Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 640
Filter
1.
World J Gastroenterol ; 30(17): 2308-2310, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38813049

ABSTRACT

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.


Subject(s)
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
2.
J Transl Med ; 22(1): 429, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711123

ABSTRACT

BACKGROUND: Previous literature has explored the relationship between chronic atrophic gastritis (CAG) and isolated cancers within the upper gastrointestinal cancers; However, an integrative synthesis across the totality of upper gastrointestinal cancers was conspicuously absent. The research objective was to assess the relationship between CAG and the risk of incident upper gastrointestinal cancers, specifically including gastric cancer, oesophageal cancer, and oesophagogastric junction cancer. METHODS: Rigorous systematic searches were conducted across three major databases, namely PubMed, Embase and Web of Science, encompassing the timeline from database inception until August 10, 2023. We extracted the necessary odds ratio (OR) and their corresponding 95% confidence interval (CI) for subsequent meta-analysis. Statistical analyses were conducted using Stata 17.0 software. RESULTS: This meta-analysis included a total of 23 articles encompassing 5858 patients diagnosed with upper gastrointestinal cancers. CAG resulted in a statistically significant 4.12-fold elevated risk of incident gastric cancer (OR = 4.12, 95% CI 3.20-5.30). Likewise, CAG was linked to a 2.08-fold increased risk of incident oesophageal cancer (OR = 2.08, 95%CI 1.60-2.72). Intriguingly, a specific correlation was found between CAG and the risk of incident oesophageal squamous cell carcinoma (OR = 2.29, 95%CI 1.77-2.95), while no significant association was detected for oesophageal adenocarcinoma (OR = 0.62, 95%CI 0.17-2.26). Moreover, CAG was correlated with a 2.77-fold heightened risk of oesophagogastric junction cancer (OR = 2.77, 95%CI 2.21-3.46). Notably, for the same type of upper gastrointestinal cancer, it was observed that diagnosing CAG through histological methods was linked to a 33-77% higher risk of developing cancer compared to diagnosing CAG through serological methods. CONCLUSION: This meta-analysis indicated a two- to fourfold increased risk of gastric cancer, oesophageal cancer, and oesophagogastric junction cancer in patients with CAG. Importantly, for the same upper gastrointestinal cancer, the risk of incident cancer was higher when CAG was diagnosed histologically compared to serological diagnosis. Further rigorous study designs are required to explore the impact of CAG diagnosed through both diagnostic methods on the risk of upper gastrointestinal cancers.


Subject(s)
Gastritis, Atrophic , Gastrointestinal Neoplasms , Humans , Gastritis, Atrophic/complications , Gastritis, Atrophic/epidemiology , Risk Factors , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/pathology , Chronic Disease , Incidence , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/pathology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology , Male , Odds Ratio , Female , Publication Bias
3.
Nat Rev Dis Primers ; 10(1): 25, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605021

ABSTRACT

Gastric neuroendocrine neoplasms (gNENs) display peculiar site-specific features among all NENs. Their incidence and prevalence have been rising in the past few decades. gNENs comprise gastric neuroendocrine carcinomas (gNECs) and gastric neuroendocrine tumours (gNETs), the latter further classified into three types. Type I anatype II gNETs are gastrin-dependent and develop in chronic atrophic gastritis and as part of Zollinger-Ellison syndrome within a multiple endocrine neoplasia type 1 syndrome (MEN1), respectively. Type III or sporadic gNETs develop in the absence of hypergastrinaemia and in the context of a near-normal or inflamed gastric mucosa. gNECs can also develop in the context of variable atrophic, relatively normal or inflamed gastric mucosa. Each gNEN type has different clinical characteristics and requires a different multidisciplinary approach in expert dedicated centres. Type I gNETs are managed mainly by endoscopy or surgery, whereas the treatment of type II gNETs largely depends on the management of the concomitant MEN1. Type III gNETs may require both locoregional approaches and systemic treatments; NECs are often metastatic and therefore require systemic treatment. Specific data regarding the systemic treatment of gNENs are lacking and are derived from the treatment of intestinal NETs and NECs. An enhanced understanding of molecular and clinical pathophysiology is needed to improve the management and outcomes of patients' gNETs.


