Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
PLoS One ; 16(8): e0255601, 2021.
Article in English | MEDLINE | ID: mdl-34379655

ABSTRACT

BACKGROUND: Gastric intestinal metaplasia (IM) can lead to gastric cancer. Until now, there have been limited studies of predictors for regression and progression of IM. This study aimed to determine risk factors associated with regression or progression of IM for guiding proper management and prevention of gastric cancer. METHODS: 2,025 patients undergoing gastroscopy in Thammasat University Hospital, Thailand were enrolled during September 2017-August 2019. Patients' data including baseline characteristics, laboratory results, and histopathology of gastric biopsies from University medical database were extensively reviewed. RESULTS: 2,025 patients had mean age of 61.3 years and 44.2% were males. Overall H. pylori prevalence was 47.5%. There were 1,551(76.6%) patients with chronic gastritis and 361(17.8%) with IM. Of 400 patients with chronic gastritis having follow-up endoscopy and repeated gastric biopsies, 104(26%) had persistent H. pylori infection and 27(26%) developed IM during mean follow-up time of 24 months. Persistent H. pylori infection was significantly associated with development of IM (OR 3.16, 95%CI 1.56-6.39, p = 0.001). Regression, persistence, and progression of IM were demonstrated in 57.3%, 39.2%, and 3.5% of patients, respectively. Age >65 years, persistent H. pylori infection, and diabetes mellitus were significantly associated with persistent IM or progression to dysplasia with OR 2.47(95%CI 1.33-4.61, p = 0.004), OR 2.64(95%CI 1.13-6.18, p = 0.025), and OR 2.54(95%CI 1.16-5.54, p = 0.019), respectively. Patients without H. pylori infection had more IM regression than patients with persistent infection (60.4%vs.39.4%, p = 0.035). Patients with persistent H. pylori infection significantly had higher IM progression to dysplasia (15.2%vs.2.1%; OR 11.15, 95%CI 1.18-105.24, p = 0.035) than noninfected. During 24 months of study, 30 patients (1.5%) were diagnosed with gastric cancer. CONCLUSION: Regression of IM could be achieved by successful H. pylori eradication. Persistent H. pylori infection was significantly associated with development and progression of IM to dysplasia. Age >65 years and diabetes mellitus were also significant predictors for IM progression.


Subject(s)
Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Helicobacter pylori/isolation & purification , Metaplasia/pathology , Precancerous Conditions/pathology , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Follow-Up Studies , Gastritis, Atrophic/epidemiology , Gastritis, Atrophic/microbiology , Helicobacter Infections/microbiology , Humans , Male , Metaplasia/epidemiology , Metaplasia/microbiology , Middle Aged , Precancerous Conditions/epidemiology , Precancerous Conditions/microbiology , Prevalence , Prognosis , Retrospective Studies , Risk Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/microbiology , Thailand/epidemiology , Young Adult
2.
Int J Med Sci ; 18(12): 2743-2751, 2021.
Article in English | MEDLINE | ID: mdl-34104107

ABSTRACT

Background: Cytokines and their gene variants are proven to play a role in pathogenic gastritis and carcinogenesis. The study assesses associations of the cytokine gene polymorphisms with extension of atrophic gastritis/intestinal metaplasia (AGIM) in patients without Helicobacter pylori infection on immunohistochemistry study. Methods: 224 adult consecutive patients undergoing an upper digestive endoscopy were included and grouped according to localization of AGIM: 37 patients with antrum-limited AGIM, 21 corpus-limited AGIM, 15 extended-AGIM (antrum and corpus) and 151 patients had no AGIM. Medical records of the patients were checked and a structured direct interview was applied in order to collect clinical data, including digestive symptoms. In all cases, IFN-γ +874T>A, TGF-ß1 +869T>C, TNF-α-308G>A and -238G>A, and IL-6 -174C>G polymorphisms were genotyped. Results: The mean age was significantly higher in the AGIM group, while the comorbidies were similar among patients with different localization of lesions or in patients without AGIM. There were no significant differences in digestive symptoms, nor in the consumption of non-steroidal anti-inflammatory drugs or proton pump inhibitor with the different extensions of AGIM. There was a significant association between oral anticoagulant consumption and localization of AGIM (P = 0.042), frequency being higher among patients with corpus-limited AGIM than those with no AGIM (P = 0.007, adjusted P = 0.041). TGF-ß1 +869T>C was less frequent among patients with corpus-limited AGIM (n=7, 33.3%) and extended AGIM (n=5, 33.3%) than in antrum-limited AGIM (n=25, 67.6%). There were no other significant differences regarding variant and wild genotype frequencies of IFN-γ +874T>A (86.5%, 81.0%, 86.7%, p=0.814), TNF-α-308G>A (35.1%, 28.6%, 53.3%, p=0.48) and IL-6 -174C>G (70.3%. 61.9%, 73.3% p=0.656) among patients with antrum-limited, corpus-limited or extended AGIM. TGF-ß1 +869T>C was associated with a decreased risk for corpus-affected AGIM (adjusted odds ratio: 0.42, 95% confidence interval: 0.19-0.93, P = 0.032). The dominant inheritance models no revealed significant association for IFN-γ +874T>A, TNF-α-308G>A and IL-6 -174C>G gene polymorphism and the risk of localization of AGIM. Conclusion: TGF-ß1 +869T>C gene polymorphism is associated with a decreased risk for corporeal localization of premalignant lesions, while IFN-γ +874T>A, TNF-α-308G>A and IL-6 -174C>G are not associated with the risk for AGIM in immunohistochemically H. pylori negative patients.


