201
|
Toyoshima O, Nishizawa T. Kyoto classification of gastritis: Advances and future perspectives in endoscopic diagnosis of gastritis. World J Gastroenterol 2022; 28:6078-6089. [PMID: 36483157 PMCID: PMC9724483 DOI: 10.3748/wjg.v28.i43.6078] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/06/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
This editorial provides an update of the recent evidence on the endoscopy-based Kyoto classification of gastritis, clarifying the shortcomings of the Kyoto classification, and providing prospects for future research, with particular focus on the histological subtypes of gastric cancer (GC) and Helicobacter pylori (H. pylori) infection status. The total Kyoto score is designed to express GC risk on a score ranging from 0 to 8, based on the following five endoscopic findings: Atrophy, intestinal metaplasia (IM), enlarged folds (EF), nodularity, and diffuse redness (DR). The total Kyoto score reflects H. pylori status as follows: 0, ≥ 2, and ≥ 4 indicate a normal stomach, H. pylori-infected gastritis, and gastritis at risk for GC, respectively. Regular arrangement of collecting venules (RAC) predicts non-infection; EF, nodularity, and DR predict current infection; map-like redness (MLR) predicts past infection; and atrophy and IM predict current or past infection. Atrophy, IM, and EF all increase the incidence of H. pylori-infected GC. MLR is a specific risk factor for H. pylori-eradicated GC, while RAC results in less GC. Diffuse-type GC can be induced by active inflammation, which presents as EF, nodularity, and atrophy on endoscopy, as well as neutrophil and mononuclear cell infiltration on histology. In contrast, intestinal-type GC develops via atrophy and IM, and is consistent between endoscopy and histology. However, this GC risk-scoring design needs to be improved.
Collapse
Affiliation(s)
- Osamu Toyoshima
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
| | - Toshihiro Nishizawa
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
- Department of Gastroenterology and Hepatology, International University of Medicine and Welfare, Narita 286-8520, Japan
| |
Collapse
|
202
|
Kriķe P, Shums Z, Poļaka I, Kikuste I, Vanags A, Tolmanis I, Isajevs S, Liepniece-Karele I, Santare D, Tzivian L, Rudzīte D, Song M, Camargo MC, Norman GL, Leja M. The Diagnostic Value of Anti-Parietal Cell and Intrinsic Factor Antibodies, Pepsinogens, and Gastrin-17 in Corpus-Restricted Atrophic Gastritis. Diagnostics (Basel) 2022; 12:2784. [PMID: 36428844 PMCID: PMC9689963 DOI: 10.3390/diagnostics12112784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
We aimed to determine the diagnostic value of anti-parietal cell antibodies (anti-PCA), anti-intrinsic factor antibodies (anti-IFA), pepsinogen ratio (PGI/II), and gastrin-17 (G-17) in corpus-restricted atrophic gastritis (CRAG) detected by ELISA (Inova, Biohit). Our study compared 29 CRAG cases against 58 age- and sex-matched controls with mild or no atrophy. Anti-PCA and anti-IFA positive cutoff values were ≥25 units for both. PGI/II value <3 was considered characteristic for atrophy; positive cutoff values for G-17 and anti-H. pylori IgG were >5 pg/L and >30 EIU. Anti-PCA was positive in 65.5% For CRAG cases and 13.8% of the controls (p < 0.0001), anti-IFA was positive in 13.8% and 0% (p = 0.01), respectively. Decreased pepsinogen levels were present in 79.3% of CRAG cases and 10.3% of the controls (p < 0.0001). PGI/II ratio was the best single biomarker, with sensitivity = 79%, specificity = 90%, and AUC 0.90. The combined use of PGI/II and anti-PCA resulted in AUC 0.93 for detecting CRAG. Our study suggests that the best combination of non-invasive biomarkers for detecting CRAG is PGI/II with anti-PCA. The addition of G-17 and anti-IFA is of little utility in clinical application.
Collapse
Affiliation(s)
- Petra Kriķe
- Institute of Clinical and Preventive Medicine & Faculty of Medicine, University of Latvia, LV-1586 Rīga, Latvia
- Pauls Stradins Clinical University Hospital, LV-1002 Riga, Latvia
| | - Zakera Shums
- Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA
| | - Inese Poļaka
- Institute of Clinical and Preventive Medicine & Faculty of Medicine, University of Latvia, LV-1586 Rīga, Latvia
| | - Ilze Kikuste
- Digestive Diseases Centre GASTRO, Hong Kong 122001, China
| | - Aigars Vanags
- Digestive Diseases Centre GASTRO, Hong Kong 122001, China
| | - Ivars Tolmanis
- Digestive Diseases Centre GASTRO, Hong Kong 122001, China
| | | | | | - Daiga Santare
- Institute of Clinical and Preventive Medicine & Faculty of Medicine, University of Latvia, LV-1586 Rīga, Latvia
- Riga East University Hospital, LV-1038 Riga, Latvia
| | - Lilian Tzivian
- Institute of Clinical and Preventive Medicine & Faculty of Medicine, University of Latvia, LV-1586 Rīga, Latvia
| | - Dace Rudzīte
- Riga East University Hospital, LV-1038 Riga, Latvia
| | - Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - Gary L. Norman
- Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA 92121, USA
| | - Mārcis Leja
- Institute of Clinical and Preventive Medicine & Faculty of Medicine, University of Latvia, LV-1586 Rīga, Latvia
- Digestive Diseases Centre GASTRO, Hong Kong 122001, China
- Riga East University Hospital, LV-1038 Riga, Latvia
| |
Collapse
|
203
|
Fang S, Fu Y, Du S, Wang L, Qing X, Luo X, Song G, Yang Y, Wei W. The role of the endoscopic grading of gastric intestinal metaplasia in assessing gastric cancer risk: A systematic review and meta-analysis. Front Oncol 2022; 12:1018248. [PMID: 36425561 PMCID: PMC9679375 DOI: 10.3389/fonc.2022.1018248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/17/2022] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND AND AIM Patients with gastric intestinal metaplasia (IM) are at increased risk of gastric cancer (GC). The endoscopic grading of gastric intestinal metaplasia (EGGIM) with high-definition endoscopes has shown the potential to facilitate GC risk stratification. However, a comprehensive review and meta-analysis of published articles are lacking. We conducted a meta-analysis to access the value of EGGIM in the assessment of histological IM. MATERIALS Studies were selected from PubMed, Medline, Embase, and Cochrane (last selection, Jun 2022). We extracted relevant data to calculate the accuracy of EGGIM compared with the operative link of gastric intestinal metaplasia (OLGIM) and to calculate pooled odds ratio (OR) with a 95% confidence interval (CI) assessing GC risk with different grading. RESULTS Four diagnostic studies and three case-control clinical trials were included in the analysis, which included 665 patients and 738 patients, respectively. Compared with OLGIM III/IV, EGGIM(5-10) had a pooled sensitivity and specificity of 0.92(95%CI 0.86-0.96) and 0.90(95%CI 0.88-0.93), and the area under the curve(AUC) was 0.9702. In assessing early GC, the pooled OR of patients with EGGIM(5-10) was 7.46(95%CI 3.41-16.31) compared with that of EGGIM(0-4). CONCLUSIONS EGGIM is highly consistent with OLGIM, and patients with EGGIM(5-10) are at a higher risk for early GC. Some heterogeneity in the current research suggests that we need to carry out more strict control of confounding factors. SYSTEMATIC REVIEW REGISTRATION [https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=248691], (Prospero registration number: 248691).
Collapse
Affiliation(s)
- Shuangshuang Fang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuhan Fu
- Department of Internal Medicine, MetroHealth Medical Center/Case Western Reserve University, Cleveland, OH, United States
| | - Sijing Du
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Wang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiangli Qing
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoying Luo
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gengqing Song
- Department of Gastroenterology and Hepatology, MetroHealth Medical Center/Case Western Reserve University, Cleveland, OH, United States
| | - Yang Yang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Wei
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
204
|
Cifuentes JDG, Sparkman J, Graham DY. Management of upper gastrointestinal symptoms in patients with autoimmune gastritis. Curr Opin Gastroenterol 2022; 38:600-606. [PMID: 36165039 PMCID: PMC9561041 DOI: 10.1097/mog.0000000000000878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW Autoimmune gastritis is characterized by atrophy of acid secreting parietal cells resulting in achlorhydria. Upper gastrointestinal symptoms are common in autoimmune gastritis and frequently result in prescriptions for acid suppressant medications despite the inability of the stomach to secrete acid. Evidence-based recommendations for management of gastrointestinal symptoms in autoimmune gastritis are lacking. RECENT FINDINGS The most common symptoms in patients with autoimmune gastritis are dyspepsia, heartburn, and regurgitation. Gastroesophageal reflux should be confirmed by pH-impedance testing and is typically weakly acid or alkaline. Therapy for reflux focuses on mechanical prevention of reflux (i.e., elevation of the head of the bed and alginates) or when severe, antireflux surgery. The etiology of dyspepsia in autoimmune gastritis is unclear and largely unstudied. In the first half of the 20th century, oral administration of acid to "aid digestion" was widely used with reported success. However, randomized, placebo-controlled trials are lacking. Here, we provide suggestions for attempting gastric acidification therapy. SUMMARY Upper GI symptoms are common in autoimmune gastritis. Their pathogenesis and therapy remain incompletely understood. Acid suppressant medications are useless and should be discontinued. A trial of acid replacement therapy is recommended especially in the form of placebo-controlled trials.