Subject(s)
Gastritis, Atrophic , Neuroendocrine Tumors , Pancreatic Neoplasms , Stomach Neoplasms , Zollinger-Ellison Syndrome , Humans , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/therapy , Zollinger-Ellison Syndrome/complications , Gastritis, Atrophic/complications , Gastritis, Atrophic/epidemiology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology , Stomach Neoplasms/therapy
4.
BMC Gastroenterol ; 24(1): 139, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649806

ABSTRACT

BACKGROUND: Gastric hamartomatous inverted polyps (GHIPs) are not well characterized and remain diagnostically challenging due to rarity. Therefore, this study aims to investigate the clinicopathologic and endoscopic characteristics of patients with GHIP. METHODS: We retrospectively reviewed clinicopathologic and endoscopic features of ten patients with GHIP who were admitted to Beijing Friendship Hospital from March 2013 to July 2022. All patients were treated successfully by endoscopic resection. RESULTS: GHIPs were usually asymptomatic and found incidentally during gastroscopic examination. They may be sessile or pedunculated, with diffuse or local surface redness or erosion. On endoscopic ultrasonography, the sessile submucosal tumor-type GHIP demonstrated a heterogeneous lesion with cystic areas in the third layer of the gastric wall. Histologically, GHIPs were characterized by a submucosal inverted proliferation of cystically dilated hyperplastic gastric glands accompanied by a branching proliferation of smooth muscle bundles. Inflammatory cells infiltration was observed in the stroma, whereas only one patient was complicated with glandular low-grade dysplasia. Assessment of the surrounding mucosa demonstrated that six patients (60%) had atrophic gastritis or Helicobacter pylori-associated gastritis, and four patients (40%) had non-specific gastritis. Endoscopic resection was safe and effective. CONCLUSIONS: GHIPs often arise from the background of abnormal mucosa, such as atrophic or H.pylori-associated gastritis. We make the hypothesis that acquired inflammation might lead to the development of GHIPs. We recommend to make a full assessment of the background mucosa and H. pylori infection status for evaluation of underlying gastric mucosal abnormalities, which may be the preneoplastic condition of the stomach.


Subject(s)
Adenomatous Polyps , Endosonography , Gastric Mucosa , Gastroscopy , Hamartoma , Polyps , Stomach Neoplasms , Humans , Male , Female , Middle Aged , Retrospective Studies , Hamartoma/pathology , Hamartoma/diagnostic imaging , Hamartoma/surgery , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/diagnostic imaging , Gastric Mucosa/pathology , Gastric Mucosa/diagnostic imaging , Gastric Mucosa/surgery , Adult , Aged , Polyps/pathology , Polyps/surgery , Polyps/diagnostic imaging , Stomach Diseases/pathology , Stomach Diseases/surgery , Stomach Diseases/diagnostic imaging , Helicobacter Infections/complications , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Gastritis/pathology , Gastritis/complications , Gastritis/diagnostic imaging , Gastritis, Atrophic/pathology , Gastritis, Atrophic/complications , Endoscopic Mucosal Resection
5.
P R Health Sci J ; 43(1): 9-17, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38512756

ABSTRACT

Of the chronic bacterial infections that affect humans, Helicobacter pylori (H. pylori) infection is one of the most common. It inhabits the stomachs of half of the adult human population. In Puerto Rico, a US territory, it has an overall prevalence of 33%, similar to the prevalence reported in the population of the US as a whole. Helicobacter pylori infection is responsible for mucosal inflammation that may lead to chronic gastritis, most peptic ulcers, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. The International Agency for Research on Cancer identified H. pylori as a definite carcinogen in 1994, the only bacterium to be given such a classification. Its oncogenic effect has been postulated to be caused by different mechanisms, including bacterial characteristics and host factors. Epidemiologic studies have shown that gastric cancer risk differs among regions. One of the top 10 causes of cancer death in Puerto Rico is gastric cancer. Although the eradication of H. pylori has well-known benefits, there are some concerns when considering mass screening and treatment of infected patients. These include the fact that such eradication could provoke an increase in antibiotic resistance rates, the disturbance of the gut microbiota, an increase in body weight, and the aggravation of existing gastroesophageal reflux symptoms. Gastric cancer is a major health concern, and we should understand the role of H. pylori eradication in its prevention. This article is geared to summarize current knowledge and controversies.


