Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
Surg Endosc ; 37(12): 9366-9372, 2023 12.
Article in English | MEDLINE | ID: mdl-37644156

ABSTRACT

BACKGROUND: Vonoprazan is a new acid-suppressing drug that received FDA approval in 2022. It reversibly inhibits gastric acid secretion by competing with the potassium ions on the luminal surface of the parietal cells (potassium-competitive acid blockers or P-CABs). Vonoprazan has been on the market for a short time and there are many clinical trials to support its clinical application. However, medical experience and comprehensive clinical data is still limited, especially on how and if, gastric histology is altered due to therapy. METHODS: A 12-week experiment trial with 30 Wistar rats was to assess the presence of gastrointestinal morphologic abnormalities upon administration of omeprazole and vonoprazan. At six weeks of age, rats were randomly assigned to one of 5 groups: (1) saline as negative control group, (2) oral omeprazole (40 mg/kg), as positive control group, (3) oral omeprazole (40 mg/kg) for 4 weeks, proceeded by 8 weeks off omeprazole, (4) oral vonoprazan (4 mg/kg), as positive control group, and (5) oral vonoprazan (4 mg/kg) for 4 weeks, proceeded by 8 weeks off vonoprazan. RESULTS: We identified non-inflammatory alterations characterized by parietal (oxyntic) cell loss and chief (zymogen) cell hyperplasia and replacement by pancreatic acinar cell metaplasia (PACM). No significant abnormalities were identified in any other tissues in the hepatobiliary and gastrointestinal tracts. CONCLUSION: PACM has been reported in gastric mucosa, at the esophagogastric junction, at the distal esophagus, and in Barrett esophagus. However, the pathogenesis of this entity is still unclear. Whereas some authors have suggested that PACM is an acquired process others have raised the possibility of PACM being congenital in nature. Our results suggest that the duration of vonoprazan administration at a dose of 4 mg/kg plays an important role in the development of PACM.


Subject(s)
Proton Pump Inhibitors , Pyrroles , Animals , Rats , Acinar Cells , Metaplasia/chemically induced , Omeprazole/adverse effects , Potassium , Proton Pump Inhibitors/adverse effects , Pyrroles/adverse effects , Rats, Wistar
2.
World J Urol ; 40(3): 849-855, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35034167

ABSTRACT

PURPOSE: To determine the prevalence of prostatic metaplasia in an expanded cohort of transmasculine individuals undergoing gender-affirming resection of vaginal tissue. METHODS: Institutional Review Board approval was obtained. Clinical records were reviewed for all transmasculine individuals undergoing vaginal tissue resection at our institution between January 2018 and July 2021. Corresponding pathology specimens were examined grossly and microscopically, including immunohistochemical stains for NKX3.1, prostate-specific antigen (PSA), and androgen receptor (AR). Vaginal specimens from three patients without androgen supplementation were used as controls. RESULTS: Twenty-one patients met inclusion criteria. The median age at surgery was 26.4 years (range 20.6-34.5 years). All patients had been assigned female gender at birth and lacked endocrine or genetic abnormalities. All were on testosterone therapy; median duration of therapy at surgery was 4.4 years (range 1.4-12.1 years). In the transmasculine group, no gross lesions were identified. Microscopically, all specimens demonstrated patchy intraepithelial glandular proliferation along the basement membrane and/or nodular proliferation of prostate-type tissue within the subepithelial stroma. On immunohistochemical staining, performed for a subset of cases, the glandular proliferation was positive for NKX3.1 (16/16 cases; 100%), PSA (12/14 cases; 85.7%), and AR (8/8 cases; 100%). Controls showed no evidence of prostatic metaplasia. CONCLUSION: One hundred percent of vaginal specimens obtained from transmasculine individuals on testosterone therapy (21/21 cases) demonstrated prostatic metaplasia. Further investigation is warranted to characterize the natural history and clinical significance of these changes. Patients seeking hormone therapy and/or gender-affirming surgery should be counseled on the findings and their yet-undetermined significance.


