided into ten categories. Among them, most of the differentially expressed metabolites identified were lipids or lipid-like molecules. These metabolites were implicated in 6 metabolic pathways including glycine, serine and threonine metabolism, arginine and proline metabolism, sphingolipid metabolism, valine, leucine and isoleucine degradation, arachidonic acid metabolism, and tyrosine metabolism. The relative abundances of Alloprevotella, Solobacterium, Rothia, Eikenella, and Aggregatibacter in the GPL group increased significantly relative to the controls and were associated with lipids and lipid-like molecules, organic nitrogen compounds, organic oxygen compounds, phenylpropanoids and polyketides, and organoheterocyclic compounds, respectively.Conclusions: Compared with healthy people, the changes of tongue coating metabolites in GPL patients were mainly characterized by alterations in lipid metabolism and were associated with localized changes in the microbiome.
Collapse
Affiliation(s)
- Yiming Hao
- Shanghai Key Laboratory of Health Identification and Assessment/Laboratory of TCM Four Diagnostic Information, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Renling Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Robert Morris
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Feng Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Zhujing Zhu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifeng Xu
- Shanghai Key Laboratory of Health Identification and Assessment/Laboratory of TCM Four Diagnostic Information, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqin Wang
- Shanghai Key Laboratory of Health Identification and Assessment/Laboratory of TCM Four Diagnostic Information, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
243
|
Doohan D, Fauzia KA, Rathnayake J, Lamawansa MD, Waskito LA, Tuan VP, Dashdorj A, Kabamba ET, Phuc BH, Ansari S, Akada J, Matsumoto T, Uchida T, Matsuhisa T, Yamaoka Y. Pepsinogen and Serum IgG Detection Is a Valuable Diagnostic Method for Helicobacter pylori Infection in a Low-Prevalence Country: A Report from Sri Lanka. Diagnostics (Basel) 2021; 11:1364. [PMID: 34441303 PMCID: PMC8391933 DOI: 10.3390/diagnostics11081364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022] Open
Abstract
The use of serum anti-Helicobacter pylori IgG and pepsinogen (PG) detection as a diagnostic method was evaluated in Sri Lanka. Gastric biopsies were performed (353 patients), and the prevalence of H. pylori infection was 1.7% (culture) and 2.0% (histology). IgG serology testing showed an area under the curve (AUC) of 0.922 (cut-off, 2.95 U/mL; specificity, 91.56%; sensitivity, 88.89%). Histological evaluation showed mild atrophy (34.3%), moderate atrophy (1.7%), metaplasia (1.7%), chronic gastritis (6.2%), and normal tissue (56%). The PGI/PGII ratio was significantly higher in H. pylori-negative patients (p < 0.01). PGII and PGI/PGII levels were lower in patients with metaplasia than in those with normal mucosa (p = 0.049 and p < 0.001, respectively). The PGI/PGII ratio best discriminated metaplasia and moderate atrophy (AUC 0.88 and 0.76, respectively). PGI and PGII alone showed poor discriminative ability, especially in mild atrophy (0.55 and 0.53, respectively) and chronic gastritis (0.55 and 0.53, respectively). The best cut-off to discriminate metaplasia was 3.25 U/mL (95.19% specificity, 83.33% sensitivity). Anti-H. pylori IgG and PG assessment (ABC method) was performed (group B, 2.0%; group A, 92.1%). The new cut-off more accurately identified patients with metaplasia requiring follow-up (group B, 5.4%). Assessment of anti-H. pylori IgG and PG is valuable in countries with a low prevalence of H. pylori infection.