Subject(s)
Gastric Mucosa/pathology , Gastritis, Atrophic/epidemiology , Genetic Predisposition to Disease , Precancerous Conditions/epidemiology , Transforming Growth Factor beta1/genetics , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Biopsy , Female , Gastric Mucosa/microbiology , Gastritis, Atrophic/genetics , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/diagnosis , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Immunohistochemistry , Interferon-gamma/genetics , Interleukin-6/genetics , Male , Metaplasia/epidemiology , Metaplasia/genetics , Metaplasia/microbiology , Metaplasia/pathology , Middle Aged , Polymorphism, Single Nucleotide , Precancerous Conditions/genetics , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Protective Factors , Risk Assessment/statistics & numerical data , Tumor Necrosis Factor-alpha/genetics , Young Adult
3.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Article in English | MEDLINE | ID: mdl-33189701

ABSTRACT

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Subject(s)
Cell Transformation, Neoplastic/immunology , Colitis-Associated Neoplasms/immunology , Lipids/immunology , Precancerous Conditions/immunology , Stomach Neoplasms/immunology , Animals , Benzylamines/pharmacology , Benzylamines/therapeutic use , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/drug effects , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Disease Models, Animal , Epithelial Cells , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Gerbillinae , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Helicobacter pylori/isolation & purification , Humans , Lipids/antagonists & inhibitors , Metaplasia/immunology , Metaplasia/microbiology , Metaplasia/pathology , Mice , Mice, Transgenic , Organoids , Precancerous Conditions/drug therapy , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control
4.
Histopathology ; 78(1): 39-47, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33382492

ABSTRACT

Since the description of Helicobacter pylori (HP) as the most common cause of gastritis and its neoplastic complications, numerous articles have been written about the epidemiology, clinical features, diagnostic methods, histopathology, pathogenesis, molecular biology and treatment of this infection. This review focuses on those aspects of the infection that challenge the universality of the medical implications through the lens of evolutionary science applied to medicine. The divergent epidemiological and clinical outcomes observed in different populations and the possible beneficial aspects of the infection are discussed. Also reviewed are Correa's seminal contributions to our understanding of gastric cancer in particular and postinflammatory tumours in general, and the renewed interest in intestinal metaplasia and its clinical implications.


Subject(s)
Gastric Mucosa/pathology , Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Precancerous Conditions/pathology , Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/complications , Humans , Metaplasia/microbiology , Metaplasia/pathology , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
5.
Gastroenterology ; 160(4): 1106-1117.e3, 2021 03.
Article in English | MEDLINE | ID: mdl-33220252