Subject(s)
Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Adult , Humans , Stomach Neoplasms/epidemiology , Stomach Neoplasms/etiology , Helicobacter Infections/complications , Helicobacter Infections/epidemiology , Gastritis, Atrophic/complications , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Puerto Rico
6.
Nutrients ; 16(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474790

ABSTRACT

Autoimmune gastritis (AIG) is characterized by the destruction of gastric parietal cells, resulting in hypochlorhydria and eventual achlorhydria, as oxyntic glands in the corpus are destroyed and become atrophic. The permanent loss of gastric acid has many impacts-both theoretical and documented. The most concerning of these are hypergastrinemia and increased N-nitroso compounds, both of which increase the risk of gastric cancers. While known deficiencies of B12 and iron are often replaced in AIG, acid is not. Moreover, patients with AIG are often prescribed acid suppression for a stomach that is decidedly no longer acidic, worsening the sequelae of gastric atrophy. Betaine hydrochloride (BHCL) is a short-acting acidifying agent, available over the counter in capsule form. Mealtime acid supplementation has an historic basis and could ameliorate many AIG-related gastrointestinal symptoms. Theoretically, acidification could also reduce the potential for hypergastrinemia and the production of N-nitroso compounds, consequently reducing the risk of gastric cancers. Supplemental vitamin C may also help in preventing gastric N-nitroso formation, regardless of the gastric pH. This narrative review describes the functions of gastric acid in gastrointestinal and immune health, documents the effects of hypochlorhydria in AIG, and proposes potential options for safely re-establishing the acid milieu of the stomach for patients with AIG.


Subject(s)
Achlorhydria , Autoimmune Diseases , Gastritis, Atrophic , Gastritis , Stomach Neoplasms , Humans , Stomach Neoplasms/complications , Gastritis, Atrophic/complications , Gastritis, Atrophic/diagnosis , Gastric Mucosa , Nitroso Compounds
7.
Dig Dis ; 42(2): 145-153, 2024.
Article in English | MEDLINE | ID: mdl-38198775

ABSTRACT

INTRODUCTION: Iron and vitamin B12 deficiencies are common in patients with atrophic gastritis, but there are limited data on the prevalence of these deficiencies in different types of atrophic gastritis. METHODS: This multicenter, prospective study assessed micronutrient concentrations in histologically confirmed autoimmune gastritis (AIG, n = 45), Helicobacter pylori-related non-autoimmune gastritis (NAIG, n = 109), and control patients (n = 201). A multivariate analysis was performed to determine factors influencing those deficiencies. RESULTS: The median vitamin B12 concentration was significantly lower in AIG (367.5 pg/mL, Q1, Q3: 235.5, 524.5) than in NAIG (445.0 pg/mL, Q1, Q3: 355.0, 565.0, p = 0.001) and control patients (391.0 pg/mL, Q1, Q3: 323.5, 488.7, p = 0.001). Vitamin B12 deficiency was found in 13.3%, 1.5%, and 2.8% of AIG, NAIG, and control patients, respectively. Similarly, the median ferritin concentration was significantly lower in AIG (39.5 ng/mL, Q1, Q3: 15.4, 98.3 ng/mL) than in NAIG (80.5 ng/mL, Q1, Q3: 43.6, 133.9, p = 0.04) and control patients (66.5 ng/mL, Q1, Q3: 33.4, 119.8, p = 0.007). Iron deficiency and iron deficiency adjusted to CRP were present in 28.9% and 33.3% of AIG, 12.8% and 16.5% of NAIG, and 12.9% and 18.4% of controls, respectively. Multivariate analysis demonstrated that AIG patients had a higher risk of developing vitamin B12 deficiency (OR: 11.52 [2.85-57.64, p = 0.001]) and iron deficiency (OR: 2.92 [1.32-6.30, p = 0.007]) compared to control patients. Factors like age, sex, and H. pylori status did not affect the occurrence of vitamin B12 or iron deficiency. CONCLUSION: Iron and vitamin B12 deficiencies are more commonly observed in patients with AIG than in those with NAIG or control patients. Therefore, it is essential to screen for both iron and vitamin B12 deficiencies in AIG patients and include the treatment of micronutrient deficiencies in the management of atrophic gastritis patients.