Collapse
Affiliation(s)
| | | | - David Y. Graham
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
205
|
Mosiychuk L, Tatarchuk O, Konenko I, Petishko O. Cytokine profile in patients with atrophic gastritis in comorbidity with thyroid gland pathology. Gastroenterology 2022; 56:143-148. [DOI: 10.22141/2308-2097.56.3.2022.502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Optimization of the management of patients with precancerous conditions of the stomach in comorbid pathology is an urgent task of gastroenterology. The purpose of the study: to evaluate the content of pro- and anti-inflammatory cytokines, as well as the level of vascular endothelial growth factor (VEGF) at different degrees of vascularization of the thyroid gland in patients with atrophic gastritis. Materials and methods. A study was conducted in 120 patients with atrophic gastritis and thyroid pathology. All of them underwent a sonological examination of the thyroid gland using an ultrasound scanner Toshiba Xario (Japan), which revealed poor vascularization of the parenchyma in 82 (68.3%) cases, moderate vascularization was diagnosed in 20 (16.7%) patients and marked— in 18 (15.0%). Quantitative content of cytokines (interleukins (IL) 8, 10, 18, tumor necrosis factorα), vascular endothelial growth factor in the blood serum was determined by immunoenzymatic assay using Vector-Best reagents. Results. Among patients with poor vascularization, only atrophic changes of the gastric mucosa were diagnosed in almost half— 39 (47.6%) people, while dysplastic changes of the gastric mucosa were found in a third of patients with marked vascularization. Intestinal metaplasia was detected in half of cases, regardless of the degree of vascularization of the thyroid parenchyma. Cytokine imbalance was found in patients with atrophic gastritis, with the greatest shift in case of marked vascularization of the thyroid parenchyma due to a 4-fold increase in the level of pro-inflammatory cytokines IL-8 (р<0.05) and by 1.7 times of IL-18 (p<0.05), with a simultaneous decrease in the content of the anti-inflammatory cytokine IL-10 by 3.2 times (p<0.05) compared to control indicators. The level of VEGF in the blood serum of patients with atrophic gastritis with moderate and marked vascularization of the thyroid parenchyma was significantly increased by 1.5 (p<0.05) and 1.7 times (p<0.05), respectively, compared to that of people with poor vascularization. Correlations were revealed between the degree of vascularization of the thyroid parenchyma and the level of IL-8 (r=0.491; p=0.031) and VEGF (r=0.444; p=0.019) in patients with atrophic gastritis. Conclusions. Patients with atrophic gastritis had a cytokine imbalance, which deepens as the vascularization of the thyroid gland increases that is possibly related to common pathogenetic mechanisms of the development of comorbid pathology.
Collapse
|
206
|
Stepanov Y, Mosiychuk L, Gaidar Y, Shevtsova O, Petishko O, Mylostiva D. Features of the histostructure of the gastric mucosa inpatients with atrophic gastritis combined with thyroid pathology. Gastroenterology 2022; 56:156-162. [DOI: 10.22141/2308-2097.56.3.2022.504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. The search for factors that influence the progression of structural remodeling of the gastric mucosa and carcinogenesis remains a promising area of gastroenterology. The purpose of the study: to study the microstructure of the gastric mucosa in patients with chronic atrophic gastritis depending on the presence of structural changes in the thyroid gland. Materials and methods. A study of histostructural changes of the gastric mucosa was carried out in 121 patients with atrophic gastritis, taking into account nodular and diffuse changes in the parenchyma of the thyroid gland. The degree and stage of gastritis were assessed according to the OLGA system. We studied the allergic component, the severity of atrophic changes in the gastric mucosa, the presence and severity of sclerotic changes in the stroma, and epithelial hyperplasia. Ultrasound examination of the thyroid gland was performed on an ultrasound scanner Toshiba Xario (Japan). Results. The distribution of patients according to the frequency of detection of atrophy in different loci of the stomach showed that intestinal metaplasia in the body of the stomach was observed 1.9 times more often in patients with nodular changes than in those with diffuse changes (χ2= 5.33; p< 0.05) and 2.8 times— than in people without structural changes of the thyroid gland (χ2= 8.12; p< 0.01). Similarly, atrophy in the angle of the stomach was most often detected in patients with nodular changes in the thyroid gland— in 65.0% of cases, which is 25.7% more often compared to those with diffuse changes (p> 0.05) and 38.3% more often compared to people without structural changes of the thyroid gland (χ2=6.27; p< 0.05). According to the results of the correlation analysis, a connection was found between the presence of nodular changes in the thyroid gland and the degree of atrophy of the mucous membrane of the body of the stomach (r= 0.49; p= 0.011) and the angle of the stomach (r= 0.52; p= 0.037), the presence of intestinal metaplasia in the mucous membrane of the antral part of the stomach (r= 0.54; p=0.013) and the angle of the stomach (r= 0.41; p= 0.028). Conclusions. The progression of structural changes in the gastric mucosa occurs most often in patients with nodular changes in the thyroid gland. For early diagnosis of precancerous conditions and changes in the mucous membrane of the stomach in patients with structural changes of the thyroid gland, it is advisable to conduct a thorough endoscopic examination of the stomach using modern highly informative technologies.
Collapse
|
207
|
Yang H, Guan L, Hu B. Detection and Treatment of Helicobacter pylori: Problems and Advances. Gastroenterol Res Pract 2022; 2022:4710964. [PMID: 36317106 PMCID: PMC9617708 DOI: 10.1155/2022/4710964] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 12/24/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection is chronic and etiologically linked to gastric cancer (GC) derived from gastric epithelium. The potential mechanism is complex, covering chronic inflammation, epithelial senescence, NF-κB activation, the cytotoxin-associated gene A protein translocation, and related abnormal signaling pathways. In clinical practice, the test-and-treat strategy, endoscopy-based strategy, and (family-based) screen-and-treat strategy are recommended to detect H. pylori and prevent GC. It has been demonstrated that the decreasing annual incidence of GC is largely attributable to the management of H. pylori. This study reviews the current clinical practice of H. pylori on the detection and eradication, alternative treatment strategies, and related problems and advances, and hopes to contribute to the better clinical management of H. pylori.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liwen Guan
- Department of Gastroenterology, Sanya Central Hospital (Hainan Third People's Hospital), Sanya, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
208
|
Yu XT, Chen M, Guo J, Zhang J, Zeng T. Noninvasive detection and interpretation of gastrointestinal diseases by collaborative serum metabolite and magnetically controlled capsule endoscopy. Comput Struct Biotechnol J 2022; 20:5524-5534. [PMID: 36249561 PMCID: PMC9550535 DOI: 10.1016/j.csbj.2022.10.001] [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: 04/15/2022] [Revised: 09/15/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Gastrointestinal diseases are complex diseases that occur in the gastrointestinal tract. Common gastrointestinal diseases include chronic gastritis, peptic ulcers, inflammatory bowel disease, and gastrointestinal tumors. These diseases may manifest a long course, difficult treatment, and repeated attacks. Gastroscopy and mucosal biopsy are the gold standard methods for diagnosing gastric and duodenal diseases, but they are invasive procedures and carry risks due to the necessity of sedation and anesthesia. Recently, several new approaches have been developed, including serological examination and magnetically controlled capsule endoscopy (MGCE). However, serological markers lack lesion information, while MGCE images lack molecular information. This study proposes combining these two technologies in a collaborative noninvasive diagnostic scheme as an alternative to the standard procedures. We introduce an interpretable framework for the clinical diagnosis of gastrointestinal diseases. Based on collected blood samples and MGCE records of patients with gastrointestinal diseases and comparisons with normal individuals, we selected serum metabolite signatures by bioinformatic analysis, captured image embedding signatures by convolutional neural networks, and inferred the location-specific associations between these signatures. Our study successfully identified five key metabolite signatures with functional relevance to gastrointestinal disease. The combined signatures achieved discrimination AUC of 0.88. Meanwhile, the image embedding signatures showed different levels of validation and testing accuracy ranging from 0.7 to 0.9 according to different locations in the gastrointestinal tract as explained by their specific associations with metabolite signatures. Overall, our work provides a new collaborative noninvasive identification pipeline and candidate metabolite biomarkers for image auxiliary diagnosis. This method should be valuable for the noninvasive detection and interpretation of gastrointestinal and other complex diseases.
Collapse
Affiliation(s)
- Xiang-Tian Yu
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China,Corresponding authors at: Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, China (X.-T. Yu); Guangzhou Laboratory, Guangzhou, China (T. Zeng).
| | - Ming Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jingyi Guo
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jing Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tao Zeng
- Guangzhou Laboratory, Guangzhou, China,Corresponding authors at: Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, China (X.-T. Yu); Guangzhou Laboratory, Guangzhou, China (T. Zeng).
| |
Collapse
|
209
|
Shah SC. Improving the Endoscopic Detection and Management of Gastric Intestinal Metaplasia Through Training: A Practical Guide. Gastroenterology 2022; 163:806-811. [PMID: 35931107 PMCID: PMC10027538 DOI: 10.1053/j.gastro.2022.07.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Shailja C Shah
- Gastroenterology Section, VA San Diego Healthcare System, La Jolla, California; Division of Gastroenterology, University of California, San Diego, La Jolla, California; Cancer Prevention and Control Program, UCSD Moores Cancer Center, La Jolla, California.