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori eradication and endoscopic surveillance of gastric precancerous lesions are strategies to reduce gastric cancer (GC) risk. To our knowledge, this study is the longest prospective cohort of an H pylori eradication trial in a Hispanic population. METHODS: A total of 800 adults with precancerous lesions were randomized to anti-H pylori treatment or placebo. Gastric biopsy samples taken at baseline and 3, 6, 12, 16, and 20 years were assessed by our Correa histopathology score. A generalized linear mixed model with a participant-level random intercept was used to estimate the effect of H pylori status on the score over time. Logistic regression models were used to estimate progression by baseline diagnosis and to estimate GC risk by intestinal metaplasia (IM) subtype and anatomic location. RESULTS: Overall, 356 individuals completed 20 years of follow-up. Anti-H pylori therapy (intention-to-treat) reduced progression of the Correa score (odds ratio [OR], 0.59; 95% confidence interval [CI], 0.38-0.93). H pylori-negative status had a beneficial effect on the score over time (P = .036). Among individuals with IM (including indefinite for dysplasia) at baseline, incidence rates per 100 person-years were 1.09 (95% CI, 0.85-1.33) for low-grade/high-grade dysplasia and 0.14 (95% CI, 0.06-0.22) for GC. Incomplete-type (vs complete-type) IM at baseline presented higher GC risk (OR, 13.4; 95% CI, 1.8-103.8). Individuals with corpus (vs antrum-restricted) IM showed an OR of 2.1 (95% CI, 0.7-6.6) for GC. CONCLUSIONS: In a high-GC-risk Hispanic population, anti-H pylori therapy had a long-term beneficial effect against histologic progression. Incomplete IM is a strong predictor of GC risk.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gastric Mucosa/pathology , Helicobacter Infections/drug therapy , Precancerous Conditions/epidemiology , Stomach Neoplasms/prevention & control , Adult , Aged , Biopsy , Colombia/epidemiology , Disease Progression , Female , Follow-Up Studies , Gastric Mucosa/diagnostic imaging , Gastric Mucosa/microbiology , Gastroscopy/statistics & numerical data , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Incidence , Male , Metaplasia/diagnosis , Metaplasia/epidemiology , Metaplasia/microbiology , Metaplasia/pathology , Middle Aged , Precancerous Conditions/diagnosis , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Prospective Studies , Risk Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Treatment Outcome
6.
Gut ; 69(12): 2093-2112, 2020 12.
Article in English | MEDLINE | ID: mdl-33004546

ABSTRACT

OBJECTIVE: A global consensus meeting was held to review current evidence and knowledge gaps and propose collaborative studies on population-wide screening and eradication of Helicobacter pylori for prevention of gastric cancer (GC). METHODS: 28 experts from 11 countries reviewed the evidence and modified the statements using the Delphi method, with consensus level predefined as ≥80% of agreement on each statement. The Grading of Recommendation Assessment, Development and Evaluation (GRADE) approach was followed. RESULTS: Consensus was reached in 26 statements. At an individual level, eradication of H. pylori reduces the risk of GC in asymptomatic subjects and is recommended unless there are competing considerations. In cohorts of vulnerable subjects (eg, first-degree relatives of patients with GC), a screen-and-treat strategy is also beneficial. H. pylori eradication in patients with early GC after curative endoscopic resection reduces the risk of metachronous cancer and calls for a re-examination on the hypothesis of 'the point of no return'. At the general population level, the strategy of screen-and-treat for H. pylori infection is most cost-effective in young adults in regions with a high incidence of GC and is recommended preferably before the development of atrophic gastritis and intestinal metaplasia. However, such a strategy may still be effective in people aged over 50, and may be integrated or included into national healthcare priorities, such as colorectal cancer screening programmes, to optimise the resources. Reliable locally effective regimens based on the principles of antibiotic stewardship are recommended. Subjects at higher risk of GC, such as those with advanced gastric atrophy or intestinal metaplasia, should receive surveillance endoscopy after eradication of H. pylori. CONCLUSION: Evidence supports the proposal that eradication therapy should be offered to all individuals infected with H. pylori. Vulnerable subjects should be tested, and treated if the test is positive. Mass screening and eradication of H. pylori should be considered in populations at higher risk of GC.


Subject(s)
Helicobacter Infections/diagnosis , Helicobacter Infections/drug therapy , Stomach Neoplasms/microbiology , Stomach Neoplasms/prevention & control , Anti-Bacterial Agents/administration & dosage , Antimicrobial Stewardship , Clinical Decision-Making , Cost-Benefit Analysis , Delphi Technique , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Bacterial , Early Detection of Cancer , Endoscopy, Gastrointestinal , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/prevention & control , Gastroesophageal Reflux , Gastrointestinal Microbiome , Genetic Markers , Global Health , Helicobacter Infections/epidemiology , Helicobacter pylori , Humans , Metabolic Syndrome , Metaplasia/microbiology , Metaplasia/prevention & control , Proton Pump Inhibitors/administration & dosage , Reinfection , Stomach Neoplasms/epidemiology
7.
PLoS One ; 15(10): e0239434, 2020.
Article in English | MEDLINE | ID: mdl-33002050

