Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 17.128
1.
Zhonghua Yi Xue Za Zhi ; 104(21): 1998-2002, 2024 Jun 04.
Article Zh | MEDLINE | ID: mdl-38825944

The study aims to summarize the clinical characteristics of patients with ectopic gastric mucosa in the small intestine, comparing clinical presentation differences between domestic and foreign patients through literature review. The clinical characteristics of cases diagnosed with ectopic gastric mucosa in the small intestine at Peking Union Medical College Hospital from January 2000 to January 2024 were retrospectively analyzed. By searching databanks, such as PubMed, EMBASE, the Cochrane Library, Wanfang, VIP, CNKI, and etc (the inclusion period was from the establishment of the database to January 1, 2024). The literature review was conducted on ectopic gastric mucosa in the small intestine. A total of 10 cases were included, all male, age [M (Q1, Q3)] was 27 (13-69) years old. Gastrointestinal bleeding was the first manifestation in most cases, with severe cases leading to hemorrhagic shock. Abdominal CT indicated local intestinal wall thickening and luminal narrowing in 3 cases. Four cases lesions were located at the beginning of the jejunum and 6 lesions were located in the end segment of ileum. All cases underwent local lesion resection, with postoperative pathology confirming ectopic gastric mucosa. Symptoms disappeared postoperatively, with a follow-up period of 0.5-3.0 years. Literature review indicates that the main clinical manifestation of gastric mucosa ectopia in the small intestine in China is gastrointestinal bleeding, while foreign patients are often complicated with intestinal duplication and intussusception, with abdominal pain and vomiting as the primary and main symptoms. The occurrence rate of intestinal obstruction in female patients, both domestically and abroad, is higher than that in male patients. The occurrence rate of ileal lesions with intestinal obstruction and small intestinal duplication is higher than that of duodenal lesions in both domestic and foreign patients. Local small intestine resection is an effective treatment method with generally good prognosis. Ectopic small intestinal mucosa is relatively rare, with symptoms of gastrointestinal bleeding and intestinal obstruction being common presentations, which can serve as one of the differential diagnoses for unexplained gastrointestinal bleeding.


Choristoma , Gastric Mucosa , Gastrointestinal Hemorrhage , Intestine, Small , Humans , Gastric Mucosa/pathology , Male , Adult , Middle Aged , Adolescent , Aged , Gastrointestinal Hemorrhage/etiology , Young Adult , Retrospective Studies , Female , China
2.
Sci Rep ; 14(1): 13003, 2024 06 06.
Article En | MEDLINE | ID: mdl-38844563

Helicobacter pylori eradication therapy reduces the risk of gastric cancer. However, it is unclear whether the severity of risk factors for gastric cancer such as atrophy and intestinal metaplasia are reduced after eradication in the long term. We aimed to study long-term changes in endoscopic risk factors for gastric cancer up to 20 years post-eradication. The endoscopic severity of gastritis according to the Kyoto Classification of Gastritis in 167 patients was retrospectively evaluated over an average follow-up 15.7 years. A significant improvement in mean total gastric cancer risk score (4.36 ± 1.66 to 2.69 ± 1.07, p < 0.001), atrophy (1.73 ± 0.44 to 1.61 ± 0.49, p = 0.004), and diffuse redness (1.22 ± 0.79 to 0.02 ± 0.13, p < 0.001) was observed compared to baseline in the Eradication group. However, there was no change in the never infection and current infection groups. The frequency of map-like redness increased over time until 15 years (3.6% to 18.7%, p = 0.03). The Cancer group had significantly higher risk scores at all time points. Endoscopic atrophy significantly improved in eradicated patients over long-term, suggested that eradication is one of the key elements in gastric cancer prevention. Individualized surveillance strategies based on endoscopic gastritis severity before eradication may be important for those at risk of gastric cancer.


Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Male , Helicobacter Infections/drug therapy , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Female , Helicobacter pylori/drug effects , Middle Aged , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/drug effects , Retrospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/microbiology , Aged , Adult , Risk Factors , Gastritis/microbiology , Gastritis/drug therapy , Gastritis/pathology , Gastroscopy , Follow-Up Studies , Anti-Bacterial Agents/therapeutic use
3.
BMC Gastroenterol ; 24(1): 157, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720287