| |
Collapse
|
210
|
Song B, Cao Q, Li T, Liu Y, Sun Q, Fan S, Li X. Biomarker identification of chronic atrophic gastritis and its potential drug analysis. FRONTIERS IN GASTROENTEROLOGY 2022; 1. [DOI: 10.3389/fgstr.2022.948323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
BackgroundChronic atrophic gastritis (CAG) is the first step of gastric precancerous lesions, and the study of the pathogenesis of CAG is helpful for the prevention and treatment of gastric cancer(GC). The purpose of this study is to explore the potential biomarkers and therapeutic drugs of CAG through bioinformatics analysis.MethodsThe GSE11632 dataset was downloaded from Gene Expression Omnibus (GEO) database and the differentially expressed genes (DEGs) were obtained by using GEO2R online tool. We searched GeneCard and DisGeNET databases for genes related to CAG and used the overlapping genes as final DEGs for further functional enrichment analysis and Protein-protein Interaction (PPI) network analysis. Tissue-specific expressed genes were identified by BioGPS database. Cytoscape software was used to identify key hub genes and validated them in GSE27411 data sets. The upstream miRNAs of hub gene was predicted by TargetScan, miRDB and miRWalk. Finally, run the Connectivity Map (CMap) to identify new potential drugs for the treatment of CAG.ResultsA total of 430 differentially expressed mRNA were identified in this study, including 315 up-regulated genes and 115 down-regulated genes. After intersecting with CAG-related genes in GeneCard and DisGeNET databases, 42 DEGs were obtained. 24 DEGs were identified as tissue-specific expressed genes, most of which were expressed in stomach. GO and KEGG pathway analysis showed that DGEs was mainly enriched in digestion, IL-1 production, gastric acid secretion and so on. A total of 6 hub genes were generated by cytoHubba plug-in, among which ATP4A, CFTR and EPCAM had high diagnostic value. A total of 13 overlapping miRNA were predicted by 6 hub genes.ConclusionATP4A, CFTR and EPCAM may be potential biomarkers of CAG. hsa-miR-185-5p-CFTR, hsa-miR-4644-CFTR and hsa-miR-4505-CFTR are potential RNA regulatory pathways to control the progression of CAG disease. Finally, amonafide, etoposide, mycophenolate-mofetil, cycloheximide and Emetine may be potential therapeutic drugs for CAG.
Collapse
|
211
|
Mechanisms of Banxia Xiexin Decoction Underlying Chronic Atrophic Gastritis via Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulations. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4237040. [PMID: 36158124 PMCID: PMC9499768 DOI: 10.1155/2022/4237040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022]
Abstract
Chronic atrophic gastritis (CAG) is a common chronically digestive disease which is notoriously characterized by atrophy of the epithelium and glands of the gastric mucosa, reduced number, thinning of the gastric mucosa, thickening of the mucosal base, or pyloric glandular hyperplasia and intestinal glandular hyperplasia, or with atypical hyperplasia. Banxia Xiexin decoction (BXD) has been applied for two thousand years and is considered an effective therapy for functional dyspepsia, gastroesophageal reflux disease and colon cancer. In this current study, to probe into the underlying mechanism of BXD on CAG, network pharmacology was conducted to collect druggable ingredients and predicted targets of BXD and the CAG-associated targets were harvested to take intersection with druggable ingredients from BXD predicted targets to obtain potential critical action targets. Subsequently, GO enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were conducted to elucidate the underlying mechanisms and roles from the perspective of overall pathways and cellular functions. Eventually, molecular docking integrated with molecular dynamics simulations was conducted to further investigate the mechanism of action of BXD active ingredients on CAG from drug molecule-target interactions and to provide a theoretical basis for BXD drug development.
Collapse
|
212
|
Yin Z, Xiao S, Tian X, Yuan Z, Zhou L. The necessity and appropriate range of the diagnostic "gray zone" of 13C-urea breath test. Saudi J Gastroenterol 2022; 28:385-392. [PMID: 35259858 DOI: 10.4103/sjg.sjg-638-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The 13C-urea breath test (13C-UBT) is preferred for non-invasive detection of Helicobacter pylori (H. pylori); however, its accuracy drops when results fall between 2‰ and 6‰ (called the gray zone). This study aimed to evaluate the accuracy of 13C-UBT (cut-off point 4‰) between 2‰ and 6‰, find a more appropriate gray zone, and identify the factors influencing 13C-UBT. METHODS Patients with 13C-UBT results 2‰-6‰, over an eight-year period, were studied. H. pylori infection was diagnosed if patients were positive for either Warthin-Starry staining or quantitative real-time polymerase chain reaction (real-time PCR), and excluded if both were negative. Accuracy of 13C-UBT under different cut-off points was calculated, and the factors affecting 13C-UBT were analyzed. RESULTS A total of 208 patients were included, of whom 129 were H. pylori-positive. Sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV) of 13C-UBT were 71.32%, 83.54%, 64.08%, and 87.62%, respectively. When the cut-off point was changed to 2.15‰, the NPV of 13C-UBT reached a maximum (76.47%); when the cut-off point was changed to 4.95‰, PPV reached its maximum (93.22%). Therefore, the original gray zone (2‰-6‰) was adjusted to 2‰-4.95‰. Gastric antral intestinal metaplasia (OR = 3.055, 95% CI: 1.003-9.309) was an independent risk factor for false-negative 13C-UBT. CONCLUSIONS Accuracy of 13C-UBT over 2‰-6‰ was poor, and the gray zone was changed to 2‰-4.95‰. 13C-UBT results over 2‰-4.95‰ should be interpreted with caution during mass screening of H. pylori, especially for patients with gastric antral intestinal metaplasia.
Collapse
Affiliation(s)
- Zhihao Yin
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Xueli Tian
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Ziying Yuan
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liya Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| |
Collapse
|
213
|
Endoscopic methods for the detection and treatment of gastric cancer. Curr Opin Gastroenterol 2022; 38:436-442. [PMID: 35881962 DOI: 10.1097/mog.0000000000000867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW The intent of this review is to describe new advances in endoscopic approaches to surveillance and management of gastric cancer. RECENT FINDINGS There are new endoscopic techniques and approaches that have improved the detection of gastric cancer, including narrow band imaging, confocal laser endocytomicroscopy and magnetically controlled capsule endoscopy. This article highlights the role of endoscopic submucosal dissection in the treatment of focal and diffuse gastric dysplasia and early gastric cancer with a discussion of indications, complications and outcomes. We review several recent guidelines addressing the surveillance strategies for individuals at high-risk for developing gastric cancer, such as those with atrophic gastritis and intestinal metaplasia, how gastric dysplasia and early gastric cancer can be endoscopically managed, and recommended surveillance after endoscopic intervention. SUMMARY Endoscopic approaches are evolving rapidly that will improve detection of dysplasia and early gastric cancer in high-risk individuals. Surveillance guidelines from various international societies reflect differences in local experience and prevalence of gastric cancer. Endoscopic submucosal dissection is now widely accepted as a first-line approach to early gastric cancers that can be resected en-bloc .
Collapse
|
214
|
Bai Y, Lei C, Zhang N, Liu Y, Hu Z, Li Y, Qi R. Peri-Ulcerative Mucosal Inflammation Appearance is an Independent Risk Factor for 30-Day Rebleeding in Patients with Gastric Ulcer Bleeding: A Multicenter Retrospective Study. J Inflamm Res 2022; 15:4951-4961. [PMID: 36065317 PMCID: PMC9440673 DOI: 10.2147/jir.s378263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/12/2022] [Indexed: 12/07/2022] Open
Abstract
AIM The aim of this study was to identify clinical endoscopic indicators related to peri-ulcerative mucosal inflammation and to analyze whether the degree of peri-ulcerative mucosal inflammation appearance is an independent risk factor for gastric ulcer rebleeding. METHODS We conducted a retrospective study that included patients with gastric ulcer bleeding who were hospitalized at three medical centers in China from January 1, 2016 to December 31, 2019. Ulcer rebleeding that occurred within 30 days of successful initial hemostasis was analyzed to determine whether this event was related to the degree of peri-ulcerative mucosal inflammation appearance or other mucosal inflammation-related factors. RESULTS We enrolled 1111 patients and determined that GBS-Rebleeding-ROC (P<0.001), age (P=0.01), use of NSAIDs (P=0.001), bile reflux (P<0.001), and Helicobacter pylori (P<0.001) are all risk factors for peri-ulcerative mucosal inflammation appearance. Through multivariate analysis, we determined that severe peri-ulcerative mucosal inflammation appearance (P=0.002) was an independent risk factor for ulcer rebleeding within 30 days. Finally, we developed a risk assessment model using factors associated with mucosal inflammation that may be useful for early prediction of rebleeding. CONCLUSION The risk factors for peri-ulcerative mucosal inflammation appearance were identified. Severe peri-ulcerative mucosal inflammation appearance is an independent risk factor for ulcer rebleeding.