ABSTRACT

BACKGROUND: Gastric intestinal metaplasia (IM) is precancerous lesion of gastric cancer related to H. pylori infection. There has been limited data about IM and associated risk factors. This study aimed to determine risk factors related to development of IM to guide proper management. METHODS: 1,370 patients undergoing UGI endoscopy at Thammasat University Hospital, Thailand were included between January 2018-August 2019. Patients' data including baseline characteristics, laboratory results, and histopathology from medical database were extensively reviewed. Immunohistochemical staining for p53 expression from gastric biopsies was also performed. RESULTS: Overall H. pylori prevalence was 43.8%. Mean age was 60.7 years and 45% of whom were males. Chronic gastritis was observed in 1,064(77.7%) patients, while 223(16.3%) had IM. Of 223 patients with IM, 194(87%) patients had complete IM, while 29 (13%) had incomplete IM. In groups of complete and incomplete IM, current H. pylori infection rates were 66.5% and 58.6%, respectively. The BMI of incomplete IM group(27.4) was significantly higher than BMI of complete IM group (23.6). Overweight and obese patients (BMI ≥23 kg/m2) were significantly associated with higher risk for the development of incomplete IM (OR 3.25; 95%CI 1.14-9.27, p = 0.027). Males, age >50 years, and current H. pylori infection were significantly higher in IM than chronic gastritis group with OR 1.43 (95%CI 1.01-2.03, p = 0.048), OR 1.67 (95% CI 1.08-2.57, p = 0.021), and OR 3.14 (95% CI 2.29-4.30, p<0.001), respectively. During 20 months of study, there were 15 patients (1.1%) diagnosed with gastric cancer and 1-year survival rate was only 60%. CONCLUSIONS: Males, age >50 years, and current H. pylori infection are significant predictors for the presence of intestinal metaplasia. BMI might be beneficial for using as a predictive risk factor to reduce the development of incomplete intestinal metaplasia. H. pylori eradication could be an effective way to prevent the development of gastric precancerous lesions.


Subject(s)
Helicobacter pylori/physiology , Stomach Neoplasms/epidemiology , Stomach/microbiology , Stomach/pathology , Aged , Cohort Studies , Female , Humans , Male , Metaplasia/microbiology , Middle Aged , Prevalence , Retrospective Studies , Risk Factors , Stomach Neoplasms/microbiology
8.
Int J Cancer ; 147(9): 2437-2445, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32363734

ABSTRACT

Helicobacter pylori (Hp) infects the stomach of about half of the human population and is strongly associated with the risk of gastric cancer (GC) and its premalignant precursors. The cag pathogenicity island (cagPAI) is a region of the Hp genome encoding for key molecular machinery involved in the infection process. Following a sequencing study, we selected 50 genetic polymorphisms located in seven cagPAI genes and tested their associations with the risk of advanced gastric premalignant lesions and GC in 1220 subjects from various Latin American populations showing the whole spectrum of phenotypes from gastritis to GC. We found that three polymorphisms of cagA are associated with the risk of advanced gastric premalignant lesions (incomplete intestinal metaplasia [ie, Type 2 and 3] or dysplasia), and that six polymorphisms located in cagA, cagL and cagI were associated with risk of GC. When corrected for multiple testing none of the associations were statistically significant. However, scores built by integrating the individual polymorphisms were significantly associated with the risk of advanced gastric premalignant lesions and GC. These results have the potential of establishing markers for risk stratification in the general population, in view of targeting Hp eradication to high-risk population groups.


Subject(s)
Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Precancerous Conditions/microbiology , Stomach Neoplasms/epidemiology , Adult , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Biopsy , Colombia/epidemiology , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Female , Gastric Mucosa/microbiology , Gastritis/microbiology , Gastritis/pathology , Genetic Markers , Genome, Bacterial/genetics , Genomic Islands , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Humans , Male , Metaplasia/microbiology , Metaplasia/pathology , Mexico/epidemiology , Middle Aged , Polymorphism, Genetic , Precancerous Conditions/pathology , Risk Assessment/methods , Risk Factors , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Whole Genome Sequencing
9.
PLoS One ; 15(3): e0230220, 2020.
Article in English | MEDLINE | ID: mdl-32163505