BACKGROUND: At present, eradication regimens for non-Helicobacter pylori Helicobacter (NHPH) have not been established yet. We investigated effectiveness of the standard triple-drug combination therapy for Helicobacter pylori eradication and of a proton pump inhibitor (PPI) monotherapy in eradication of NHPH. METHODS: Subjects were the patients who were diagnosed with NHPH-infected gastritis based on microscopic findings, helical-shaped organisms obviously larger than Helicobacter pylori, in the gastric mucosal specimens using Giemsa staining at Kenwakai Hospital between November 2010 and September 2021, whose NHPH species were identified by polymerase chain reaction (PCR) analysis of urease genes in endoscopically-biopsied samples, and who consented to NHPH eradication with either the triple-drug combination therapy for one week or a PPI monotherapy for six months. Six months after the completion of eradication, its result was determined with esophagogastroduodenoscopy, microscopic examination, and PCR analysis. In cases of unsuccessful eradication, a second eradication with the other therapy was suggested to the patient. RESULTS: PCR analysis detected NHPH in 38 patients: 36 as Helicobacter suis and two as Helicobacter heilmannii/Helicobacter ailurogastricus. Fourteen Helicobacter suis-infected and one Helicobacter heilmannii/Helicobacter ailurogastricus-infected patients requested eradication therapy. The triple-drug combination therapy succeeded in four of five patients, while the PPI monotherapy succeeded in five of 10 patients. Three of five patients who had been unsuccessful with the latter therapy requested the triple-drug combination therapy as the second eradication and all three were successful. In total, the triple-drug combination therapy succeeded in seven out of eight (87.5%) attempted cases, while the PPI monotherapy in five out of 10 (50%) attempted cases. CONCLUSIONS: In NHPH eradication, the triple-drug combination therapy was considered to be effective to some extent and to become the first-line therapy. While, although less successful, PPI monotherapy appeared to be a potentially promising option particularly for patients with allergy or resistance to antibiotics. Effectiveness of PPI monotherapy may be attributed to hyperacid environment preference of Helicobacter suis and PPI's acid-suppressive effect. Additionally, male predominance in NHPH-infected gastritis patients may be explained by gender difference in gastric acid secretory capacity. However, further evidence needs to be accumulated. STUDY REGISTRATION: This study was approved by the Research Ethics Committee of Kenwakai Hospital (No. 2,017,024).


Anti-Bacterial Agents , Drug Therapy, Combination , Gastritis , Helicobacter Infections , Helicobacter heilmannii , Proton Pump Inhibitors , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/therapeutic use , Humans , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Male , Female , Middle Aged , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Gastritis/drug therapy , Gastritis/microbiology , Adult , Aged , Helicobacter heilmannii/isolation & purification , Helicobacter pylori/drug effects , Helicobacter pylori/isolation & purification , Amoxicillin/administration & dosage , Amoxicillin/therapeutic use , Clarithromycin/administration & dosage , Clarithromycin/therapeutic use , Helicobacter/isolation & purification , Helicobacter/drug effects , Treatment Outcome , Gastric Mucosa/microbiology , Gastric Mucosa/pathology
4.
Int J Biol Sci ; 20(7): 2658-2685, 2024.
Article En | MEDLINE | ID: mdl-38725851

Mucosal epithelial death is an essential pathological characteristic of portal hypertensive gastropathy (PHG). FADDosome can regulate mucosal homeostasis by controlling mitochondrial status and cell death. However, it remains ill-defined whether and how the FADDosome is involved in the epithelial death of PHG. The FADDosome formation, mitochondrial dysfunction, glycolysis process and NLRP3 inflammasome activation in PHG from both human sections and mouse models were investigated. NLRP3 wild-type (NLRP3-WT) and NLRP3 knockout (NLRP3-KO) littermate models, critical element inhibitors and cell experiments were utilized. The mechanism underlying FADDosome-regulated mitochondrial dysfunction and epithelial death in PHG was explored. Here, we found that FADD recruited caspase-8 and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) to form the FADDosome to promote Drp1-dependent mitochondrial fission and dysfunction in PHG. Also, FADDosome modulated NOX2 signaling to strengthen Drp1-dependent mitochondrial fission and alter glycolysis as well as enhance mitochondrial reactive oxygen species (mtROS) production. Moreover, due to the dysfunction of electron transport chain (ETC) and alteration of antioxidant enzymes activity, this altered glycolysis also contributed to mtROS production. Subsequently, the enhanced mtROS production induced NLRP3 inflammasome activation to result in the epithelial pyroptosis and mucosal injury in PHG. Thus, the FADDosome-regulated pathways may provide a potential therapeutic target for PHG.


Fas-Associated Death Domain Protein , Gastric Mucosa , Hypertension, Portal , Mitochondria , Animals , Mice , Mitochondria/metabolism , Fas-Associated Death Domain Protein/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Hypertension, Portal/metabolism , Hypertension, Portal/pathology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Knockout , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Inflammasomes/metabolism
5.
PLoS One ; 19(5): e0302015, 2024.
Article En | MEDLINE | ID: mdl-38728332

Nature has proven to be a treasure resource of bioactive metabolites. In this regard, Tamarix aphylla (F. Tamaricaceae) leaves crude extract was investigated for its gastroprotective effect against indomethacin-induced damage to the gastric mucosa. Additionally, phytochemical investigation of the methanolic extract afforded eight flavonoids' derivatives (1-8). On pharmacology networking study, the isolated compounds identified 123 unique targets where only 45 targets were related to peptic ulcer conditions, these 45 targets include 11 targets specifically correlate to gastric ulcer. The protein-protein interaction defined the PTGS2 gene as one of the highly interacted genes and the complete pharmacology network defined the PTGS2 gene as the most represented gene. The top KEGG signaling pathways according to fold enrichment analysis was the EGFR tyrosine kinase inhibitor resistance pathway. As a result, these findings highlighted the significance of using T. aphylla leaves crude extract as an anti-gastric ulcer candidate, which provides a safer option to chemical antisecretory medicines, which are infamous for their negative side effects. Our findings have illuminated the potent anti-inflammatory and antioxidant effects of T. aphylla, which are likely mediated by suppressing IL-1ß, IL-6, TNF-α, and MAPK signaling pathways, without compromising gastric acidity.