Collapse
Affiliation(s)
- Yixuan Bai
- Department of Digestive Internal Medicine, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Chenggang Lei
- Department of Hepatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, People’s Republic of China
| | - Na Zhang
- Department of Digestive Internal Medicine, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yuhui Liu
- Department of Digestive Internal Medicine, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Zhengyu Hu
- Department of General Surgery, Shanghai Tenth People’s Hospital, Affiliated to Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Affiliated to Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ran Qi
- Department of General Surgery, Tongji Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
215
|
Yamamoto Y, Yoshida N, Yano T, Horimatsu T, Uedo N, Kawata N, Kanzaki H, Hori S, Yao K, Abe S, Katada C, Yokoi C, Ohata K, Doyama H, Yoshimura K, Ishikawa H, Muto M. Assessment of Outcomes From 1-Year Surveillance After Detection of Early Gastric Cancer Among Patients at High Risk in Japan. JAMA Netw Open 2022; 5:e2227667. [PMID: 35984658 PMCID: PMC9391963 DOI: 10.1001/jamanetworkopen.2022.27667] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Single endoscopic examination often misses early gastric cancer (GC), even when both high-definition white light imaging and narrow-band imaging are used. It is unknown whether new GC can be detected approximately 1 year after intensive index endoscopic examination. OBJECTIVE To examine whether new GC can be detected approximately 1 year after intensive index endoscopic examination using both white light and narrow-band imaging. DESIGN, SETTING, AND PARTICIPANTS This case-control study was a preplanned secondary analysis of a randomized clinical trial involving 4523 patients with a high risk of GC who were enrolled between October 1, 2014, and September 22, 2017. Data were analyzed from December 26, 2019, to April 21, 2021. Participants in the clinical trial received index endoscopy to detect early GC via 2 examinations of the entire stomach using white light and narrow-band imaging. The duration of follow-up was 15 months. The secondary analysis included 107 patients with newly detected GC (case group) and 107 matched patients without newly detected GC (control group) within 15 months after index endoscopy. INTERVENTIONS Surveillance endoscopy was scheduled between 9 and 15 months after index endoscopy. If new lesions suspected of being early GC were detected during surveillance endoscopy, biopsies were obtained to confirm the presence of cancer. MAIN OUTCOMES AND MEASURES The primary end point was the rate of new GC detected within 15 months after index endoscopy. The main secondary end point was identification of risk factors associated with new GC detected within 15 months after index endoscopy. RESULTS Among 4523 patients (mean [SD] age, 70.6 [7.5] years; 3527 men [78.0%]; all of Japanese ethnicity) enrolled in the clinical trial, 4472 received index endoscopy; the rate of early GC detected on index endoscopy was 3.0% (133 patients). Surveillance endoscopy was performed in 4146 of 4472 patients (92.7%) who received an index endoscopy; the rate of new GC detected within 15 months after index endoscopy was 2.6% (107 patients). Among 133 patients for whom early GC was detected during index endoscopy, 110 patients (82.7%) received surveillance endoscopy within 15 months after index endoscopy; the rate of newly detected GC was 10.9% (12 patients). For the secondary analysis of risk factors associated with newly detected GC, characteristics were well balanced between the 107 patients included in the case group vs the 107 patients included in the matched control group (mean [SD] age, 71.7 [7.2] years vs 71.8 [7.0] years; 94 men [87.9%] in each group; 82 patients [76.6%] vs 87 patients [81.3%] with a history of gastric neoplasm). Multivariate analysis revealed that the presence of open-type atrophic gastritis (odds ratio, 6.00; 95% CI, 2.25-16.01; P < .001) and early GC detection by index endoscopy (odds ratio, 4.67; 95% CI, 1.08-20.21; P = .04) were independent risk factors associated with new GC detection. CONCLUSIONS AND RELEVANCE In this study, the rate of new GC detected by surveillance endoscopy approximately 1 year after index endoscopy was similar to that of early GC detected by index endoscopy. These findings suggest that 1-year surveillance is warranted for patients at high risk of GC.
Collapse
Affiliation(s)
- Yoshinobu Yamamoto
- Department of Gastrointestinal Oncology, Hyogo Cancer Center, Akashi, Hyogo, Japan
| | - Naohiro Yoshida
- Department of Gastroenterology, Ishikawa Prefectural Central Hospital, Kanazawa, Ishikawa, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Takahiro Horimatsu
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Osaka, Japan
| | - Noboru Kawata
- Division of Endoscopy, Shizuoka Cancer Center, Suntogun, Shizuoka, Japan
| | - Hiromitsu Kanzaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Shinichiro Hori
- Department of Endoscopy, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Kenshi Yao
- Department of Endoscopy, Fukuoka University Chikushi Hospital, Chikushino, Fukuoka, Japan
| | - Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Chuo, Tokyo, Japan
| | - Chikatoshi Katada
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Chizu Yokoi
- Endoscopy Division, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| | - Ken Ohata
- Department of Gastroenterology, NTT Medical Center Tokyo, Shinagawa, Tokyo, Japan
| | - Hisashi Doyama
- Department of Gastroenterology, Ishikawa Prefectural Central Hospital, Kanazawa, Ishikawa, Japan
| | - Kenichi Yoshimura
- Department of Biostatistics, Hiroshima University Hospital, Hiroshima University, Hiroshima, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| |
Collapse
|
216
|
Sun Z, Yu X. A case report: subacute combined degeneration of the spinal cord and pernicious anemia caused by autoimmune gastritis. Medicine (Baltimore) 2022; 101:e29226. [PMID: 35777065 PMCID: PMC9239667 DOI: 10.1097/md.0000000000029226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Autoimmune gastritis causing both subacute combined degeneration of the spinal cord and pernicious anemia is rare in clinical practice. Here, we report a case of subacute combined degeneration of the spinal cord and pernicious anemia resulting from vitamin B12 deficiency due to autoimmune gastritis. PATIENT CONCERNS A 66-year-old woman presented with a 2-month history of numbness in her extremities. DIAGNOSES The diagnoses were (1) autoimmune gastritis (2) subacute combined degeneration of the spinal cord (3) pernicious anemia (4) hypergastrinemia (5) chronic lymphocytic thyroiditis. INTERVENTIONS The patient received intramuscular methylcobalamin treatment for 5 days, followed by oral methylcobalamin daily.Outcomes: Symptoms improved, and anemia recovered in the second month after discharge. She discontinued her medication afterward, and the neurological symptoms recurred. CONCLUSIONS Autoimmune gastritis can lead to several diseases if not intervened in the early course. Neuropathy and hematopathy recur with treatment discontinuity. Methylcobalamin and adenosylcobalamin are unlikely to be more effective than vitamin B12.
Collapse
Affiliation(s)
- Zhihuan Sun
- From the Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Pudong New District, Shanghai, China
| | - Xiaofei Yu
- From the Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Pudong New District, Shanghai, China
- *Correspondence: Xiaofei Yu, the Institute of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Pudong New District, Shanghai, China (e-mail: )
| |
Collapse
|
217
|
Lenti MV, Facciotti F, Miceli E, Vanoli A, Fornasa G, Lahner E, Spadoni I, Giuffrida P, Arpa G, Pasini A, Rovedatti L, Caprioli F, Travelli C, Lattanzi G, Conti L, Klersy C, Vecchi M, Paulli M, Annibale B, Corazza GR, Rescigno M, Di Sabatino A. Mucosal Overexpression of Thymic Stromal Lymphopoietin and Proinflammatory Cytokines in Patients With Autoimmune Atrophic Gastritis. Clin Transl Gastroenterol 2022; 13:e00510. [PMID: 35905420 PMCID: PMC10476748 DOI: 10.14309/ctg.0000000000000510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 09/06/2023] Open
Abstract
INTRODUCTION The immune mechanisms underlying human autoimmune atrophic gastritis (AAG) are poorly understood. We sought to assess immune mucosal alterations in patients with AAG. METHODS In 2017-2021, we collected gastric corpus biopsies from 24 patients with AAG (median age 62 years, interquartile range 56-67, 14 women), 26 age-matched and sex-matched healthy controls (HCs), and 14 patients with Helicobacter pylori infection (HP). We investigated the lamina propria mononuclear cell (LPMC) populations and the mucosal expression of thymic stromal lymphopoietin (TSLP) and nicotinamide phosphoribosyltransferase (NAMPT). Ex vivo cytokine production by organ culture biopsies, under different stimuli (short TSLP and zinc-l-carnosine), and the gastric vascular barrier through plasmalemma vesicle-associated protein-1 (PV1) were also assessed. RESULTS In the subset of CD19+ LPMC, CD38+ cells (plasma cells) were significantly higher in AAG compared with HC. Ex vivo production of tumor necrosis factor (TNF)-α, interleukin (IL)-15, and transforming growth factor β1 was significantly higher in AAG compared with HC. At immunofluorescence, both IL-7R and TSLP were more expressed in AAG compared with HC and HP, and short TSLP transcripts were significantly increased in AAG compared with HC. In the supernatants of AAG corpus mucosa, short TSLP significantly reduced TNF-α, while zinc-l-carnosine significantly reduced interferon-γ, TNF-α, IL-21, IL-6, and IL-15. NAMPT transcripts were significantly increased in AAG compared with HC. PV1 was almost absent in AAG, mildly expressed in HC, and overexpressed in HP. DISCUSSION Plasma cells, proinflammatory cytokines, and altered gastric vascular barrier may play a major role in AAG. TSLP and NAMPT may represent potential therapeutic targets, while zinc-l-carnosine may dampen mucosal inflammation.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Emanuela Miceli
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giulia Fornasa
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Ilaria Spadoni
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Paolo Giuffrida
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giovanni Arpa
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Alessandra Pasini
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Laura Rovedatti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, IRCCS Ca' Granda Hospital Foundation, University of Milan, Milan, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Laura Conti
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Catherine Klersy
- Clinical Epidemiology & Biometry, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, IRCCS Ca' Granda Hospital Foundation, University of Milan, Milan, Italy
| | - Marco Paulli
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Gino Roberto Corazza
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| |
Collapse
|
218
|
Sugiyama Y, Tanabe H, Matsuya T, Kobayashi Y, Murakami Y, Sasaki T, Kunogi T, Takahashi K, Ando K, Ueno N, Kashima S, Moriichi K, Tanino M, Mizukami Y, Fujiya M, Okumura T. Severe immune checkpoint inhibitor-associated gastritis: A case series and literature review. Endosc Int Open 2022; 10:E982-E989. [PMID: 35845030 PMCID: PMC9286772 DOI: 10.1055/a-1839-4303] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023] Open
Abstract
Background and study aims Recent advances in cancer treatment have involved the clinical application of immune checkpoint inhibitors (ICIs) for various type of cancers. The adverse events associated with ICIs are generally referred to as immune-related adverse events (irAEs). Gastrointestinal irAEs are a major disorder, but gastritis is not frequently observed. The aims of this study were to elucidate the clinical, endoscopic, and histological characteristics of irAE gastritis. Patients and methods Information on patients treated with ICIs were collected from a single institute over 3 years. IrAE gastritis was identified based on the clinical course and endoscopic and histopathological findings. Of the 359 patients treated with ICIs, four cases of irAE gastritis were identified in clinical records from the endoscopy unit. The endoscopic and histopathological findings were analyzed, and further immunohistochemical studies with immune subtype markers and programmed cell death ligand-1 (PD-L1) antibody were conducted. Results Among four patients with irAE gastritis, the remarkable endoscopic characteristics were network-pattern erosion, erythematous and edematous mucosa with thick purulent discharge, and fragile mucosa. Corresponding histological features were fibrinopurulent exudate, severe inflammatory cell infiltration, and epithalaxia, respectively. The PD-L1 expression rate was ≥ 1 % in the gastric tissue of all patients with gastritis. These patients were treated with prednisolone (PSL) and their symptoms improved within a few days to 2 weeks. Conclusions IrAE gastritis were characterized by specific endoscopic findings. The appropriate endoscopic diagnosis may lead to effective treatment with PSL.