ABSTRACT

Helicobacter pylori is a Gram-negative bacterium that causes chronic atrophic gastritis and peptic ulcers and it has been associated with the development of gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT). One of the more remarkable characteristics of H. pylori is its ability to survive in the hostile environment of the stomach. H. pylori regulates the expression of specific sets of genes allowing it to survive high acidity levels and nutrient scarcity. In the present study, we determined the expression of virulence associated protein D (VapD) of H. pylori inside adenocarcinoma gastric (AGS) cells and in gastric biopsies. Using qRT-PCR, VapD expression was quantified in intracellular H. pylori-AGS cell cultures at different time points and in gastric mucosa biopsies from patients suffering from chronic atrophic gastritis, follicular gastritis, peptic ulcers, gastritis precancerous intestinal metaplasia and adenocarcinoma. Our results show that vapD of H. pylori presented high transcription levels inside AGS cells, which increased up to two-fold above basal values across all assays over time. Inside AGS cells, H. pylori acquired a coccoid form that is metabolically active in expressing VapD as a protection mechanism, thereby maintaining its permanence in a viable non-cultivable state. VapD of H. pylori was expressed in all gastric biopsies, however, higher expression levels (p = 0.029) were observed in gastric antrum biopsies from patients with follicular gastritis. The highest VapD expression levels were found in both antrum and corpus gastric biopsies from older patients (>57 years old). We observed that VapD in H. pylori is a protein that is only produced in response to interactions with eukaryotic cells. Our results suggest that VapD contributes to the persistence of H. pylori inside the gastric epithelial cells, protecting the microorganism from the intracellular environment, reducing its growth rate, enabling long-term infection and treatment resistance.


Subject(s)
Bacterial Proteins/genetics , Gastritis, Atrophic/etiology , Helicobacter pylori/genetics , Membrane Glycoproteins/genetics , Stomach/microbiology , Stomach/pathology , Adenocarcinoma/etiology , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Coculture Techniques/methods , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Gastroscopy/methods , Helicobacter Infections/complications , Helicobacter Infections/pathology , Humans , Intestines/microbiology , Intestines/pathology , Male , Metaplasia/microbiology , Metaplasia/pathology , Middle Aged , Peptic Ulcer/metabolism , Peptic Ulcer/pathology , Precancerous Conditions/etiology , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Pyloric Antrum/microbiology , Pyloric Antrum/pathology , Stomach Neoplasms/etiology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Virulence/genetics , Young Adult
10.
Rev Esp Patol ; 53(2): 79-87, 2020.
Article in English | MEDLINE | ID: mdl-32199598

ABSTRACT

The association between Helicobacter-pylori-induced inflammation and gastric adenocarcinoma is well documented and it has been suggested that the pro-mitotic and apoptotic effect of Cyclooxygenase-2 and Osteopontin on the epithelial cells of the gastric mucosa may have a role in carcinogenesis of the gastric mucosa. The aim of this study was to determine the expression of Cyclooxygenase-2 and Osteopontin in normal gastric mucosa, mucosa with gastritis and gastric mucosa with intestinal metaplasia, in relation to Helicobacter-pylori infection and grade of inflammation. Immunohistochemistry was performed on 108 gastric biopsies in order to detect Cyclooxygenase-2 and Osteopontin expression. The intensity and percentage of staining were evaluated using the H-Score, and its association with grade of inflammation, Helicobacter pylori infection and intestinal metaplasia was determined. Expression of Cyclooxygenase-2 and Osteopontin was higher in gastric biopsies (values shown respectively) with Helicobacter-pylori infection (179.9/142.3), intestinal metaplasia (208.8/179.3) or higher grades of inflammation (190/135.7) in comparison to normal gastric mucosa (100.7/80) or mild grade of inflammation (128.4/128.4), (p<0.05).There is an overexpression of Cyclooxygenase-2 and Osteopontin in gastric mucosa with H. pylori infection, intestinal metaplasia and high grades of inflammation, suggesting a constant up-regulation of protein expression in response to the inflammatory process generated by a Helicobacter-pylori infection, leading to the development of intestinal metaplasia.