Collapse
Grants
- DK62813, Ministry of Education, Culture, Sports, Science and Technology
- 221S0002 Ministry of Education, Culture, Sports, Science and Technology
- 16H06279 Ministry of Education, Culture, Sports, Science and Technology
- 18KK0266 Ministry of Education, Culture, Sports, Science and Technology
- 18K16182 Ministry of Education, Culture, Sports, Science and Technology
- 17K09353 Ministry of Education, Culture, Sports, Science and Technology
- Young Researcher Overseas Visits Japan Society for the Promotion of Science
- Strategic Funds Japan Science and Technology Agency
Collapse
Affiliation(s)
- Dalla Doohan
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Kartika Afrida Fauzia
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
- Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Jeewantha Rathnayake
- Department of Surgery, Teaching Hospital Peradeniya, University of Peradeniya, Kandy 20404, Sri Lanka; (J.R.); (M.D.L.)
| | - Meegahalande Durage Lamawansa
- Department of Surgery, Teaching Hospital Peradeniya, University of Peradeniya, Kandy 20404, Sri Lanka; (J.R.); (M.D.L.)
| | - Langgeng Agung Waskito
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Vo Phuoc Tuan
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Department of Endoscopy, Cho Ray Hospital, Ho Chi Minh 749000, Vietnam
| | - Azzaya Dashdorj
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
| | - Evariste Tshibangu Kabamba
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Research Center for Infectious Sciences, Department of Parasitology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
| | - Bui Hoang Phuc
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
| | - Shamshul Ansari
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Department of Microbiology, Teaching Hospital, Chitwan Medical College, Bharatpur 44200, Nepal
| | - Junko Akada
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
| | - Takashi Matsumoto
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
| | - Tomohisa Uchida
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan;
| | - Takeshi Matsuhisa
- Department of Gastroenterology, Tama Nagayama University Hospital, Nippon Medical School, Tokyo 206-8512, Japan;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Japan; (D.D.); (K.A.F.); (L.A.W.); (V.P.T.); (A.D.); (E.T.K.); (B.H.P.); (S.A.); (J.A.); (T.M.)
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
- Global Oita Medical Advanced Research Center for Health (GO-MARCH), Faculty of Medicine, Oita University, Yufu 879-5593, Japan
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
244
|
Tokat M, van Tilburg L, Koch AD, Spaander MCW. Artificial Intelligence in Upper Gastrointestinal Endoscopy. Dig Dis 2021; 40:395-408. [PMID: 34348267 DOI: 10.1159/000518232] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/23/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Over the past decade, several artificial intelligence (AI) systems are developed to assist in endoscopic assessment of (pre-)cancerous lesions of the gastrointestinal (GI) tract. In this review, we aimed to provide an overview of the possible indications of AI technology in upper GI endoscopy and hypothesize about potential challenges for its use in clinical practice. SUMMARY Application of AI in upper GI endoscopy has been investigated for several indications: (1) detection, characterization, and delineation of esophageal and gastric cancer (GC) and their premalignant conditions; (2) prediction of tumor invasion; and (3) detection of Helicobacter pylori. AI systems show promising results with an accuracy of up to 99% for the detection of superficial and advanced upper GI cancers. AI outperformed trainee and experienced endoscopists for the detection of esophageal lesions and atrophic gastritis. For GC, AI outperformed mid-level and trainee endoscopists but not expert endoscopists. KEY MESSAGES Application of artificial intelligence (AI) in upper gastrointestinal endoscopy may improve early diagnosis of esophageal and gastric cancer and may enable endoscopists to better identify patients eligible for endoscopic resection. The benefit of AI on the quality of upper endoscopy still needs to be demonstrated, while prospective trials are needed to confirm accuracy and feasibility during real-time daily endoscopy.