Collapse
Affiliation(s)
- Yuya Sugiyama
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Hiroki Tanabe
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Taisuke Matsuya
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yu Kobayashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuki Murakami
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Takahiro Sasaki
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Takehito Kunogi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Keitaro Takahashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Katsuyoshi Ando
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Nobuhiro Ueno
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Shin Kashima
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kentaro Moriichi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mishie Tanino
- Department of Diagnostic Pathology, Asahikawa Medical University Hospital, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yusuke Mizukami
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mikihiro Fujiya
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
219
|
Yu Z, Wang A, Hu C, Yu T, Chen J. Type-1 Grade 2 Multi-Focal Gastric Neuroendocrine Tumors Secondary to Chronic Autoimmune Gastritis. Front Med (Lausanne) 2022; 9:856125. [PMID: 35783638 PMCID: PMC9247202 DOI: 10.3389/fmed.2022.856125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundChronic autoimmune gastritis (CAG) refers to chronic atrophic gastritis due to autoimmunity. Loss of gastric glands in CAG results in hypergastrinemia and achlorhydria leading to Vitamin B12 deficiency and hyperplasia of G cells and enterochromaffin-like (ECL) cells. Vitamin B12 deficiency could cause pernicious anemia and subacute combined degeneration, while G cells and ECL cells hyperplasia might develop gastric neuroendocrine tumor (G-NET).Case PresentationA 35-year-old Chinese female presented with multi-focal type-1 Grade 2 (G2) NETs with a 14-year history of pernicious anemia and subacute combined degeneration.ConclusionHere, we report a rare case of a Chinese patient presenting G-NET combined with pernicious anemia and subacute combined degeneration, which are secondary to chronic autoimmune gastritis. This case also illustrates the importance of routine gastroscopy in patients with Vitamin B12 deficiency.
Collapse
Affiliation(s)
- Ziqi Yu
- Munich Medical Research School, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
| | - Aiyao Wang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, China
| | - Chong Hu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, China
| | - Tao Yu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, China
| | - Jianyong Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People’s Hospital, Nanchang, China
- *Correspondence: Jianyong Chen,
| |
Collapse
|
220
|
Zou Y, Chen X, Sun Y, Li P, Xu M, Fang P, Zhang S, Yuan G, Deng X, Hu H. Antibiotics-free nanoparticles eradicate Helicobacter pylori biofilms and intracellular bacteria. J Control Release 2022; 348:370-385. [PMID: 35662575 DOI: 10.1016/j.jconrel.2022.05.044] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023]
Abstract
Biofilms and intracellular survival tremendously help Helicobacter pylori (H. pylori) escape from antibacterial agents attacking, therefore issuing extreme challenges to clinical therapies. Herein, we constructed fucoidan (FU)-coated nanoparticles (FU/ML-LA/EB NPs) via simple self-assembly of biguanide derivative (metformin-linoleic acid, ML) and linoleic acid (LA), encapsulating urease inhibitor ebselen (EB) instead of antibiotics to take antibacterial effect. Negatively charged FU/ML-LA/EB NPs easily penetrated through the gastric mucus layer to arrive at infection sites, then eradicated extracellular polymeric substances (EPS) to destroy H. pylori biofilms structure. After strengthening bacterial membrane permeability, the nanoparticles could enter H. pylori and kill bacteria by inhibiting the activity of urease. FU/ML-LA/EB NPs also entered H. pylori-infected host cells through receptor-mediated internalization, in which they activated AMPK to recover lysosomal acidification for killing intracellular H. pylori. Additionally, FU/ML-LA/EB NPs alleviated oxidative stress, hence reducing gastric mucosal damage and cutting off the pathways of carcinogenesis. Notably, H. pylori burden after FU/ML-LA/EB NPs treatment was reduced to a great extent in vivo, which was significantly lower than that after treatment with clinical therapy. Antibiotics-free FU/ML-LA/EB NPs improving bacterial eradication and alleviating oxidation stress made it a powerful approach against H. pylori.
Collapse
Affiliation(s)
- Yiqing Zou
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Xiaonan Chen
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Yingying Sun
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Pengyu Li
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Mao Xu
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Pengchao Fang
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Shuqi Zhang
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Gang Yuan
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR 999077, China
| | - Haiyan Hu
- Lab of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-Sen University, Guangzhou 510006, PR China.
| |
Collapse
|
221
|
AbiMansour JP, Van Treeck BJ, Chandrasekhara V. Gastric Polyps in a Patient With Dyspepsia. Gastroenterology 2022; 162:1852-1854. [PMID: 35247460 DOI: 10.1053/j.gastro.2022.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Jad P AbiMansour
- Department of Internal Medicine and Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Benjamin J Van Treeck
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Vinay Chandrasekhara
- Department of Internal Medicine and Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota.
| |
Collapse
|
222
|
Yang H, Zhou X, Hu B. The 'reversibility' of chronic atrophic gastritis after the eradication of Helicobacter pylori. Postgrad Med 2022; 134:474-479. [PMID: 35382697 DOI: 10.1080/00325481.2022.2063604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023]
Abstract
Gram-negative bacterium Helicobacter pylori (H. pylori) infection is lifelong and usually acquired in childhood, which is etiologically linked to gastric cancer (GC). H. pylori gastritis is defined as an infectious disease with varying severity in virtually all infected subjects. Chronic atrophic gastritis (CAG) is the precancerous condition with the decrease or the loss of gastric glands, which can further be replaced by metaplasia or fibrosis. Patients with advanced stages of CAG are at higher risk of GC and should be followed up with a high-quality endoscopy every 3 years. H. pylori infection is the most common cause and its eradication is recommended, which may contribute to the regression of CAG. However, it is controversial whether CAG is reversible after eradication therapy. In the review, we discuss recent studies which provide important insights into whether CAG is 'reversibility' and when it may progress into GC after eradicating H. pylori.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinyue Zhou
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
223
|
Rugge M. Big Data on Gastric Dysplasia Support Gastric Cancer Prevention. Clin Gastroenterol Hepatol 2022; 20:1226-1228. [PMID: 35123082 DOI: 10.1016/j.cgh.2022.01.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Massimo Rugge
- Department of Medicine DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy; Azienda Zero Padova, Veneto Tumor Cancer Registry, Padova, Italy; Baylor College of Medicine, Houston, Texas
| |
Collapse
|
224
|
He J, Hu W, Ouyang Q, Zhang S, He L, Chen W, Li X, Hu C. Helicobacter pylori infection induces stem cell-like properties in Correa cascade of gastric cancer. Cancer Lett 2022; 542:215764. [PMID: 35654291 DOI: 10.1016/j.canlet.2022.215764] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 02/09/2023]
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer-related death. Its poor prognosis is attributed to unclear pathogenesis. Currently, the most widely accepted model for elucidating the mechanism of GC is the Correa cascade, which covers several histological lesions of the gastric mucosa. GC stem cells (CSCs) are crucial for oncogenesis in the Correa cascade and GC progression. As Helicobacter pylori (H. pylori) is the etiological factor in the Correa cascade, growing evidence suggests that enhancement of gastric stem cell-like properties and increase in CSCs correlate with H. pylori infection. In this paper, we review recent studies that present pathogenic mechanisms by which H. pylori induces gastric stem cell-like properties and CSCs, which may supplement the existing Correa model of GC. First, the dysfunction of developmental signaling pathways associated with H. pylori infection leads to the enhancement of gastric stemness. Second, H. pylori infection promotes alteration of the gastric mucosal microenvironment. In addition, epithelial-mesenchymal transition (EMT) may contribute to H. pylori-induced gastric stemness. Taken together, understanding these pathogeneses will provide potential therapeutic targets for the treatment of CSCs and malignant GC in H. pylori induced-Correa cascade of GC.
Collapse
Affiliation(s)
- JunJian He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiChao Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - ShengWei Zhang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - LiJiao He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiYan Chen
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - XinZhe Li
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - ChangJiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
225
|
Kametaka D, Iwamuro M, Takahashi T, Hirabata A, Hamada K, Kono Y, Kanzaki H, Kawano S, Tanaka T, Otsuka F, Kawahara Y, Okada H. Characterization of Gastric Tissue-Resident T Cells in Autoimmune and Helicobacter pylori-Associated Gastritis. Curr Issues Mol Biol 2022; 44:2443-2452. [PMID: 35735608 PMCID: PMC9221633 DOI: 10.3390/cimb44060167] [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: 04/22/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022] Open
Abstract
Data regarding the in-depth surface marker profiles of gastric tissue-resident lymphocytes in autoimmune and Helicobacter pylori-associated gastritis are lacking. In this study, we investigated potential differences in lymphocyte composition between these profiles. We enrolled patients with autoimmune (n = 14), active (current infection of H. pylori in the stomach; n = 10), and inactive gastritis (post-eradication of H. pylori; n = 20). Lymphocytes were isolated from the greater curvature of the stomach and lesser curvature of the body and analyzed using flow cytometry. The CD8+/CD3+ and CD4+/CD3+ ratios differed between the samples. Body CD4+/antrum CD4+, which is calculated by dividing the CD4+/CD3+ ratio in the body by that in the antrum, was significantly higher in autoimmune gastritis (3.54 ± 3.13) than in active (1.47 ± 0.41) and inactive gastritis (1.42 ± 0.77). Antrum CD8+/CD4+ in autoimmune gastritis (7.86 ± 7.23) was also higher than that in active (1.49 ± 0.58) and inactive gastritis (2.84 ± 2.17). The area under the receiver operating characteristic curve of antrum CD8+/CD4+ was 0.842, and the corresponding optimal cutoff point was 4.0, with a sensitivity of 71.4% and a specificity of 93.3%. We propose that an antrum CD8+/CD4+ ratio > 4.0 is a potential diagnostic marker for autoimmune gastritis.