Subject(s)
Cyclooxygenase 2/metabolism , Gastric Mucosa/metabolism , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Osteopontin/metabolism , Precancerous Conditions/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Chronic Disease , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/complications , Humans , Inflammation/metabolism , Inflammation/pathology , Intestines/pathology , Metaplasia/metabolism , Metaplasia/microbiology , Metaplasia/pathology , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Stomach/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Up-Regulation
11.
Gastroenterology ; 158(6): 1650-1666.e15, 2020 05.
Article in English | MEDLINE | ID: mdl-32032583

ABSTRACT

BACKGROUND & AIMS: Gastric chief cells, a mature cell type that secretes digestive enzymes, have been proposed to be the origin of metaplasia and cancer through dedifferentiation or transdifferentiation. However, studies supporting this claim have had technical limitations, including issues with the specificity of chief cell markers and the toxicity of drugs used. We therefore sought to identify genes expressed specifically in chief cells and establish a model to trace these cells. METHODS: We performed transcriptome analysis of Mist1-CreERT-traced cells, with or without chief cell depletion. Gpr30-rtTA mice were generated and crossed to TetO-Cre mice, and lineage tracing was performed after crosses to R26-TdTomato mice. Additional lineage tracing experiments were performed using Mist1-CreERT, Kitl-CreERT, Tff1-Cre, and Tff2-Cre mice crossed to reporter mice. Mice were given high-dose tamoxifen or DMP-777 or were infected with Helicobacter pylori to induce gastric metaplasia. We studied mice that expressed mutant forms of Ras in gastric cells, using TetO-KrasG12D, LSL-KrasG12D, and LSL-HrasG12V mice. We analyzed stomach tissues from GPR30-knockout mice. Mice were given dichloroacetate to inhibit pyruvate dehydrogenase kinase (PDK)-dependent cell competition. RESULTS: We identified GPR30, the G-protein-coupled form of the estrogen receptor, as a cell-specific marker of chief cells in gastric epithelium of mice. Gpr30-rtTA mice crossed to TetO-Cre;R26-TdTomato mice had specific expression of GPR30 in chief cells, with no expression noted in isthmus stem cells or lineage tracing of glands. Expression of mutant Kras in GPR30+ chief cells did not lead to the development of metaplasia or dysplasia but, instead, led to a reduction in labeled numbers of chief cells and a compensatory expansion of neck lineage, which was derived from upper Kitl+ clones. Administration of high-dose tamoxifen, DMP-777, or H pylori decreased the number of labeled chief cells. Chief cells were eliminated from epithelia via GPR30- and PDK-dependent cell competition after metaplastic stimuli, whereas loss of GRP30 or inhibition of PDK activity preserved chief cell numbers and attenuated neck lineage cell expansion. CONCLUSIONS: In tracing studies of mice, we found that most chief cells are lost during metaplasia and therefore are unlikely to contribute to gastric carcinogenesis. Expansion of cells that coexpress neck and chief lineage markers, known as spasmolytic polypeptide-expressing metaplasia, does not occur via dedifferentiation from chief cells but, rather, through a compensatory response from neck progenitors to replace the eliminated chief cells.


Subject(s)
Chief Cells, Gastric/physiology , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Azetidines/toxicity , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/physiology , Dichloroacetic Acid/administration & dosage , Disease Models, Animal , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Helicobacter Infections/microbiology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Metaplasia/chemically induced , Metaplasia/microbiology , Metaplasia/pathology , Mice , Mice, Knockout , Piperazines/toxicity , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/physiology , Tamoxifen/toxicity
13.
Gut ; 69(9): 1572-1580, 2020 09.
Article in English | MEDLINE | ID: mdl-31974133

ABSTRACT

OBJECTIVE: Helicobacter pylori is associated with gastric inflammation, precancerous gastric atrophy (GA) and intestinal metaplasia (IM). We aimed to identify microbes that are associated with progressive inflammation, GA and IM 1 year after H. pylori eradication. DESIGN: A total of 587 H. pylori-positive patients were randomised to receive H. pylori eradication therapy (295 patients) or placebo (292 patients). Bacterial taxonomy was analysed on 404 gastric biopsy samples comprising 102 pairs before and after 1 year H. pylori eradication and 100 pairs before and after 1 year placebo by 16S rRNA sequencing. RESULTS: Analysis of microbial sequences confirmed the eradication of H. pylori in treated group after 1 year. Principal component analysis revealed distinct microbial clusters reflected by increase in bacterial diversity (p<0.00001) after H. pylori eradication. While microbial interactions remained largely unchanged after placebo treatment, microbial co-occurrence was less in treated group. Acinetobacter lwoffii, Streptococcus anginosus and Ralstonia were enriched while Roseburia and Sphingomonas were depleted in patients with persistent inflammation 1 year after H. pylori eradication. A distinct cluster of oral bacteria comprising Peptostreptococcus, Streptococcus, Parvimonas, Prevotella, Rothia and Granulicatella were associated with emergence and persistence of GA and IM. Probiotic Faecalibacterium praustznii was depleted in subjects who developed GA following H. pylori eradication. Functional pathways including amino acid metabolism and inositol phosphate metabolism were enriched while folate biosynthesis and NOD-like receptor signalling decreased in atrophy/IM-associated gastric microbiota. CONCLUSION: This study identified that gastric microbes contribute to the progression of gastric carcinogenesis after H. pylori eradication.