Collapse
Affiliation(s)
- Meltem Tokat
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Laurelle van Tilburg
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Arjun D Koch
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
245
|
Effects of orally administered Euglena gracilis and its reserve polysaccharide, paramylon, on gastric dysplasia in A4gnt knockout mice. Sci Rep 2021; 11:13640. [PMID: 34210998 PMCID: PMC8249615 DOI: 10.1038/s41598-021-92013-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 05/24/2021] [Indexed: 12/28/2022] Open
Abstract
Euglena gracilis is widely utilized as food or supplement to promote human and animal health, as it contains rich nutrients. In this study, we administered spray-dried powder of E. gracilis and paramylon, β-glucan stored in E. gracilis cells, to A4gnt knockout (KO) mice. A4gnt KO mice are a mutant mouse model that spontaneously develops gastric cancer through hyperplasia-dysplasia-adenocarcinoma sequence in the antrum of the stomach, and we observed the effects of E. gracilis and paramylon on the early involvements of A4gnt KO mice. Male and female 10-week-old A4gnt KO mice and their age-matched wildtype C57BL/6J mice were orally administered with 50 mg of E. gracilis or paramylon suspended in saline or saline as a control. After 3-week administration, animals were euthanatized and the stomach was examined histopathologically and immunohistochemically. Gene expression patterns of the stomach, which have been reported to be altered with A4gnt KO, and IgA concentration in small intestine were also analyzed with real-time PCR and ELISA, respectively. Administration of Euglena significantly reduced the number of stimulated CD3-positive T-lymphocytes in pyloric mucosa of A4gnt KO mice and tend to reduce polymorphonuclear leukocytes infiltration. Euglena administration further downregulated the expression of Il11 and Cxcl1 of A4gnt KO mice. Euglena administration also affected IgA concentration in small intestinal contents of A4gnt KO mice. Paramylon administration reduced the number of CD3-positive lymphocytes in pyloric mucosa of A4gnt KO mice, and downregulated the expressions of Il11 and Ccl2 of A4gnt KO mice. Although we found no significant effects on gross and microscopic signs of gastric dysplasia and cell proliferation, the present study suggests that the administration of Euglena and paramylon may ameliorate the early involvements of A4gnt mice through the effects on inflammatory reactions in the gastric mucosa. The cancer-preventing effects should be studied with long-term experiments until actual gastric cancer formation.
Collapse
|
246
|
Doulberis M, Pierre NT, Manzini G, Papaefthymiou A, Kountouras J, Klukowska-Rötzler J, Polyzos SA, Srivastava S, Exadaktylos AK, Knuchel J, Kuntzen T, Srivastava DS. Helicobacter pylori-Related Metabolic Parameters and Premalignant Gastric Mucosa Histological Lesions in Swiss Bariatric Patients. Microorganisms 2021; 9:microorganisms9071361. [PMID: 34201748 PMCID: PMC8303942 DOI: 10.3390/microorganisms9071361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 02/08/2023] Open
Abstract
Obesity, as a major risk factor of metabolic syndrome (MetS), represents a pandemic, especially in Western societies, and is considered a risk factor for malignancies. Helicobacter pylori (Hp), is a definite carcinogen with global distribution. We aimed to investigate, for the first time in Switzerland, the main gastric mucosa premalignant histological lesions of bariatric patients in correlation with MetS components and Hp Infection (Hp-I). By reviewing retrospectively 94304 patient cases, a total of 116 eligible patients having undergone bariatric surgery were identified. The mean patient age was 48.66 years. Hp(+) patients were 24% (28/116). Presence of gastric mucosa atrophy was documented in 8/28 Hp(+) patients (29%) and (2/88) Hp(-) ones (2%) (p = 0.006). Gastric mucosa intestinal metaplasia was observed in 14/28 (50%) Hp(+) patients versus 3/88 (3.4%) of Hp(-) group (p < 0.0001). Hp(+) patients exhibited statistically higher arterial hypertension (p = 0.033). The homeostatic model of assessment insulin resistance was also statistically significantly higher for the Hp(+) group (p < 0.001). In a multivariate analysis, including arterial hypertension, gastric mucosa atrophy, and intestinal metaplasia as variables, statistical significance remained only for intestinal metaplasia (p = 0.001). In conclusion, Hp-I is associated with premalignant gastric mucosa histologic lesions and MetS components, including arterial hypertension and IR. Further large-scale prospective studies are required to confirm these findings.
Collapse
Affiliation(s)
- Michael Doulberis
- Emergency Department, University Hospital Inselspital of Bern, 3010 Bern, Switzerland; (N.T.P.); (J.K.-R.); (A.K.E.); (D.S.S.)
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland; (J.K.); (T.K.)
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642 Thessaloniki, Macedonia, Greece; (A.P.); (J.K.)