Collapse
Affiliation(s)
- Daisuke Kametaka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
- Department of Gastroenterology, Iwakuni Clinical Center, 1-1-1 Atago-cho, Iwakuni, Yamaguchi 740-8510, Japan
| | - Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| | - Takahide Takahashi
- Division of Medical Support, Okayama University Hospital, Okayama 700-8558, Japan; (T.T.); (A.H.)
| | - Araki Hirabata
- Division of Medical Support, Okayama University Hospital, Okayama 700-8558, Japan; (T.T.); (A.H.)
| | - Kenta Hamada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| | - Yoshiyasu Kono
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| | - Hiromitsu Kanzaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| | - Seiji Kawano
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| | - Takehiro Tanaka
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Yoshiro Kawahara
- Department of Practical Gastrointestinal Endoscopy, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (D.K.); (K.H.); (Y.K.); (H.K.); (S.K.); (H.O.)
| |
Collapse
|
226
|
Lenti MV, Vanoli A, Miceli E, Arpa G, Di Stefano M, Soriano S, Capuano F, Gentile A, Aronico N, Coppola L, Pasini A, Luinetti O, Mauro A, Paulli M, Klersy C, Corazza GR, Di Sabatino A. Increase of Deep Intraepithelial Lymphocytes in the Oxyntic Mucosa of Patients With Potential and Overt Autoimmune Gastritis. Front Immunol 2022; 13:866167. [PMID: 35603187 PMCID: PMC9114815 DOI: 10.3389/fimmu.2022.866167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/31/2022] [Indexed: 01/23/2023] Open
Abstract
Pathological correlates of potential autoimmune gastritis (AIG), defined by anti-parietal cell antibody (PCA) positivity in the absence of gastric atrophy, have never been described. We herein aimed to assess intraepithelial lymphocyte (IEL) infiltration in gastric corpus of AIG patients. From 2000 to 2021, among 53 potential AIG patients, we focused on nine (median age 61 years, IQR 53-82; four females) who subsequently developed overt AIG. IEL infiltration of the oxyntic mucosa was assessed before and after developing overt AIG by measuring deep and superficial CD3+ IEL. AIG patients with different degrees of corpus atrophy, healthy controls (HC), active H. pylori gastritis, celiac disease (CD), and Hashimoto’s thyroiditis patients were included as controls. Of note, deep, but not superficial, CD3+ IEL count was higher (p<0.001) in potential AIG compared to HC and H. pylori gastritis. Deep CD3+ IEL infiltration did not change before or after the evolution into atrophy (median 9.6, IQR 8.8-12.4, vs 11.3, IQR 9.4-12.9). No difference was found in deep CD3+ IEL infiltration among potential, mild, and severe AIG, and compared to Hashimoto’s thyroiditis or CD. A deep CD3+ IEL cut-off of >7/100 epithelial cells allowed discrimination of any AIG stage and severity (AUC=0.842). We conclude that an increased deep CD3+ IEL infiltration of the oxyntic mucosa could represent a marker of potential AIG. Prospective studies including a larger number of potential AIG patients are needed.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Emanuela Miceli
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Giovanni Arpa
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Michele Di Stefano
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Simone Soriano
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Francesca Capuano
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Antonella Gentile
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Nicola Aronico
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Luigi Coppola
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandra Pasini
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Ombretta Luinetti
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Aurelio Mauro
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marco Paulli
- Department of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Catherine Klersy
- Unit of Clinical Epidemiology and Biometry, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| |
Collapse
|
227
|
Tsukanov VV, Smirnova OV, Kasparov EV, Sinyakov AA, Vasyutin AV, Tonkikh JL, Cherepnin MA. Dynamics of Oxidative Stress in Helicobacter pylori-Positive Patients with Atrophic Body Gastritis and Various Stages of Gastric Cancer. Diagnostics (Basel) 2022; 12:1203. [PMID: 35626358 PMCID: PMC9141138 DOI: 10.3390/diagnostics12051203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is a global health problem. The pathogenesis of this disease remains unclear. This study included 198 H. pylori (+) men aged 45 to 60 years old. Group A included 63 practically healthy men, group B included 45 men with severe atrophic body gastritis, group C included 37 men with epithelial gastric cancer stages I-II according to TNM, and group D included 54 men with epithelial gastric cancer stages III-IV according to the TNM scale. The content of malondialdehyde (MDA), diene conjugates (DCs), superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), and glutathione peroxidase (GPO) was detected using an enzyme immunoassay (ELISA) or spectrophotometric methods in the blood plasma. The concentrations of MDA and DC were increased in the patients of group B compared with group A, and in patients of groups C and D compared with groups A and B. The ratio of MDA/SOD and MDA/CAT was decreased in the patients in group D compared with the patients in group C, and was significantly higher compared with group A. The ratios of MDA/GPO and MDA/GST increased linearly and were at a maximum in groups C and D. Our work determined that indicators of oxidative stress may be the biochemical substrate, which brings together the various stages of the Correa cascade, and may explain disease progression. The dynamics of changes in the content of SOD and CAT in the plasma in patients with gastric cancer may be a target of future investigations.
Collapse
Affiliation(s)
- Vladislav Vladimirovich Tsukanov
- Scientific Research Institute of Medical Problems of the North, Federal Research Centre “Krasnoyarsk Science Centre” of the Siberian Branch of Russian Academy of Science, 660022 Krasnoyarsk, Russia; (O.V.S.); (E.V.K.); (A.A.S.); (A.V.V.); (J.L.T.); (M.A.C.)
| | | | | | | | | | | | | |
Collapse
|
228
|
Assa A, Borrelli O, Broekaert I, Saccomani MD, Dolinsek J, Martin-de-Carpi J, Mas E, Miele E, Sila S, Thomson M, Tzivinikos C, Benninga MA. Helicobacter pylori-negative Chronic Gastritis in Children: A Systematic Review. J Pediatr Gastroenterol Nutr 2022; 74:956-967. [PMID: 35175996 DOI: 10.1097/mpg.0000000000003414] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES To systematically review the current evidence on Helicobacter pylori-negative chronic gastritis including natural history, available therapies and outcomes. METHODS Articles providing data on the prevalence, treatment or outcomes of Helicobacter pylori-negative gastritis were identified through a systematic search in the MEDLINE and EMBASE databases. All original research articles from human studies until October 31, 2021, were included. RESULTS A total of 54 studies were included consisted of eosinophilic gastritis (n = 9), autoimmune gastritis (n = 11), collagenous gastritis (n = 16), focally enhanced gastritis (n = 6), lymphocytic gastritis (n = 5) and other causes including idiopathic gastritis and chronic renal failure related (n = 7). Most of the included studies were either cross-sectional or longitudinal cohorts except for collagenous gastritis, which mainly included case reports and case series. The prevalence of paediatric eosinophilic gastritis ranges between 5 and 7/100,000 and patients have generally favourable outcome with 50% to 70% clinical and histological response to either corticosteroids or elimination diets. Autoimmune gastritis and collagenous gastritis are extremely rare entities, commonly present with refractory iron deficiency anaemia, while lymphocytic gastritis is relatively common (10%-45%) in children with coeliac disease. Data on treatments and outcomes of autoimmune, collagenous, and focally enhanced gastritis are lacking with limited data implying poor response to therapy in the former 2 diagnoses. CONCLUSIONS Helicobacter pylori-negative gastritis is uncommonly reported, mainly in small cohorts, mixed adult-paediatric cohorts or as sporadic case reports. As common symptoms are not specific, thus not always result in an endoscopic evaluation, the true prevalence of these distinct disorders may be underestimated, and thus under reported.
Collapse
Affiliation(s)
- Amit Assa
- The Juliet Keidan institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, The Hebrew University, Jerusalem, Israel
| | - Osvaldo Borrelli
- Division of Neurogastroenterology & Motility, Department of Paediatric Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Ilse Broekaert
- Department of Paediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Jernej Dolinsek
- Department of pediatrics, University Medical Center Maribor, Ljubljanska 5, Maribor, Slovenia
| | - Javier Martin-de-Carpi
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Emmanuel Mas
- Unité de Gastroenterologie, Hepatologie, Nutrition et Maladies Héréditaires du Metabolisme, Hôpital des Enfants, and IRSD, Universite de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Sara Sila
- Referral Centre for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Mike Thomson
- Centre for Paediatric Gastroenterology, Sheffield Children's Hospital NHS Foundation Trust, Weston Bank, Sheffield, UK
| | - Christos Tzivinikos
- Paediatric Gastroenterology Department, Al Jalila Children's Specialty Hospital, Mohammed Bin Rashid University, Dubai Medical College, Dubai, United Arab Emirates
| | - Marc A Benninga
- Department ofPaediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
229
|
Kamada T, Maruyama Y, Monobe Y, Haruma K. Endoscopic features and clinical importance of autoimmune gastritis. Dig Endosc 2022; 34:700-713. [PMID: 34674318 DOI: 10.1111/den.14175] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022]
Abstract
Autoimmune gastritis (AIG) is a special type of chronic gastritis characterized by autoimmune disorders caused by cellular immunity, resulting in the destruction of parietal cells and production of antiparietal cell antibodies. Endoscopic findings of AIG are mainly characterized by corpus-dominant advanced atrophy. The antral area is generally considered to have no or mild atrophy; however, there are cases wherein the gastric mucosa is red or faded due to past infection with Helicobacter pylori or bile reflux. Currently, there are no diagnostic criteria for AIG in Japan, and it is important to make a diagnosis based on the presence of gastric autoantibodies and characteristic endoscopic and histological findings. AIG is associated with gastric cancer, neuroendocrine tumors (NETs), and other autoimmune diseases, such as thyroid diseases, anemia, and neurological symptoms due to impaired absorption of iron and vitamin B12 , and thus requires systemic treatment. The significance of diagnosing AIG is to include patients as a high-risk group for the development of gastric cancer and gastric NETs, provide an opportunity to detect autoimmune endocrine diseases, and initiate therapeutic intervention before anemia and neurological symptoms develop. It is important to pay close attention to the occurrence of AIG comorbidities not only at the time of AIG diagnosis but also during follow-up after detection.