Indomethacin , MAP Kinase Signaling System , Oxidative Stress , Plant Extracts , Stomach Ulcer , Tamaricaceae , Stomach Ulcer/drug therapy , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Animals , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Indomethacin/adverse effects , Indomethacin/toxicity , Rats , Tamaricaceae/chemistry , MAP Kinase Signaling System/drug effects , Male , Plant Leaves/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Rats, Sprague-Dawley , Network Pharmacology , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Anti-Ulcer Agents/chemistry , Flavonoids/pharmacology , Flavonoids/chemistry
6.
Sci Rep ; 14(1): 10445, 2024 05 07.
Article En | MEDLINE | ID: mdl-38714774

Conventional endoscopy is widely used in the diagnosis of early gastric cancers (EGCs), but the graphical features were loosely defined and dependent on endoscopists' experience. We aim to establish a more accurate predictive model for infiltration depth of early gastric cancer including a standardized colorimetric system, which demonstrates promising clinical implication. A retrospective study of 718 EGC cases was performed. Clinical and pathological characteristics were included, and Commission Internationale de l'Eclariage (CIE) standard colorimetric system was used to evaluate the chromaticity of lesions. The predicting models were established in the derivation set using multivariate backward stepwise logistic regression, decision tree model, and random forest model. Logistic regression shows location, macroscopic type, length, marked margin elevation, WLI color difference and histological type are factors significantly independently associated with infiltration depth. In the decision tree model, margin elevation, lesion located in the lower 1/3 part, WLI a*color value, b*color value, and abnormal thickness in enhanced CT were selected, which achieved an AUROC of 0.810. A random forest model was established presenting the importance of each feature with an accuracy of 0.80, and an AUROC of 0.844. Quantified color metrics can improve the diagnostic precision in the invasion depth of EGC. We have developed a nomogram model using logistic regression and machine learning algorithms were also explored, which turned out to be helpful in decision-making progress.


Machine Learning , Neoplasm Invasiveness , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/diagnosis , Humans , Male , Female , Middle Aged , Retrospective Studies , Aged , Color , Gastric Mucosa/pathology , Gastric Mucosa/diagnostic imaging , Early Detection of Cancer/methods , Logistic Models , Gastroscopy/methods , Decision Trees
7.
World J Gastroenterol ; 30(17): 2308-2310, 2024 May 07.
Article En | MEDLINE | ID: mdl-38813049

Gastric cystica profunda (GCP) is an uncommon but underestimated gastric lesion. Its precancerous potential determines its significance. In addition to previous mucosa injury due to operations, biopsy or polypectomy, chronic active and atrophic gastritis may also lead to the development of GCPs. By carefully examining the stomach and taking biopsy samples from the susceptible regions, the stage of atrophy can be determined. Chronic atrophic gastritis is a risk factor for cancer evolvement and it can also contribute to GCPs formation. GCPs frequently occur close to early gastric cancers (EGCs) or EGC can arise from the cystic glands. Endoscopic resection is an effective and minimally invasive treatment in GCP.


Gastric Mucosa , Gastritis, Atrophic , Precancerous Conditions , Stomach Neoplasms , Humans , Biopsy , Chronic Disease , Cysts/surgery , Cysts/pathology , Cysts/etiology , Gastric Mucosa/pathology , Gastric Mucosa/surgery , Gastric Mucosa/diagnostic imaging , Gastritis, Atrophic/pathology , Gastritis, Atrophic/complications , Gastritis, Atrophic/surgery , Gastroscopy , Precancerous Conditions/pathology , Precancerous Conditions/surgery , Precancerous Conditions/etiology , Risk Factors , Stomach Diseases/etiology , Stomach Diseases/surgery , Stomach Diseases/pathology , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/etiology
8.
J Transl Med ; 22(1): 468, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760813