Subject(s)
Autoimmune Diseases , Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Iron Deficiencies , Vitamin B 12 Deficiency , Humans , Gastritis, Atrophic/complications , Gastritis, Atrophic/epidemiology , Prospective Studies , Iron , Gastritis/complications , Gastritis/epidemiology , Vitamin B 12 Deficiency/complications , Vitamin B 12 Deficiency/epidemiology , Helicobacter Infections/complications , Helicobacter Infections/epidemiology , Vitamin B 12 , Micronutrients , Autoimmune Diseases/complications
8.
Cleve Clin J Med ; 91(1): 33-39, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167394

ABSTRACT

Gastric intestinal metaplasia (GIM), a common histologic finding, is associated with increased risk of gastric cancer, and GIM associated with Helicobacter pylori infection is classified as an environmental metaplastic atrophic gastritis. Patients may be asymptomatic or present with various dyspeptic symptoms. Autoimmune metaplastic atrophic gastritis is a less common but important cause of chronic gastritis. The Correa cascade describes the evolution of precancerous mucosal changes that lead to development of GIM, with differentiation of 2 histologic types of GIM (complete and incomplete) and the consequences of each type. The risk of progression to malignancy is higher with incomplete GIM. It is also higher for those who immigrate from regions with a high incidence of H pylori infection to areas where the incidence is low. Guidelines regarding endoscopic management of GIM vary by geographic region.


Subject(s)
Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Precancerous Conditions , Stomach Neoplasms , Humans , Stomach Neoplasms/etiology , Stomach Neoplasms/prevention & control , Gastritis, Atrophic/complications , Helicobacter Infections/complications , Watchful Waiting , Gastritis/complications , Gastritis/diagnosis , Gastritis/pathology , Precancerous Conditions/complications , Precancerous Conditions/diagnosis , Precancerous Conditions/epidemiology , Metaplasia/complications
10.
Clin J Gastroenterol ; 17(2): 216-221, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38072907

ABSTRACT

A 61-year-old female patient underwent upper gastrointestinal endoscopy, which confirmed the presence of Helicobacter pylori (H. pylori)-positive nodular gastritis (NG). Routine upper gastrointestinal endoscopy after H. pylori eradication revealed atrophic changes of the corpus, having gradually progressed over the 10 years after successful eradication. Serological and biopsy specimen examination showed hypergastrinemia (1200 pg/mL), positive anti-parietal cell antibody (with a titer of more 160), and endocrine cell micronests after 11 years of H. pylori eradication. The patient was diagnosed with autoimmune gastritis (AIG) based on endoscopic, serological, and histological findings. This is the first report of AIG diagnosed in a patient with NG over a long period of time after H. pylori eradication.


Subject(s)
Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Female , Humans , Middle Aged , Gastritis, Atrophic/complications , Gastritis, Atrophic/drug therapy , Gastritis, Atrophic/pathology , Gastritis/drug therapy , Helicobacter Infections/complications , Helicobacter Infections/drug therapy , Atrophy
11.
Eur J Pediatr ; 183(2): 515-522, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37947925

ABSTRACT

We would like to provide an updated comprehensive perspective and identify the components linked to chronic spontaneous urticaria (CSU) without specific triggers in autoimmune atrophic gastritis (AAG). AAG is an organ-specific autoimmune disease that affects the corpus-fundus gastric mucosa. Although we lack a unified explanation of the underlying pathways, when considering all paediatric patients reported in the literature, alterations result in gastric neuroendocrine enterochromaffin-like (ECL) cell proliferation and paracrine release of histamine. Several mechanisms have been proposed for the pathogenesis of CSU, with much evidence pointing towards AAG and ECL cell responses, which may be implicated as potential factors contributing to CSU. The excessive production/release of histamine into the bloodstream could cause or trigger exacerbations of CSU in AAG, independent of Helicobacter pylori; thus, the release of histamine from ECL cells may be the primary modulator. CONCLUSION: Considering the understanding of these interactions, recognising the respective roles of AAG in the pathogenesis of CSU may strongly impact the diagnostic workup and management of unexplained/refractory CSU and may inform future research and interventions in the paediatric population. WHAT IS KNOWN: • Autoimmune atrophic gastritis is a chronic immune-mediated inflammatory disease characterised by the destruction of the oxyntic mucosa in the gastric body and fundus, mucosal atrophy, and metaplastic changes. • Autoimmune atrophic gastritis in paediatric patients is important because of the poor outcome and risk of malignancy and possibly underestimated entities primarily reported in single-case reports. WHAT IS NEW: • Upper gastrointestinal inflammatory disorders, independent of H. pylori, have been implicated as potential inducing factors in the development of chronic spontaneous urticaria. • If a paediatric patient presents with symptoms such as anaemia, reduced vitamin B12 levels, recurrent urticaria with no other detectable aetiology, positive anti-parietal cell antibodies, and elevated gastrin levels, autoimmune atrophic gastritis should be considered a possible cause of chronic urticaria.