Collapse
Affiliation(s)
- Tomoari Kamada
- Department of, Health Care Medicine, Kawasaki Medical School, Okayama, Japan
| | - Yasuhiko Maruyama
- Department of Gastroenterology, Fujieda Municipal General Hospital, Shizuoka, Japan
| | - Yasumasa Monobe
- Department of, Pathology, Kawasaki Medical School, Okayama, Japan
| | - Ken Haruma
- Department of, General Internal Medicine 2, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
230
|
Li Y, Zhou D, Liu Q, Zhu W, Ye Z, He C. Gene Polymorphisms of m6A Erasers FTO and ALKBH1 Associated with Susceptibility to Gastric Cancer. Pharmgenomics Pers Med 2022; 15:547-559. [PMID: 35669943 PMCID: PMC9166898 DOI: 10.2147/pgpm.s360912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Yue Li
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
| | - Dalei Zhou
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
| | - Qing Liu
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
| | - Weijie Zhu
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
| | - Zulu Ye
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
| | - Caiyun He
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China
- Correspondence: Caiyun He; Zulu Ye, Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road, Yuexiu District, Guangzhou, 510060, People’s Republic of China, Tel +86-18665593050; +86-15017590433, Fax +20-87340921, Email ;
| |
Collapse
|
231
|
Kawai Y, Sugimoto M, Hamada M, Iwata E, Niikura R, Nagata N, Fukuzawa M, Itoi T, Kawai T. Linked color imaging effectively detects the endoscopic atrophic border in transnasal endoscopy. J Clin Biochem Nutr 2022; 70:290-296. [PMID: 35692679 PMCID: PMC9130059 DOI: 10.3164/jcbn.21-145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022] Open
Abstract
In oral endoscopy, linked color imaging (LCI) detects atrophic border and gastric mucosal diseases better than white light imaging (WLI), but its usefulness in transnasal endoscopy has not been fully investigated. Here, we retrospectively compared WLI and LCI using the L*a*b* color space in images from 57 patients aged ≥20 years who had undergone transnasal endoscopy as part of a health check-up from May 2016 to January 2017. We measured color differences at the atrophic/non-atrophic and fundic/pyloric mucosal borders. Gastritis severity scored using the Kyoto classification of gastritis was similar between the two techniques. However, in patients with current and with past Helicobacter pylori infection, color difference at the atrophic border was greater with LCI (21.58 ± 6.97 and 27.34 ± 10.32, respectively) than with WLI [14.42 ± 5.95 (p = 0.004) and 17.9 ± 8.48 (p<0.001)]; in those never infected with Helicobacter pylori, color difference at the fundic/pyloric mucosal border was greater with LCI than with WLI (p<0.001). Because of its enhancement of atrophic border detection, we recommend linked color imaging as the method of choice for transnasal endoscopy in health check-ups, particularly for identifying people at high risk of gastric cancer.
Collapse
Affiliation(s)
- Yusuke Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Mitsushige Sugimoto
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Mariko Hamada
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Eri Iwata
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Ryota Niikura
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Masakatsu Fukuzawa
- Department of Gastroenterology, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takao Itoi
- Department of Gastroenterology, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
232
|
Esposito G, Dottori L, Pivetta G, Ligato I, Dilaghi E, Lahner E. Pernicious Anemia: The Hematological Presentation of a Multifaceted Disorder Caused by Cobalamin Deficiency. Nutrients 2022; 14:nu14081672. [PMID: 35458234 PMCID: PMC9030741 DOI: 10.3390/nu14081672] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
Pernicious anemia is still a neglected disorder in many medical contexts and is underdiagnosed in many patients. Pernicious anemia is linked to but different from autoimmune gastritis. Pernicious anemia occurs in a later stage of autoimmune atrophic gastritis when gastric intrinsic factor deficiency and consequent vitamin B12 deficiency may occur. The multifaceted nature of pernicious anemia is related to the important role of cobalamin, which, when deficient, may lead to several dysfunctions, and thus, the proteiform clinical presentations of pernicious anemia. Indeed, pernicious anemia may lead to potentially serious long-term complications related to micronutrient deficiencies and their consequences and the development of gastric cancer and type 1 gastric neuroendocrine tumors. When not recognized in a timely manner or when pernicious anemia is diagnosed with delay, these complications may be potentially life-threatening and sometimes irreversible. The current review aimed to focus on epidemiology, pathogenesis, and clinical presentations of pernicious anemia in an attempt to look beyond borders of medical specialties. It aimed to focus on micronutrient deficiencies besides the well-known vitamin B12 deficiency, the diagnostic approach for pernicious anemia, its long-term complications and optimal clinical management, and endoscopic surveillance of patients with pernicious anemia.
Collapse
|
233
|
Venerito M, Sulzer S, Jechorek D. [Clinical management of autoimmune gastritis]. Dtsch Med Wochenschr 2022; 147:451-459. [PMID: 35405749 DOI: 10.1055/a-1520-3562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Autoimmune gastritis (AIG) is a chronic immune-mediated inflammation of the gastric corpus/fundus mucosa leading to progressive atrophy of the oxyntic gastric glands (AOM) and their consecutive loss of function. Possible clinical consequences of AIG include iron deficiency anemia, pernicious anemia, gastric neuroendocrine tumors (gNET), and gastric adenocarcinoma. This article provides a review of interdisciplinary aspects of the diagnosis and treatment of AIG.
Collapse
|
234
|
Yin Z, Xiao S, Tian X, Yuan Z, Zhou L. The necessity and appropriate range of the diagnostic "gray zone" of 13C-urea breath test. Saudi J Gastroenterol 2022; 28:385-392. [PMID: 35259858 PMCID: PMC9752539 DOI: 10.4103/sjg.sjg_638_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The 13C-urea breath test (13C-UBT) is preferred for non-invasive detection of Helicobacter pylori (H. pylori); however, its accuracy drops when results fall between 2‰ and 6‰ (called the gray zone). This study aimed to evaluate the accuracy of 13C-UBT (cut-off point 4‰) between 2‰ and 6‰, find a more appropriate gray zone, and identify the factors influencing 13C-UBT. METHODS Patients with 13C-UBT results 2‰-6‰, over an eight-year period, were studied. H. pylori infection was diagnosed if patients were positive for either Warthin-Starry staining or quantitative real-time polymerase chain reaction (real-time PCR), and excluded if both were negative. Accuracy of 13C-UBT under different cut-off points was calculated, and the factors affecting 13C-UBT were analyzed. RESULTS A total of 208 patients were included, of whom 129 were H. pylori-positive. Sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV) of 13C-UBT were 71.32%, 83.54%, 64.08%, and 87.62%, respectively. When the cut-off point was changed to 2.15‰, the NPV of 13C-UBT reached a maximum (76.47%); when the cut-off point was changed to 4.95‰, PPV reached its maximum (93.22%). Therefore, the original gray zone (2‰-6‰) was adjusted to 2‰-4.95‰. Gastric antral intestinal metaplasia (OR = 3.055, 95% CI: 1.003-9.309) was an independent risk factor for false-negative 13C-UBT. CONCLUSIONS Accuracy of 13C-UBT over 2‰-6‰ was poor, and the gray zone was changed to 2‰-4.95‰. 13C-UBT results over 2‰-4.95‰ should be interpreted with caution during mass screening of H. pylori, especially for patients with gastric antral intestinal metaplasia.
Collapse
Affiliation(s)
- Zhihao Yin
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Xueli Tian
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China
| | - Ziying Yuan
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liya Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China,Address for correspondence: Prof. Liya Zhou, Department of Gastroenterology, Beijing Key Laboratory of Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, China. E-mail:
| |
Collapse
|
235
|
Pyloric Incompetence Associated with Helicobactor pylori Infection and Correlated to the Severity of Atrophic Gastritis. Diagnostics (Basel) 2022; 12:diagnostics12030572. [PMID: 35328125 PMCID: PMC8947545 DOI: 10.3390/diagnostics12030572] [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: 01/20/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Duodenogastric reflux (DGR) causes bile reflux gastritis (BRG) and may develop into gastric cancer. DGR is classified as primary in non-operated stomachs or secondary to surgical intervention. Primary DGR and Helicobacter pylori (H. pylori) infection are reportedly related. However, the mechanism is not fully understood. This study aimed to elucidate the relationship between H. pylori infection and pyloric incompetence in a non-operated stomach. A total of 502 non-operated participants who underwent an upper intestinal endoscopy were prospectively enrolled. Endoscopic findings (EAC, endoscopic atrophy classification; nodular gastritis; xanthoma; fundic gland polyp; and incompetence of pylorus), sex, age, gastrin, pepsinogen (PG) I and PG II levels were evaluated. PG I/PG II ratio, anti-H. pylori-Ab positivity, and atrophic gastritis status were significantly different between the normal and incompetent pylori (p = 0.043, <0.001, and 0.001, respectively). Open-type atrophic gastritis was significantly higher in the incompetent pylori. Incompetence of the pylorus and EAC were moderately correlated (Cramer’s V = 0.25). Multivariate analysis revealed that the presence of anti-H. pylori-Ab was the only independent factor associated with the incompetence of the pylorus, with an adjusted odds ratio of 2.70 (95% CI: 1.47−4.94, p = 0.001). In conclusion, pyloric incompetence was associated with H. pylori infection and moderately correlated to the severity of atrophic gastritis in non-operated stomachs.