Subject(s)
Bacteria , Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Metaplasia , Stomach , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/pathogenicity , Biopsy/methods , Biopsy/statistics & numerical data , Carcinogenesis , Disease Eradication/methods , Disease Eradication/statistics & numerical data , Disease Progression , Female , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/diagnosis , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Helicobacter pylori/isolation & purification , Humans , Male , Metaplasia/microbiology , Metaplasia/pathology , Microbial Interactions/drug effects , Middle Aged , Sequence Analysis, RNA/methods , Stomach/microbiology , Stomach/pathology
15.
Digestion ; 101(5): 624-630, 2020.
Article in English | MEDLINE | ID: mdl-31336366

ABSTRACT

INTRODUCTION: The diagnosis of Helicobacter pylori infection status with white light imaging (WLI) is difficult. We evaluated the accuracies of using WLI and linked color imaging (LCI) for diagnosing H. pylori-active gastritis in a multicenter prospective study setting. METHODS: Patients who underwent esophagogastroduodenoscopy were prospectively included. The image collection process was randomized and anonymous, and the image set included 4 images with WLI or 4 images with LCI in the corpus that 5 reviewers separately evaluated. Active gastritis was defined as positive when there was diffuse redness in WLI and crimson coloring in LCI. The H. pylori infection status was determined by the urea breath test and the serum antibody test. Cases in which both test results were negative but atrophy or intestinal metaplasia was histologically confirmed were defined as past infections. The primary endpoint was the diagnostic accuracies of WLI and LCI, and the secondary endpoint was inter-observer agreement. RESULTS: Data for 127 patients were analyzed. The endoscopic diagnostic accuracy for active gastritis was 79.5 (sensitivity of 84.4 and specificity of 74.6) with WLI and 86.6 (sensitivity of 84.4 and specificity of 88.9) with LCI (p = 0.029). LCI significantly improved the accuracy in patients with past infections over WLI (36.8 in WLI and 78.9 in LCI, p < 0.01). The κ values were 0.59 in WLI and 0.70 in LCI. CONCLUSIONS: LCI is useful for endoscopic diagnosis of H. pylori-active or inactive gastritis, and it is advantageous for patients with past infections of inactive gastritis.


Subject(s)
Gastric Mucosa/diagnostic imaging , Gastritis/diagnosis , Gastroscopy/methods , Helicobacter Infections/diagnosis , Image Enhancement/methods , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Breath Tests , Color , Feasibility Studies , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/blood , Gastritis/microbiology , Gastritis/pathology , Gastroscopy/instrumentation , Gastroscopy/statistics & numerical data , Helicobacter Infections/blood , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Helicobacter pylori/isolation & purification , Humans , Image Enhancement/instrumentation , Male , Metaplasia/blood , Metaplasia/diagnosis , Metaplasia/microbiology , Metaplasia/pathology , Middle Aged , Narrow Band Imaging/instrumentation , Narrow Band Imaging/methods , Narrow Band Imaging/statistics & numerical data , Prospective Studies
16.
Cell Mol Gastroenterol Hepatol ; 9(2): 257-276, 2020.
Article in English | MEDLINE | ID: mdl-31669263