BACKGROUND: Gastric intestinal metaplasia (GIM) is an essential precancerous lesion. Although the reversal of GIM is challenging, it potentially brings a state-to-art strategy for gastric cancer therapeutics (GC). The lack of the appropriate in vitro model limits studies of GIM pathogenesis, which is the issue this work aims to address for further studies. METHOD: The air-liquid interface (ALI) model was adopted for the long-term culture of GIM cells in the present work. This study conducted Immunofluorescence (IF), quantitative real-time polymerase chain reaction (qRT-PCR), transcriptomic sequencing, and mucoproteomic sequencing (MS) techniques to identify the pathways for differential expressed genes (DEGs) enrichment among different groups, furthermore, to verify novel biomarkers of GIM cells. RESULT: Our study suggests that GIM-ALI model is analog to the innate GIM cells, which thus can be used for mucus collection and drug screening. We found genes MUC17, CDA, TRIM15, TBX3, FLVCR2, ONECUT2, ACY3, NMUR2, and MAL2 were highly expressed in GIM cells, while GLDN, SLC5A5, MAL, and MALAT1 showed down-regulated, which can be used as potential biomarkers for GIM cells. In parallel, these genes that highly expressed in GIM samples were mainly involved in cancer-related pathways, such as the MAPK signal pathway and oxidative phosphorylation signal pathway. CONCLUSION: The ALI model is validated for the first time for the in vitro study of GIM. GIM-ALI model is a novel in vitro model that can mimic the tissue micro-environment in GIM patients and further provide an avenue for studying the characteristics of GIM mucus. Our study identified new markers of GIM as well as pathways associated with GIM, which provides outstanding insight for exploring GIM pathogenesis and potentially other related conditions.


Metaplasia , Humans , Air , Models, Biological , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Stomach/pathology , Organoids/pathology , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Transcriptome/genetics , Intestines/pathology
10.
Curr Oncol ; 31(5): 2769-2779, 2024 05 14.
Article En | MEDLINE | ID: mdl-38785491

Gastric cancer is the fifth most common cancer and the fourth cause of global cancer mortality. The identification of new biomarkers and drug targets is crucial to allow the better prognosis and treatment of patients. The mitotic spindle positioning (MISP) protein has the function of correcting mitotic spindle positioning and centrosome clustering and has been implicated in the cytokinesis and migration of cancer cells. The goal of this work was to evaluate the expression and clinical relevance of MISP in gastric cancer. MISP expression was evaluated by immunohistochemistry in a single hospital series (n = 286) of gastric adenocarcinomas and compared with normal gastric mucosa and intestinal metaplasia, a preneoplastic lesion. MISP was detected on the membrane in 83% of the cases, being overexpressed in gastric cancer compared to normal gastric mucosa (n = 10). Its expression was negatively associated with diffuse and poorly cohesive types. On the other hand, it was strongly expressed in intestinal metaplasia where it was associated with MUC2 and CDX2 expression. Furthermore, when we silenced MISP in vitro, a significant decrease in the viability of gastric carcinoma cells was observed. In conclusion, MISP is overexpressed in gastric cancer, being associated with an intestinal phenotype in gastric carcinogenesis and having a role in cellular proliferation.


Metaplasia , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Male , Female , Middle Aged , Aged , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Biomarkers, Tumor , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor
11.
Helicobacter ; 29(3): e13097, 2024.
Article En | MEDLINE | ID: mdl-38819071

BACKGROUND: Helicobacter pylori (H. pylori) can evade the host's immune response and persist for a long time on the gastric mucosa. T helper (Th) cells appear to be involved in the control of H. pylori bacteria but promote mucosal inflammation. In contrast, regulatory T cells (Tregs) may reduce inflammation but promote H. pylori persistence. CC motif chemokine receptor 6 (CCR6) is involved in the migration of various cells into inflamed gastric mucosa. In this study, we examined CCR6+ Th cells and CCR6+ Tregs during H. pylori infection in humans. MATERIALS AND METHODS: Isolation of cells from blood and mucosal biopsies, magnetic separation of В cells, CD4+ and CD4+CCR6+CD45RO+ T cells, antigen-specific activation, B cell response in vitro, flow cytometry, determination of CD4+CD25hiFoxP3+ Tregs and various groups of Th cells. RESULTS: CD4+CCR6+ blood lymphocytes from healthy donors included Th cells and Tregs. These CCR6+ Th cells produced proinflammatory cytokines and also stimulated plasma cell maturation and antibody production in vitro. H. pylori gastritis and peptic ulcer disease were associated with an increase in the number of circulate CD4+CCR6+CD45RO+ cells and the percentage of Th1, Th17 and Th1/17 cells in this lymphocyte subgroup. In H. pylori-positive patients, circulating CD4+CCR6+ cells contained a higher proportion of H. pylori-specific cells compared with their CD4+CCR6- counterparts. H. pylori infection strongly increased the content of CD4+ lymphocytes in the inflamed gastric mucosa, with the majority of these CD4+ lymphocytes expressing CCR6. CD4+CCR6+ lymphocytes from H. pylori-infected stomach included Tregs and in vivo activated T cells, some of which produced interferon-γ without ex vivo stimulation. CONCLUSION: H. pylori infection causes an increase in the number of mature CD4+CCR6+ lymphocytes in the blood, with a pro-inflammatory shift in their composition and enrichment of the gastric mucosa with CD4+CCR6+ lymphocytes, including CCR6+ Th1 cells and Tregs.


Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Receptors, CCR6 , T-Lymphocytes, Regulatory , Humans , Receptors, CCR6/metabolism , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , T-Lymphocytes, Regulatory/immunology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter pylori/immunology , T-Lymphocytes, Helper-Inducer/immunology , Male , Female , Adult , Middle Aged , Flow Cytometry , Aged
13.
BMC Gastroenterol ; 24(1): 187, 2024 May 29.
Article En | MEDLINE | ID: mdl-38811868

BACKGROUND: Proton-pump inhibitors (PPIs) prevent aspirin-associated gastric and duodenal mucosal damage. However, long-term use of PPIs can lead to various adverse reactions, such as gastric polyps and enterochromaffin-like cell hyperplasia. Current research indicates that the abovementioned adverse reactions are mainly related to hypergastrinemia. We investigated whether low-frequency administration of omeprazole could effectively repair aspirin-induced mucosal damage and reduce the increase in gastrin levels associated with long-term use of PPIs. METHODS: Sprague‒Dawley rats were divided into four treatment groups: daily aspirin, daily aspirin and omeprazole once every day (qd), daily aspirin and omeprazole once every other day (qod), and daily aspirin and omeprazole once every three days (1/d3). After 15 days of feeding, blood samples were collected, and the stomachs of sacrificed rats were subjected to macroscopic, histological, and immunohistochemical studies. Moreover, in clinical practice, patients with peptic ulcers caused by aspirin took a standard dose of omeprazole (20 mg) every other day. Two months later, gastroscopy was performed to examine the healing of the ulcers. RESULTS: Both the omeprazole qd and omeprazole qod administrations effectively prevented aspirin-induced gastric peptic ulcers, with no significant difference between the two groups in the inhibition of parietal cell secretion of gastric acid and cell apoptosis. However, omeprazole 1/d3 failed to completely prevent aspirin-induced gastric mucosal injury. Notably, the gastrin levels, cell proliferation ability and cholecystokinin B receptor expression of the omeprazole qd group were significantly higher than those of the omeprazole qod group. In clinical work, patients with peptic ulcers caused by aspirin were given a standard dose of omeprazole every other day, and their ulcers healed after 2 months, as observed by gastroscopy. CONCLUSIONS: Omeprazole administration once every other day can effectively prevent aspirin-induced peptic ulcers and reduce hypergastrinemia, which may reduce the long-term adverse effects of PPI treatment.


Aspirin , Gastric Mucosa , Gastrins , Omeprazole , Proton Pump Inhibitors , Rats, Sprague-Dawley , Animals , Aspirin/adverse effects , Aspirin/administration & dosage , Omeprazole/pharmacology , Omeprazole/administration & dosage , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/administration & dosage , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastrins/blood , Male , Rats , Drug Administration Schedule , Humans , Peptic Ulcer/prevention & control , Peptic Ulcer/chemically induced , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Stomach Ulcer/prevention & control , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology
14.
Drug Des Devel Ther ; 18: 1651-1672, 2024.
Article En | MEDLINE | ID: mdl-38774485

Background: The Zuojin Pill (ZJP) is widely used for treating chronic atrophic gastritis (CAG) in clinical practice, effectively ameliorating symptoms such as vomiting, pain, and abdominal distension in patients. However, the underlying mechanisms of ZJP in treating CAG has not been fully elucidated. Purpose: This study aimed to clarify the characteristic function of ZJP in the treatment of CAG and its potential mechanism. Methods: The CAG model was established by alternant administrations of ammonia solution and sodium deoxycholate, as well as an irregular diet. Therapeutic effects of ZJP on body weight, serum biochemical indexes and general condition were analyzed. HE staining and AB-PAS staining were analyzed to characterize the mucosal injury and the thickness of gastric mucosa. Furthermore, network pharmacology and molecular docking were used to predict the regulatory mechanism and main active components of ZJP in CAG treatment. RT-PCR, immunohistochemistry, immunofluorescence and Western blotting were used to measure the expression levels of apoptosis-related proteins, gastric mucosal barrier-associated proteins and PI3K/Akt signaling pathway proteins. Results: The results demonstrated that ZJP significantly improved the general state of CAG rats, alleviated weight loss and gastric histological damage and reduced the serum biochemical indicators. Network pharmacology and molecular docking found that ZJP in treating CAG by inhibiting inflammation, suppressing apoptosis, and protecting the gastric mucosal barrier via the PI3K/Akt signaling pathway. Further experiments confirmed that ZJP obviously modulated the expression of key proteins involved in gastric mucosal cell apoptosis, such as Bax, Bad, Apaf-1, cleaved-caspase-3, cleaved-caspase-9, Cytochrome C, Bcl-2, and Bcl-xl. Moreover, ZJP significantly reversed the protein expression of Occludin, ZO-1, Claudin-4 and E-cadherin. Conclusion: Our study revealed that ZJP treats CAG by inhibiting the PI3K/Akt signaling pathway. This research provided a scientific basis for the rational use of ZJP in clinical practice.