Subject(s)
Prostate , Transgender Persons , Adult , Androgens/therapeutic use , Female , Humans , Infant, Newborn , Male , Metaplasia/chemically induced , Metaplasia/drug therapy , Vagina , Young Adult
3.
Gastroenterology ; 159(6): 2077-2091.e8, 2020 12.
Article in English | MEDLINE | ID: mdl-32891625

ABSTRACT

BACKGROUND & AIMS: Severe injury to the lining of the stomach leads to changes in the epithelium (reprogramming) that protect and promote repair of the tissue, including development of spasmolytic polypeptide-expressing metaplasia (SPEM) and tuft and foveolar cell hyperplasia. Acute gastric damage elicits a type-2 inflammatory response that includes production of type-2 cytokines and infiltration by eosinophils and alternatively activated macrophages. Stomachs of mice that lack interleukin 33 (IL33) or interleukin 13 (IL13) did not undergo epithelial reprogramming after drug-induced injury. We investigated the role of group 2 innate lymphoid cells (ILC2s) in gastric epithelial repair. METHODS: Acute gastric injury was induced in C57BL/6J mice (wild-type and RAG1 knockout) by administration of L635. We isolated ILC2s by flow cytometry from stomachs of mice that were and were not given L635 and performed single-cell RNA sequencing. ILC2s were depleted from wild-type and RAG1-knockout mice by administration of anti-CD90.2. We assessed gastric cell lineages, markers of metaplasia, inflammation, and proliferation. Gastric tissue microarrays from patients with gastric adenocarcinoma were analyzed by immunostaining. RESULTS: There was a significant increase in the number of GATA3-positive ILC2s in stomach tissues from wild-type mice after L635-induced damage, but not in stomach tissues from IL33-knockout mice. We characterized a marker signature of gastric mucosal ILC2s and identified a transcription profile of metaplasia-associated ILC2s, which included changes in expression of Il5, Il13, Csf2, Pd1, and Ramp3; these changes were validated by quantitative polymerase chain reaction and immunocytochemistry. Depletion of ILC2s from mice blocked development of metaplasia after L635-induced injury in wild-type and RAG1-knockout mice and prevented foveolar and tuft cell hyperplasia and infiltration or activation of macrophages after injury. Numbers of ILC2s were increased in stomach tissues from patients with SPEM compared with patients with normal corpus mucosa. CONCLUSIONS: In analyses of stomach tissues from mice with gastric tissue damage and patients with SPEM, we found evidence of type 2 inflammation and increased numbers of ILC2s. Our results suggest that ILC2s coordinate the metaplastic response to severe gastric injury.


Subject(s)
Gastric Mucosa/pathology , Immunity, Innate , Lymphocyte Subsets/immunology , Animals , Disease Models, Animal , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Humans , Interleukin-33/genetics , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/immunology , Mice , Mice, Knockout
4.
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
5.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34066250

ABSTRACT

TMEM16A, a Ca2+-activated chloride channel (CaCC), and its regulator, CLCA1, are associated with inflammatory airway disease and goblet cell metaplasia. CLCA1 is a secreted protein with protease activity that was demonstrated to enhance membrane expression of TMEM16A. Expression of CLCA1 is particularly enhanced in goblet cell metaplasia and is associated with various lung diseases. However, mice lacking expression of CLCA1 showed the same degree of mucous cell metaplasia and airway hyperreactivity as asthmatic wild-type mice. To gain more insight into the role of CLCA1, we applied secreted N-CLCA1, produced in vitro, to mice in vivo using intratracheal instillation. We observed no obvious upregulation of TMEM16A membrane expression by CLCA1 and no differences in ATP-induced short circuit currents (Iscs). However, intraluminal mucus accumulation was observed by treatment with N-CLCA1 that was not seen in control animals. The effects of N-CLCA1 were augmented in ovalbumin-sensitized mice. Mucus production induced by N-CLCA1 in polarized BCi-NS1 human airway epithelial cells was dependent on TMEM16A expression. IL-13 upregulated expression of CLCA1 and enhanced mucus production, however, without enhancing purinergic activation of Isc. In contrast to polarized airway epithelial cells and mouse airways, which express very low levels of TMEM16A, nonpolarized airway cells express large amounts of TMEM16A protein and show strong CaCC. The present data show an only limited contribution of TMEM16A to airway ion secretion but suggest a significant role of both CLCA1 and TMEM16A for airway mucus secretion.