Subject(s)
Autoimmune Diseases , Chronic Urticaria , Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Humans , Child , Gastritis, Atrophic/complications , Gastritis, Atrophic/pathology , Histamine , Gastritis/complications , Gastritis/diagnosis , Gastric Mucosa/pathology , Chronic Urticaria/etiology , Chronic Urticaria/pathology , Autoimmune Diseases/complications , Autoimmune Diseases/diagnosis , Chronic Disease , Helicobacter Infections/complications
13.
Gut Liver ; 18(1): 60-69, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37720994

ABSTRACT

Background/Aims: : A latex agglutination turbidity (LA) assay to test for serum antibodies has been approved in Japan and Korea for mass screening of Helicobacter pylori infection. In this study, we evaluated the LA assay for diagnosing H. pylori infection and predicting gastric mucosal changes in a Mongolian population. Methods: : In total, 484 individuals were classified into H. pylori-positive (n=356) and H. pylori-negative (n=128) groups, as determined by histology and H. pylori culture. Results: : The best cutoff, sensitivity, and specificity values for the LA assay were 18.35 U/mL, 74.2%, and 65.6%, respectively. The LA values in the atrophic gastritis group were statistically higher than those in the other groups (healthy, chronic gastritis, intestinal metaplasia, and gastric cancer, p<0.0001). The cutoff value to distinguish the atrophic gastritis group from the other four groups was 32.0 U/mL, and its area under the curve was 0.673, which was the highest among the E-plate, pepsinogen (PG) I, PG II, and PG I/II ratio tests in our data. The odds ratios for atrophic gastritis determined by the LA assay and PG I test in multiple logistic regression were 2.5 and 1.9, respectively, which were significantly higher than for the other tests. Conclusions: : The LA assay can determine the risk of atrophic gastritis, which in turn is a considerable risk factor for gastric cancer. We propose using this assay in combination with the PG I/II ratio to avoid missing gastric cancer patients who have a low LA value (less than 32.0 U/mL).


Subject(s)
Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Gastritis, Atrophic/complications , Stomach Neoplasms/pathology , Helicobacter Infections/diagnosis , Helicobacter Infections/complications , Latex Fixation Tests , Pepsinogen A , Pepsinogen C
14.
Adv Med Sci ; 68(2): 491-498, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37945439

ABSTRACT

PURPOSE: Atrophic gastritis, one of the processes leading to gastric cancer (GC), is closely related to Helicobacter pylori (HP) infection. This study aimed to understand how HP causes chronic inflammation that leads to ulcers and stomach problems. METHODS: Twenty-eight CAG patients were included in the study (9 HP-infected and 19 HP-uninfected). Endoscopy, histopathology, and high-throughput mRNA sequencing were performed. Differentially expressed genes (DEGs) were validated via qRT-PCR. RESULTS: Principal component analysis (PCA) results showed that more than 88.9 â€‹% of the samples were classified into the HP (+) group. A total of 157 DEGs were identified, of which 38 were up-regulated and 119 were down-regulated. The DEGs were mainly enriched in the biological process (BP) terms associated with immune system process, adaptive immune response, G protein-coupled receptor signaling pathway, as well as point to numerous key pathways, including fat digestion and absorption, retinol metabolism, steroid hormone biosynthesis, ascorbate and aldarate metabolism, and chemical carcinogenesis. APOA1, APOA4, FOXP3, NR1H4, ABCG5, ACTA1, CCL19, CCR7, CYP3A4, and PDCD had the highest degrees in protein-protein interaction network as the hub genes; they were also included into the transcription factor (TF)-target network except for PDCD. APOA1 and CYP3A4 were extremely significantly up-regulated in HP (+) CAG patients compared with the HP (-) CAG patients, while FOXP3, CCR7 and CCL19 were significantly down-regulated. CONCLUSION: The expression of APOA1, CYP3A4, FOXP3, CCR7, and CCL19 are the potential indicators for CAG to GC development, being the biomarkers to predict progression of CAG and poor prognosis of GC.