- First Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece;
- Correspondence:
| | - Noah Thierry Pierre
- Emergency Department, University Hospital Inselspital of Bern, 3010 Bern, Switzerland; (N.T.P.); (J.K.-R.); (A.K.E.); (D.S.S.)
- Private Practice, 4704 Niederbipp, Switzerland
| | - Giulia Manzini
- Department of General and Visceral Surgery, Kantonsspital Aarau, 5001 Aarau, Switzerland;
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642 Thessaloniki, Macedonia, Greece; (A.P.); (J.K.)
- First Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece;
- Department of Gastroenterology, University Hospital of Larisa, Mezourlo, 41110 Larisa, Thessaly, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642 Thessaloniki, Macedonia, Greece; (A.P.); (J.K.)
| | - Jolanta Klukowska-Rötzler
- Emergency Department, University Hospital Inselspital of Bern, 3010 Bern, Switzerland; (N.T.P.); (J.K.-R.); (A.K.E.); (D.S.S.)
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece;
| | | | - Aristomenis K. Exadaktylos
- Emergency Department, University Hospital Inselspital of Bern, 3010 Bern, Switzerland; (N.T.P.); (J.K.-R.); (A.K.E.); (D.S.S.)
| | - Jürg Knuchel
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland; (J.K.); (T.K.)
| | - Thomas Kuntzen
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland; (J.K.); (T.K.)
| | - David S. Srivastava
- Emergency Department, University Hospital Inselspital of Bern, 3010 Bern, Switzerland; (N.T.P.); (J.K.-R.); (A.K.E.); (D.S.S.)
- Department of General Internal Medicine, Kliniken Hirslanden Beau-Site, 3013 Bern, Switzerland
| |
Collapse
|
247
|
Hsiao YJ, Wen YC, Lai WY, Lin YY, Yang YP, Chien Y, Yarmishyn AA, Hwang DK, Lin TC, Chang YC, Lin TY, Chang KJ, Chiou SH, Jheng YC. Application of artificial intelligence-driven endoscopic screening and diagnosis of gastric cancer. World J Gastroenterol 2021; 27:2979-2993. [PMID: 34168402 PMCID: PMC8192292 DOI: 10.3748/wjg.v27.i22.2979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/10/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
The landscape of gastrointestinal endoscopy continues to evolve as new technologies and techniques become available. The advent of image-enhanced and magnifying endoscopies has highlighted the step toward perfecting endoscopic screening and diagnosis of gastric lesions. Simultaneously, with the development of convolutional neural network, artificial intelligence (AI) has made unprecedented breakthroughs in medical imaging, including the ongoing trials of computer-aided detection of colorectal polyps and gastrointestinal bleeding. In the past demi-decade, applications of AI systems in gastric cancer have also emerged. With AI's efficient computational power and learning capacities, endoscopists can improve their diagnostic accuracies and avoid the missing or mischaracterization of gastric neoplastic changes. So far, several AI systems that incorporated both traditional and novel endoscopy technologies have been developed for various purposes, with most systems achieving an accuracy of more than 80%. However, their feasibility, effectiveness, and safety in clinical practice remain to be seen as there have been no clinical trials yet. Nonetheless, AI-assisted endoscopies shed light on more accurate and sensitive ways for early detection, treatment guidance and prognosis prediction of gastric lesions. This review summarizes the current status of various AI applications in gastric cancer and pinpoints directions for future research and clinical practice implementation from a clinical perspective.