Collapse
|
236
|
Huang MY, Liu XY, Shao Q, Zhang X, Miao L, Wu XY, Xu YX, Wang F, Wang HY, Zeng L, Deng L. Phosphoserine phosphatase as a prognostic biomarker in patients with gastric cancer and its potential association with immune cells. BMC Gastroenterol 2022; 22:1. [PMID: 34979926 PMCID: PMC8722028 DOI: 10.1186/s12876-021-02073-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Background Because of dismal prognosis in gastric cancer, identifying relevant prognostic factors is necessary. Phosphoserine phosphatase (PSPH) exhibits different expression patterns in many cancers and has been reported to affect the prognosis of patients with cancer. In this study, we examined the prognostic role of metabolic gene PSPH in gastric cancer based on the TCGA dataset and our hospital–based cohort cases. Methods We collected and analysed RNA-seq data of Pan-cancer and gastric cancer in the TCGA dataset and PSPH expression data obtained from immunohistochemical analysis of 243 patients with gastric cancer from Sun Yat-sen University cancer center. Further, Kaplan–Meier survival analysis and Cox analysis were used to assess the effect of PSPH on prognosis. The ESTIMATE and Cibersort algorithms were used to elucidate the relationship between PSPH and the abundance of immune cells using the TCGA dataset. Results We observed that PSPH expression displayed considerably high in gastric cancer and it was significantly associated with inferior prognosis (P = 0.043). Surprisingly, there was a significant relationship between lower immune scores and high expression of PSPH (P < 0.05). Furthermore, patients with a low amount of immune cells exhibited poor prognosis (P = 0.046). The expression of PSPH significantly increased in activated memory CD4 T cells, resting NK cells and M0 macrophages (P = 0.037, < 0.001, and 0.005, respectively). Conclusions This study highlighted that PSPH influences the prognosis of patients with gastric cancer, and this is associated with the infiltration of tumour immune cells, indicating that PSPH may be a new immune-related target for treating gastric cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-021-02073-0.
Collapse
Affiliation(s)
- Ma-Yan Huang
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiao-Yun Liu
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Qiong Shao
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xu Zhang
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, People's Republic of China
| | - Xiao-Yan Wu
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Yu-Xia Xu
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Hai-Yun Wang
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, People's Republic of China.,Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, People's Republic of China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, People's Republic of China.
| | - Ling Deng
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Dongfeng East Road 651, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
237
|
Kim YI, Choi IJ. Current Evidence for a Paradigm Shift in Gastric Cancer Prevention From Endoscopic Screening to Helicobacter pylori Eradication in Korea. J Gastric Cancer 2022; 22:169-183. [PMID: 35938364 PMCID: PMC9359887 DOI: 10.5230/jgc.2022.22.e22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Gastric cancer is prevalent in Korea and ranked as the third most common cancer in 2019, followed by lung and thyroid cancers. The National Cancer Screening Program (NCSP) for gastric cancer has been implemented in adults aged ≥40 since 1999 and involves endoscopic screening every 2 years. The beneficial effects of the current NCSP on early cancer detection, cost-effectiveness, and mortality reduction are evident. However, the screening program results in a large socioeconomic burden and the consumption of medical resources, as it focuses solely on secondary prevention (early detection) rather than primary prevention of cancer. Helicobacter pylori is defined as a group I carcinogen by the International Agency for Research on Cancer. Hence, its eradication has been suggested as an important primary gastric cancer prevention strategy. Well-designed randomized controlled trials involving high-risk groups (post-endoscopic resection of early gastric cancer and family history of gastric cancer) and long-term follow-up studies in the general population have provided high-quality evidence regarding the effects of H. pylori eradication on gastric cancer prevention. In this review, we discussed the evidences for a possible modification of the current gastric cancer secondary prevention strategy by introducing primary prevention through H. pylori eradication. Areas for future research to optimize primary prevention strategies were also suggested.
Collapse
Affiliation(s)
- Young-Il Kim
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Il Ju Choi
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| |
Collapse
|
238
|
Livzan MA, Gaus OV, Mozgovoi SI, Bordin DS. Chronic Autoimmune Gastritis: Modern Diagnostic Principles. Diagnostics (Basel) 2021; 11:2113. [PMID: 34829460 PMCID: PMC8621657 DOI: 10.3390/diagnostics11112113] [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: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/18/2022] Open
Abstract
This article summarizes and systematizes the available data from the literature on chronic autoimmune gastritis (CAG) in order to increase the awareness of specialists about the modern possibilities for diagnosing the disease, including its early stages. The clinical manifestation of the disease includes possible variants such as gastrointestinal, hematological (first of all, the formation of iron deficiency and B12-deficiency anemia), and neurological variants. Patients with chronic autoimmune gastritis are characterized by comorbidity with other autoimmune diseases. In this paper, data on the most informative serological markers for the diagnosis of CAG, as well as laboratory tests to detect micronutrient deficiencies, information on the characteristic changes in the gastric mucosa, and the prognosis of the disease, are presented. The diagnosis of CAG should be based on a multidisciplinary approach that combines a thorough analysis of a patient's complaints with a mandatory assessment of nutritional status, as well as the results of serological, endoscopic, and histological research methods.
Collapse
Affiliation(s)
- Maria A. Livzan
- Omsk Sate Medical University, 644099 Omsk, Russia; (M.A.L.); (O.V.G.); (S.I.M.)
| | - Olga V. Gaus
- Omsk Sate Medical University, 644099 Omsk, Russia; (M.A.L.); (O.V.G.); (S.I.M.)
| | - Sergei I. Mozgovoi
- Omsk Sate Medical University, 644099 Omsk, Russia; (M.A.L.); (O.V.G.); (S.I.M.)
| | - Dmitry S. Bordin
- A.S. Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
- Tver State Medical University, 170100 Tver, Russia
| |
Collapse
|
239
|
Hu S, Liu H, Zhang J, Li S, Zhou H, Gao Y. Effects and prognostic values of miR-30c-5p target genes in gastric cancer via a comprehensive analysis using bioinformatics. Sci Rep 2021; 11:20584. [PMID: 34663825 PMCID: PMC8523699 DOI: 10.1038/s41598-021-00043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/30/2021] [Indexed: 12/02/2022] Open
Abstract
Gastric cancer (GC) is a common cancer and the leading cause of cancer-related death worldwide. To improve the diagnosis and treatment of GC, it is necessary to identify new biomarkers by investigating the cellular and molecular mechanisms. In this study, miR-30c-5p expression was significantly down-regulated in GC tissues by comprehensive analysis using multiple databases. The target genes of miR-30c-5p with up-regulated expression level in GC were identified, including ADAM12 (a disintegrin and metalloproteinase12), EDNRA (the Endothelin receptor type A), STC1 (stanniocalcin 1), and CPNE8 (the calcium-dependent protein, copine 8). The expression level of ADAM12 was significantly related to depth of invasion (p = 0.036) in GC patients. The expression level of EDNRA was significantly related to grade (P = 0.003), depth of invasion (P = 0.019), and lymphatic metastasis (P = 0.001). The expression level of CPNE8 was significantly related to grade (P = 0.043) and TNM stage (P = 0.027).Gene set enrichment analysis showed that they might participate in GC progression through cancer-related pathways. CIBERSORT algorithm analysis showed that their expressions were related to a variety of tumor-infiltrating immune cells. The higher expression of those target genes might be the independent risk factor for poor survival of GC patients, and they might be potential prognostic markers in GC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huaifeng Liu
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Jinyan Zhang
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China.,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Shujing Li
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huadong Zhou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.,Department of Neurology, Army Medical Center of PLA, Chongqing, 400038, China
| | - Yu Gao
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, Anhui, China.
| |
Collapse
|
240
|
Rustgi SD, Bijlani P, Shah SC. Autoimmune gastritis, with or without pernicious anemia: epidemiology, risk factors, and clinical management. Therap Adv Gastroenterol 2021; 14:17562848211038771. [PMID: 34484423 PMCID: PMC8414617 DOI: 10.1177/17562848211038771] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/22/2021] [Indexed: 02/04/2023] Open
Abstract
Autoimmune gastritis (AIG) is a chronic immune-mediated, inflammatory condition that involves the destruction of the gastric oxyntic mucosa through the autoimmune-mediated loss of parietal cells, with replacement by atrophic and metaplastic tissue. Diagnosing AIG is important, given the need for ongoing clinical management and vigilance with respect to downstream complications, the most serious of which is gastric adenocarcinoma. Other clinical consequences include gastric neuroendocrine tumors, consequences related to decreased gastric acid and decreased intrinsic factor due to parietal cell destruction and antibodies against intrinsic factor (e.g. micronutrient deficiencies), as well as concomitant autoimmune disorders. Considering the prevalence of AIG and the potential for severe clinical outcomes, it is important to engage in efforts to reduce practice pattern variability related to diagnosis and management. Accordingly, herein, we review of the epidemiology, pathogenesis, clinical presentation of AIG, including both gastric and extragastric manifestations, and provide an overview of clinical management.
Collapse
Affiliation(s)
- Sheila D Rustgi
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Priyesha Bijlani
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shailja C Shah
- Section of Gastroenterology, VA San Diego Healthcare System, 3350 La Jolla Villa Drive, San Diego, CA 92161, USA
| |
Collapse
|