Subject(s)
Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Gastritis, Atrophic/genetics , Gastritis, Atrophic/complications , Gastritis, Atrophic/metabolism , Cytochrome P-450 CYP3A , Receptors, CCR7 , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Forkhead Transcription Factors , RNA
15.
BMJ Case Rep ; 16(11)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38035681

ABSTRACT

Nitrous oxide is among the most common drugs used by adolescents and young adults, and its neuropsychiatric sequelae are severe but reversible with timely treatment. The causal mechanism relates to impaired metabolism of vitamin B12, which is necessary for the development and maintenance of the myelin sheath. Individuals most susceptible to neuropsychiatric manifestations are those with a secondary cause of vitamin B12 deficiency, including nutritional deficiency and impaired absorption, or an alternative cause of impaired metaboclism. We describe the case of a man in his thirties who developed subacute combined degeneration of the spinal cord and polyneuropathy in the setting of recreational nitrous oxide use and autoimmune atrophic gastritis. Our case highlights clinical pearls for diagnosis and treatment, differential diagnosis, common concomitant aetiologies and the importance of screening for substance use disorder and psychiatric comorbidities.


Subject(s)
Gastritis, Atrophic , Gastritis , Subacute Combined Degeneration , Vitamin B 12 Deficiency , Humans , Male , Atrophy/pathology , Gastritis/chemically induced , Gastritis/diagnosis , Gastritis/drug therapy , Gastritis, Atrophic/complications , Nitrous Oxide/adverse effects , Spinal Cord/diagnostic imaging , Spinal Cord/pathology , Subacute Combined Degeneration/drug therapy , Subacute Combined Degeneration/etiology , Vitamin B 12/metabolism , Vitamin B 12 Deficiency/chemically induced , Vitamin B 12 Deficiency/complications , Vitamin B 12 Deficiency/diagnosis , Adult
17.
PeerJ ; 11: e16243, 2023.
Article in English | MEDLINE | ID: mdl-37868053

ABSTRACT

Background: Gastric cancer (GC) is the fourth leading cause of cancer-related death worldwide. However, the precise mechanisms and specific biomarkers of GC have not been fully elucidated. We therefore sought to identify and validate the genes associated with GC. Methods: RNA sequencing was performed on gastric tissue specimens from 10 cases each of non-atrophic gastritis (NAG), intestinal metaplasia (IM), and GC. Validation of gene expression was conducted through immunohistochemistry (IHC) staining. The Kaplan-Meier Plotter database was utilized to screen genes associated with prognosis, while protein-protein interaction analysis was conducted to identify hub genes. Results: In GC-IM, the differentially expressed genes (DEGs) were predominantly enriched in pathways related to ECM-receptor interaction, focal adhesion, PI3K-Akt pathway, and pathways in cancer. Conversely, in IM-NAG, the DEGs were primarily enriched in pathways associated with fat digestion and absorption, pancreatic secretion, and retinol metabolism. IHC staining revealed elevated expression levels of KLK7 and KLK10 in GC. Specifically, KLK7 expression was found to be correlated with differentiation (P = 0.025) and depth of invasion (P = 0.007) in GC, while both KLK7 and KLK10 were associated with the overall survival (P < 0.05). Furthermore, a total of ten hub genes from DEGs in GC-NAG (COL6A2, COL1A1, COL4A1, COL1A2, SPARC, COL4A2, FN1, PCOLCE, SERPINH1, LAMB1) and five hub genes in IM-NAG (SI, DPP4, CLCA1, MEP1A, OLFM4) were demonstrated to have a significant correlation with the prognosis of GC. Conclusions: The present study successfully identified and validated crucial genes associated with GC, providing valuable insights into the underlying mechanisms of this disease. The findings of this study have the potential to inform clinical practice.