Collapse
Affiliation(s)
- Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yuan-Chih Wen
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Critical Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | | | - De-Kuang Hwang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Tai-Chi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Yun-Chia Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Ting-Yi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kao-Jung Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ying-Chun Jheng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Big Data Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
248
|
Metabolite Characteristics in Tongue Coating from Damp Phlegm Pattern in Patients with Gastric Precancerous Lesion. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5515325. [PMID: 34122594 PMCID: PMC8189775 DOI: 10.1155/2021/5515325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/12/2021] [Accepted: 05/21/2021] [Indexed: 12/23/2022]
Abstract
Objective In this study, we analyzed the metabolite profile of the tongue coating of patients having gastric precancerous lesion (GPL) with damp phlegm pattern and proposed a mechanism of pathological transition. Methods The changes in tongue-coating metabolites in patients with GPL damp phlegm pattern were analyzed using GC-TOF-MS and UHPLC-QE-MS metabolomics methods. Results When compared with 20 patients who did not exhibit a nondamp phlegm pattern, 12 metabolites were highly expressed and 10 metabolites were under expressed in 40 cases of damp phlegm pattern, of which involved 9 metabolic pathways. Compared with 15 healthy people, 134 metabolites were upregulated and 3 metabolites were downregulated in 40 cases exhibiting a damp phlegm pattern, of which involved 17 metabolic pathways. The patients with damp phlegm pattern were compared with nondamp phlegm pattern patients and healthy people, the main differential metabolites were primarily lipids and lipid-like molecules, and the main differential metabolic pathways were related to glycerophospholipid metabolism. In the glycerophospholipid metabolism, the metabolites with changes were phosphatidylethanolamine and lysoPC(18 : 1 (9z)). Among them, phosphatidylethanolamine exists in the synthesis stage of glycerophospholipid metabolism. Conclusions Abnormal expression of lipids and lipid-like molecules, as the major metabolic change, was involved in the formation of GPL patients with damp phlegm pattern.
Collapse
|
249
|
Follow-Up Study Confirms the Presence of Gastric Cancer DNA Methylation Hallmarks in High-Risk Precursor Lesions. Cancers (Basel) 2021; 13:cancers13112760. [PMID: 34199386 PMCID: PMC8199626 DOI: 10.3390/cancers13112760] [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] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/31/2022] Open
Abstract
To adopt prevention strategies in gastric cancer, it is imperative to develop robust biomarkers with acceptable costs and feasibility in clinical practice to stratified populations according to risk scores. With this aim, we applied an unbiased genome-wide CpG methylation approach to a discovery cohort composed of gastric cancer (n = 24), and non-malignant precursor lesions (n = 64). Then, candidate-methylation approaches were performed in a validation cohort of precursor lesions obtained from an observational longitudinal study (n = 264), with a 12-year follow-up to identify repression or progression cases. H. pylori stratification and histology were considered to determine their influence on the methylation dynamics. As a result, we ascertained that intestinal metaplasia partially recapitulates patterns of aberrant methylation of intestinal type of gastric cancer, independently of the H. pylori status. Two epigenetically regulated genes in cancer, RPRM and ZNF793, consistently showed increased methylation in intestinal metaplasia with respect to earlier precursor lesions. In summary, our result supports the need to investigate the practical utilities of the quantification of DNA methylation in candidate genes as a marker for disease progression. In addition, the H. pylori-dependent methylation in intestinal metaplasia suggests that pharmacological treatments aimed at H. pylori eradication in the late stages of precursor lesions do not prevent epigenome reprogramming toward a cancer signature.
Collapse
|
250
|
Association of Helicobacter pylori Infection and Host Cytokine Gene Polymorphism with Gastric Cancer. Can J Gastroenterol Hepatol 2021; 2021:8810620. [PMID: 34136433 PMCID: PMC8177986 DOI: 10.1155/2021/8810620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
The global cancer burden of new cases of various types rose with millions of death in 2018. Based on the data extracted by GLOBOCAN 2018, gastric cancer (GC) is the third leading cause of mortality related to cancer across the globe. Carcinogenic or oncogenic infections associated with Helicobacter pylori (Hp) are regarded as one of the essential risk factors for GC development. It contributes to the increased production of cytokines that cause inflammation prior to their growth in the host cells. Hp infections and specific types of polymorphisms within the host cells encoding cytokines are significant contributors to the host's increased susceptibility in terms of the development of GC. Against the backdrop of such an observation is that only a small portion of the cells infected can become malignant. The diversities are a consequence of the differences in the pathogenic pathway of the Hp, susceptibility of the host, environmental conditions, and interplay between these factors. It is evident that hosts carrying cytokine genes with high inflammatory levels and polymorphism tend to exhibit an increased risk of development of GC, with special emphasis being placed on the host cytokines gene polymorphisms.
Collapse
|