Disease Models, Animal , Drugs, Chinese Herbal , Gastric Mucosa , Gastritis, Atrophic , Molecular Docking Simulation , Rats, Sprague-Dawley , Animals , Gastritis, Atrophic/drug therapy , Gastritis, Atrophic/pathology , Gastritis, Atrophic/metabolism , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Male , Chronic Disease , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis/drug effects , Network Pharmacology , Proto-Oncogene Proteins c-akt/metabolism
15.
World J Gastroenterol ; 30(16): 2220-2232, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38690017

Several features of drug-induced mucosal alterations have been observed in the upper gastrointestinal tract, i.e., the esophagus, stomach, and duodenum. These include pill-induced esophagitis, desquamative esophagitis, worsening of gastroesophageal reflux, chemotherapy-induced esophagitis, proton pump inhibitor-induced gastric mucosal changes, medication-induced gastric erosions and ulcers, pseudomelanosis of the stomach, olmesartan-related gastric mucosal inflammation, lanthanum deposition in the stomach, zinc acetate hydrate tablet-induced gastric ulcer, immune-related adverse event gastritis, olmesartan-asso-ciated sprue-like enteropathy, pseudomelanosis of the duodenum, and lanthanum deposition in the duodenum. For endoscopists, acquiring accurate knowledge regarding these diverse drug-induced mucosal alterations is crucial not only for the correct diagnosis of these lesions but also for differential diag-nosis of other conditions. This minireview aims to provide essential information on drug-induced mucosal alterations observed on esophagogastroduodenoscopy, along with representative endoscopic images.


Endoscopy, Digestive System , Humans , Endoscopy, Digestive System/methods , Gastric Mucosa/pathology , Gastric Mucosa/drug effects , Gastric Mucosa/diagnostic imaging , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/diagnostic imaging , Proton Pump Inhibitors/adverse effects , Esophageal Mucosa/pathology , Esophageal Mucosa/drug effects , Esophageal Mucosa/diagnostic imaging
16.
Asian J Endosc Surg ; 17(3): e13324, 2024 Jul.
Article En | MEDLINE | ID: mdl-38804100

BACKGROUND: One anastomosis gastric bypass (OAGB) is now the third most common bariatric surgery worldwide. This procedure is garnering increasing attention, but its complication of bile reflux and the associated risk of gastric carcinogenesis remains controversial. OBJECTIVE: The study aims to assess the impact of bile reflux on the gastric mucosa by comparing pathological and immunohistochemical results of gastric mucosa before and 2 years after OAGB surgery. METHODS: This retrospective study analyzed gastric lesions observed in gastroscopy before and after OAGB surgery. Pathological examinations were conducted on mucosal samples from proximal, middle and distal part of stomach, with a particular focus on the expression of Ki-67, P53, and CDX2 in immunohistochemistry. Ki-67 indicates cellular proliferation, P53 is a tumor suppressor protein, and CDX2 is a marker for intestinal differentiation. RESULTS: A total of 16 patients completed the follow-up. Regarding gastritis, presurgery nonerosive gastritis was found in two cases (12.5%), and postsurgery in six cases (37.5%). Erosive gastritis increased from one case (6.2%) presurgery to three cases (18.7%) postsurgery, totaling an increase from three to nine cases (p = .028). Bile reflux in the stomach increased from one case (6.2%) presurgery to three cases (18.7%) postsurgery. Most lesions in the proximal, middle, and distal part of stomach were relatively mild, with normal tissue states being predominant. Mild inflammation was found in all three areas, whereas moderate inflammation, intestinal metaplasia, and glandular atrophy were less common. No cases of severe inflammation were noted. The expression of gastric biomarkers CDX-2, Ki67, and P53 showed no significant statistical variation in different areas. CONCLUSION: Bile reflux does occur after OAGB, but its incidence is not high. Based on the immunohistochemical and pathological results of the gastric mucosa 2 years post-OAGB, there seems to be no significant causal relationship between OAGB and oncogenic inflammation around the gastric tube.