Subject(s)
Gastritis, Atrophic , Stomach Neoplasms , Humans , Stomach Neoplasms/diagnosis , Phosphatidylinositol 3-Kinases , Prognosis , Collagen Type I , Gastritis, Atrophic/complications
18.
Korean J Gastroenterol ; 82(4): 171-179, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37876256

ABSTRACT

Chronic inflammation due to a Helicobacter pylori (H. pylori) infection is a representative cause of gastric cancer that can promote gastric carcinogenesis by abnormally activating immune cells and increasing the inflammatory cytokines levels. H. pylori infections directly cause DNA double-strand breaks in gastric epithelial cells and genetic damage by increasing the enzymatic activity of cytidine deaminase. Eventually, gastric cancer is induced through dysplasia. Hypermethylation of tumor suppressor genes is an important cause of gastric cancer because of a H. pylori infection. In addition, the changes in gastric microbiota and the mucosal inflammatory changes associated with a co-infection with the Epstein-Barr virus are associated with gastric cancer development. DNA damage induced by H. pylori and the subsequent responses of gastric stem cells have implications for gastric carcinogenesis. Although the pathogenesis of H. pylori has been established, many uncertainties remain, requiring more study.


Subject(s)
Epstein-Barr Virus Infections , Gastritis, Atrophic , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Gastritis, Atrophic/complications , Gastritis, Atrophic/pathology , Stomach Neoplasms/pathology , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Carcinogenesis/pathology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Gastric Mucosa/pathology
19.
J Radiat Res ; 64(6): 870-879, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37788485

ABSTRACT

Radiotherapy (RT) has been the standard of care for treating a multitude of cancer types. Radiation-induced gastric injury (RIGI) is a common complication of RT for thoracic and abdominal tumors. It manifests acutely as radiation gastritis or gastric ulcers, and chronically as chronic atrophic gastritis or intestinal metaplasia. In recent years, studies have shown that intracellular signals such as oxidative stress response, p38/MAPK pathway and transforming growth factor-ß signaling pathway are involved in the progression of RIGI. This review also summarized the risk factors, diagnosis and treatment of this disease. However, the root of therapeutic challenges lies in the incomplete understanding of the mechanisms. Here, we also highlight the potential mechanistic, diagnostic and therapeutic directions of RIGI.


Subject(s)
Gastritis, Atrophic , Stomach Neoplasms , Stomach Ulcer , Humans , Gastritis, Atrophic/complications , Gastritis, Atrophic/pathology , Risk Factors , Oxidative Stress , Stomach Neoplasms/radiotherapy , Stomach Neoplasms/pathology
20.
Nutrients ; 15(19)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37836482

ABSTRACT

Corpus Atrophic Gastritis (CAG) is characterised by iron malabsorption leading to iron deficiency anaemia (IDA), which rarely responds to oral therapy. Ferric carboxymaltose (FCM), shown to be a safe and effective intravenous iron therapy in other diseases, has not been investigated yet in CAG. Thus, we aimed to assess the safety and efficacy of FCM in CAG-related IDA. A retrospective study on 91 patients identified CAG as the only cause of IDA treated with FCM. Twenty-three were excluded for incomplete follow-up. Sixty-eight were evaluated for safety and efficacy, while three were evaluated for safety only due to infusion interruption for side effects. Haemoglobin and iron storage were evaluated pre-infusion (T0), at 4 weeks (T4) and 12 weeks (T12) after infusion. An eventual IDA relapse was analysed. Two cases reported mild side effects. Haemoglobin significantly increased at T4, and T12, reaching +3.1 g/dL. Ferritin increased at T4, decreasing at T12, while transferrin saturation increased progressively until reaching a plateau. IDA relapsed in 55.4% of patients at a mean of 24.6 months. The only factor associated with relapse was female gender [OR (95% CI): 6.6 (1.5-28.6)]. FCM proved to be safe and effective in treating CAG-related IDA, ensuring quick and long-lasting recovery.


Subject(s)
Anemia, Iron-Deficiency , Gastritis, Atrophic , Humans , Female , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/etiology , Retrospective Studies , Gastritis, Atrophic/chemically induced , Gastritis, Atrophic/complications , Gastritis, Atrophic/drug therapy , Ferric Compounds/adverse effects , Iron/therapeutic use , Hemoglobins/analysis , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...