ABSTRACT

BACKGROUND & AIMS: Gastric carcinoma is related mostly to CagA+-Helicobacter pylori infection, which disrupts the gastric mucosa turnover and elicits an epithelial-mesenchymal transition (EMT) and preneoplastic transdifferentiation. The tumor suppressor Hippo pathway controls stem cell homeostasis; its core, constituted by the large tumor suppressor 2 (LATS2) kinase and its substrate Yes-associated protein 1 (YAP1), was investigated in this context. METHODS: Hippo, EMT, and intestinal metaplasia marker expression were investigated by transcriptomic and immunostaining analyses in human gastric AGS and MKN74 and nongastric immortalized RPE1 and HMLE epithelial cell lines challenged by H pylori, and on gastric tissues of infected patients and mice. LATS2 and YAP1 were silenced using small interfering RNAs. A transcriptional enhanced associated domain (TEAD) reporter assay was used. Cell proliferation and invasion were evaluated. RESULTS: LATS2 and YAP1 appear co-overexpressed in the infected mucosa, especially in gastritis and intestinal metaplasia. H pylori via CagA stimulates LATS2 and YAP1 in a coordinated biphasic pattern, characterized by an early transient YAP1 nuclear accumulation and stimulated YAP1/TEAD transcription, followed by nuclear LATS2 up-regulation leading to YAP1 phosphorylation and targeting for degradation. LATS2 and YAP1 reciprocally positively regulate each other's expression. Loss-of-function experiments showed that LATS2 restricts H pylori-induced EMT marker expression, invasion, and intestinal metaplasia, supporting a role of LATS2 in maintaining the epithelial phenotype of gastric cells and constraining H pylori-induced preneoplastic changes. CONCLUSIONS: H pylori infection engages a number of signaling cascades that alienate mucosa homeostasis, including the Hippo LATS2/YAP1/TEAD pathway. In the host-pathogen conflict, which generates an inflammatory environment and perturbations of the epithelial turnover and differentiation, Hippo signaling appears as a protective pathway, limiting the loss of gastric epithelial cell identity that precedes gastric carcinoma development.


Subject(s)
Epithelial-Mesenchymal Transition/immunology , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Precancerous Conditions/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Animals , Cell Cycle Proteins/metabolism , Female , Gastric Mucosa/microbiology , Gene Expression Regulation, Neoplastic/immunology , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Male , Metaplasia/genetics , Metaplasia/microbiology , Metaplasia/pathology , Mice , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Protective Factors , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
20.
Gut ; 69(9): 1598-1607, 2020 09.
Article in English | MEDLINE | ID: mdl-31857433

ABSTRACT

OBJECTIVE: Gastrointestinal microbiota may be involved in Helicobacter pylori-associated gastric cancer development. The aim of this study was to explore the possible microbial mechanisms in gastric carcinogenesis and potential dysbiosis arising from H. pylori infection. DESIGN: Deep sequencing of the microbial 16S ribosomal RNA gene was used to investigate alterations in paired gastric biopsies and stool samples in 58 subjects with successful and 57 subjects with failed anti-H. pylori treatment, relative to 49 H. pylori negative subjects. RESULTS: In H. pylori positive subjects, richness and Shannon indexes increased significantly (both p<0.001) after successful eradication and showed no difference to those of negative subjects (p=0.493 for richness and p=0.420 for Shannon index). Differential taxa analysis identified 18 significantly altered gastric genera after eradication. The combination of these genera into a Microbial Dysbiosis Index revealed that the dysbiotic microbiota in H. pylori positive mucosa was associated with advanced gastric lesions (chronic atrophic gastritis and intestinal metaplasia/dysplasia) and could be reversed by eradication. Strong coexcluding interactions between Helicobacter and Fusobacterium, Neisseria, Prevotella, Veillonella, Rothia were found only in advanced gastric lesion patients, and were absent in normal/superficial gastritis group. Changes in faecal microbiota included increased Bifidobacterium after successful H. pylori eradication and more upregulated drug-resistant functional orthologs after failed treatment. CONCLUSION: H. pylori infection contributes significantly to gastric microbial dysbiosis that may be involved in carcinogenesis. Successful H. pylori eradication potentially restores gastric microbiota to a similar status as found in uninfected individuals, and shows beneficial effects on gut microbiota.


Subject(s)
Dysbiosis , Gastritis, Atrophic , Gastrointestinal Microbiome/genetics , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Anti-Bacterial Agents/therapeutic use , Biopsy/methods , Dysbiosis/diagnosis , Dysbiosis/microbiology , Feces/microbiology , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/drug therapy , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Humans , Male , Metaplasia/microbiology , Metaplasia/pathology , Microbial Interactions , Middle Aged , RNA, Ribosomal, 16S/isolation & purification , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...