Gastric Bypass , Gastric Mucosa , Immunohistochemistry , Humans , Retrospective Studies , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/surgery , Female , Male , Gastric Bypass/adverse effects , Middle Aged , Adult , Bile Reflux/metabolism , Bile Reflux/pathology , Bile Reflux/etiology , CDX2 Transcription Factor/metabolism , Ki-67 Antigen/metabolism , Ki-67 Antigen/analysis , Tumor Suppressor Protein p53/metabolism , Gastritis/pathology , Gastritis/metabolism , Gastritis/etiology , Postoperative Complications/metabolism , Postoperative Complications/pathology , Postoperative Complications/etiology , Gastroscopy , Aged
17.
Cell Commun Signal ; 22(1): 263, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730482

BACKGROUND: Helicobacter pylori (H. pylori) is the predominant etiological agent of gastritis and disrupts the integrity of the gastric mucosal barrier through various pathogenic mechanisms. After H. pylori invades the gastric mucosa, it interacts with immune cells in the lamina propria. Macrophages are central players in the inflammatory response, and H. pylori stimulates them to secrete a variety of inflammatory factors, leading to the chronic damage of the gastric mucosa. Therefore, the study aims to explore the mechanism of gastric mucosal injury caused by inflammatory factors secreted by macrophages, which may provide a new mechanism for the development of H. pylori-related gastritis. METHODS: The expression and secretion of CCL3 from H. pylori infected macrophages were detected by RT-qPCR, Western blot and ELISA. The effect of H. pylori-infected macrophage culture medium and CCL3 on gastric epithelial cells tight junctions were analyzed by Western blot, immunofluorescence and transepithelial electrical resistance. EdU and apoptotic flow cytometry assays were used to detect cell proliferation and apoptosis levels. Dual-luciferase reporter assays and chromatin immunoprecipitation assays were used to study CCL3 transcription factors. Finally, gastric mucosal tissue inflammation and CCL3 expression were analyzed by hematoxylin and eosin staining and immunohistochemistry. RESULTS: After H. pylori infection, CCL3 expressed and secreted from macrophages were increased. H. pylori-infected macrophage culture medium and CCL3 disrupted gastric epithelial cells tight junctions, while CCL3 neutralizing antibody and receptor inhibitor of CCL3 improved the disruption of tight junctions between cells. In addition, H. pylori-infected macrophage culture medium and CCL3 recombinant proteins stimulated P38 phosphorylation, and P38 phosphorylation inhibitor improved the disruption of tight junctions between cells. Besides, it was identified that STAT1 was a transcription factor of CCL3 and H. pylori stimulated macrophage to secret CCL3 through the JAK1-STAT1 pathway. Finally, after mice were injected with murine CCL3 recombinant protein, the gastric mucosal injury and inflammation were aggravated, and the phosphorylation level of P38 was increased. CONCLUSIONS: In summary, our findings demonstrate that H. pylori infection stimulates macrophages to secrete CCL3 via the JAK1-STAT1 pathway. Subsequently, CCL3 damages gastric epithelial tight junctions through the phosphorylation of P38. This may be a novel mechanism of gastric mucosal injury in H. pylori-associated gastritis.


Chemokine CCL3 , Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Macrophages , Helicobacter pylori/physiology , Chemokine CCL3/metabolism , Chemokine CCL3/genetics , Animals , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mice , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Homeostasis , Mice, Inbred C57BL , Humans , Apoptosis , Cell Proliferation , Male , RAW 264.7 Cells
18.
Toxicol Appl Pharmacol ; 486: 116950, 2024 May.
Article En | MEDLINE | ID: mdl-38701902

Antidepressant duloxetine has been shown protective effect on indomethacin-induced gastric ulcer, which was escorted by inflammation in the gastric mucosa. Cytokines are the principal mediators of inflammation. Thus, by screening the differential expression of cytokines in the gastric mucosa using cytokine array at 3 h after indomethacin exposure, when the gastric ulcer began to format, we found that indomethacin increased cytokines which promoted inflammation responses, whereas duloxetine decreased pro-inflammatory cytokines increased by indomethacin and increased RANTES expression. RANTES was consistently increased by pretreated with both 5 mg/kg and 20 mg/kg duloxetine at 3 h and 6 h after indomethacin exposure in male rats. Selective blockade of RANTES-CCR5 axis by a functional antagonist Met-RANTES or a CCR5 antagonist maraviroc suppressed the protection of duloxetine. Considering the pharmacologic action of duloxetine on reuptake of monoamine neurotransmitters, we examined the serotonin (5-HT), norepinephrine and dopamine contents in the blood and discovered 20 mg/kg duloxetine increased 5-HT levels in platelet-poor plasma, while treatment with 5-HT promoted expression of RANTES in the gastric mucosa and alleviated the indomethacin-induced gastric injury. Furthermore, duloxetine activated PI3K-AKT-VEGF signaling pathway, which was regulated by RANTES-CCR5, and selective inhibitor of VEGF receptor axitinib blocked the prophylactic effect of duloxetine. Furthermore, duloxetine also protected gastric mucosa from indomethacin in female rats, and RANTES was increased by duloxetine after 6 h after indomethacin exposure too. Together, our results identified the role of cytokines, particularly RANTES, and the underlying mechanisms in gastroprotective effect of duloxetine against indomethacin, which advanced our understanding in inflammatory modulation by monoamine-based antidepressants.


Chemokine CCL5 , Duloxetine Hydrochloride , Gastric Mucosa , Indomethacin , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Serotonin , Signal Transduction , Stomach Ulcer , Vascular Endothelial Growth Factor A , Animals , Duloxetine Hydrochloride/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Male , Indomethacin/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Chemokine CCL5/metabolism , Signal Transduction/drug effects , Rats , Vascular Endothelial Growth Factor A/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/metabolism , Serotonin/metabolism , Phosphatidylinositol 3-Kinases/metabolism
19.
Nutrients ; 16(10)2024 May 09.
Article En | MEDLINE | ID: mdl-38794668

INTRODUCTION: Justicia pectoralis Jacq. is traditionally applied in folk medicine in Brazil and in several Latin American countries. The leaves are used in tea form, especially in the treatment of respiratory disorders, acting as an expectorant. It also has activity in gastrointestinal disorders, and it is anti-inflammatory, antioxidant, sedative, and estrogenic, among others. AIMS: To investigate the gastroprotective activity of the methanol extract of the leaves of Justicia pectoralis Jacq. (MEJP) in different experimental models of gastric ulcers. MATERIALS AND METHODS: The adult leaves of Justicia pectoralis Jacq. were collected and cultivated in beds, with an approximate spacing of 40 × 40 cm, organic fertilization, irrigation with potable water and without shelter from light. The MEJP was prepared from the dried and pulverized leaves and concentrated under reduced pressure in a rotary evaporator. For the experimental model of gastric ulcer, Swiss male albino mice were used. The inputs used in the experiment were MEJP at three different concentrations (250, 500 and 1000 mg/kg p.o.), cimetidine (50 mg/kg p.o.), indomethacin (50 mg/kg s.c.) and vehicle (10 mL/kg p.o.). RESULTS: MEJP (250, 500 and 1000 mg/kg p.o.) demonstrated gastroprotective activity, with levels of protection of 45.65%, 44.80% and 40.22%, respectively, compared to the control (vehicle). Compared with cimetidine (48.29%), MEJP showed similar gastroprotective activity. CONCLUSIONS: This study demonstrated the gastroprotective activity of MEJP and contributes to validate the traditional use the species for gastric disorders and provides a pharmacological basis for its clinical potential.


Plant Extracts , Plant Leaves , Stomach Ulcer , Animals , Plant Extracts/pharmacology , Mice , Stomach Ulcer/drug therapy , Stomach Ulcer/prevention & control , Plant Leaves/chemistry , Male , Anti-Ulcer Agents/pharmacology , Methanol/chemistry , Justicia/chemistry , Disease Models, Animal , Cimetidine/pharmacology , Acanthaceae/chemistry , Indomethacin , Brazil , Gastric Mucosa/drug effects , Gastric Mucosa/pathology
20.
Mol Biol Rep ; 51(1): 684, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796650

BACKGROUND: Indomethacin is an anti-inflammatory drug that causes ulcers on the gastric mucosa due to its use. Probiotic bacteria are live microorganisms, and it has been stated by various studies that these bacteria have antioxidant and anti-inflammatory effects. In this study, we investigated the possible protective effect of various types of probiotic bacteria (Lactobacillus rhamnosus, Lactobacillus fermentum, and Lactobacillus brevis) against acute gastric mucosal damage caused by indomethacin. METHODS: Control group - Physiological saline was administered daily for 10 days. Indo group-Physiological saline was administered daily for 10 days. Ranitidine + Indo group 5 mg/kg ranitidine dose was administered daily for 5 days. On day 11, a single dose of 100 mg/kg of indomethacin was given to the same group. Probiotic + Indo group 1 ml/kg of oral probiotic bacteria was administered daily for 10 days. On day 11, a single 100 mg/kg dose of indomethacin was given. After the application, the rats were anesthetized with ketamine xylazine, killed under appropriate conditions, the abdominal cavity was opened and the stomach tissues were removed. The obtained gastric tissues were used in the biochemical and histopathological analyses discussed below. All data were statistically evaluated by one-way ANOVA using SPSS 20.00, followed by Duncan Post hoc test. The data were expressed as mean ± SD. P < 0.05 was considered statistically significant. RESULTS: As a result, the administration of indomethacin caused gastric damage, stimulating oxidative stress, inflammation, and apoptosis. We found that the use of probiotic bacteria reduces oxidative stress (TOC), increases the activity of antioxidant enzymes (TAC), suppresses inflammation (IL-6 and Tnf-α), and inhibits apoptosis (Bax and Bcl-2) (P < 0.05). CONCLUSION: Probiotic treatment can mitigate gastric damage and apoptosis caused by indomethacin-induced gastric damage in rats. Probiotic also enhances the restoration of biochemical oxidative enzymes as it has anti-inflammatory, antioxidant, and antiapoptotic properties.


Apoptosis , Gastric Mucosa , Indomethacin , Inflammation , Oxidative Stress , Probiotics , Stomach Ulcer , Indomethacin/adverse effects , Probiotics/pharmacology , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects , Rats , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Inflammation/metabolism , Male , Rats, Wistar , Antioxidants/metabolism , Antioxidants/pharmacology
...