Subject(s)
Anoctamin-1/metabolism , Asthma/pathology , Chloride Channels/metabolism , Goblet Cells/pathology , Metaplasia/pathology , Mucus/metabolism , Respiratory Mucosa/pathology , Animals , Anoctamin-1/genetics , Asthma/chemically induced , Asthma/metabolism , Chloride Channels/genetics , Goblet Cells/metabolism , Metaplasia/chemically induced , Metaplasia/metabolism , Mice , Ovalbumin/toxicity , Respiratory Mucosa/metabolism
6.
Gut ; 69(6): 1027-1038, 2020 06.
Article in English | MEDLINE | ID: mdl-31481545

ABSTRACT

OBJECTIVE: Spasmolytic polypeptide-expressing metaplasia (SPEM) is a regenerative lesion in the gastric mucosa and is a potential precursor to intestinal metaplasia/gastric adenocarcinoma in a chronic inflammatory setting. The goal of these studies was to define the transcriptional changes associated with SPEM at the individual cell level in response to acute drug injury and chronic inflammatory damage in the gastric mucosa. DESIGN: Epithelial cells were isolated from the gastric corpus of healthy stomachs and stomachs with drug-induced and inflammation-induced SPEM lesions. Single cell RNA sequencing (scRNA-seq) was performed on tissue samples from each of these settings. The transcriptomes of individual epithelial cells from healthy, acutely damaged and chronically inflamed stomachs were analysed and compared. RESULTS: scRNA-seq revealed a population Mucin 6 (Muc6)+gastric intrinsic factor (Gif)+ cells in healthy tissue, but these cells did not express transcripts associated with SPEM. Furthermore, analyses of SPEM cells from drug injured and chronically inflamed corpus yielded two major findings: (1) SPEM and neck cell hyperplasia/hypertrophy are nearly identical in the expression of SPEM-associated transcripts and (2) SPEM programmes induced by drug-mediated parietal cell ablation and chronic inflammation are nearly identical, although the induction of transcripts involved in immunomodulation was unique to SPEM cells in the chronic inflammatory setting. CONCLUSIONS: These data necessitate an expansion of the definition of SPEM to include Tff2+Muc6+ cells that do not express mature chief cell transcripts such as Gif. Our data demonstrate that SPEM arises by a highly conserved cellular programme independent of aetiology and develops immunoregulatory capabilities in a setting of chronic inflammation.


Subject(s)
Gastric Mucosa/metabolism , Gastritis/chemically induced , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Female , Fluorescent Antibody Technique , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastritis/metabolism , Gastritis/pathology , Gene Expression Profiling , In Situ Hybridization , Male , Metaplasia/chemically induced , Metaplasia/metabolism , Mice , Mice, Inbred BALB C , Mucin-6/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Tamoxifen/pharmacology , Trefoil Factor-2/metabolism
7.
Gastroenterology ; 152(4): 762-766.e7, 2017 03.
Article in English | MEDLINE | ID: mdl-27932312

ABSTRACT

Parietal cell atrophy is considered to cause metaplasia in the stomach. We developed mice that express the diphtheria toxin receptor specifically in parietal cells to induce their death, and found this to increase proliferation in the normal stem cell zone and neck but not to cause metaplastic reprogramming of chief cells. Furthermore, the metaplasia-inducing agents tamoxifen or DMP-777 still induced metaplasia even after previous destruction of parietal cells by diphtheria toxin. Atrophy of parietal cells alone therefore is not sufficient to induce metaplasia: completion of metaplastic reprogramming of chief cells requires mechanisms beyond parietal cell injury or death.


Subject(s)
Apoptosis , Chief Cells, Gastric/pathology , Parietal Cells, Gastric/pathology , Parietal Cells, Gastric/physiology , Stomach/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Atrophy/chemically induced , Azetidines , Cell Proliferation , Cellular Reprogramming , Chief Cells, Gastric/metabolism , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Intercellular Signaling Peptides and Proteins , Intrinsic Factor/metabolism , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/metabolism , Mice , Parietal Cells, Gastric/drug effects , Peptides/metabolism , Piperazines , Plant Lectins/metabolism , Tamoxifen
8.
Regul Toxicol Pharmacol ; 92: 55-66, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29158042

ABSTRACT

An inhalation reference concentration (RfC) was developed for diethanolamine (DEA), based principally on evaluation of three animal studies (Gamer et al., 1993, 1996, 2008). The RfC (25 µg/m3) was based on statistically significantly increased relative liver weight in female rats in Gamer et al. (2008) as the critical effect. The lower confidence limit on the benchmark dose (BMDL10 of 5.5 mg/m3) was adjusted to a human equivalent concentration and to continuous exposure before dividing the final point of departure (2.3 mg/m3) by a total factor of 90 that considered standard key areas of uncertainty (intrahuman variability, potential interspecies toxicodynamic differences, database limitations). While laryngeal effects observed in Gamer et al. (2008) were also considered as candidate critical effects, evaluation of the adversity and human relevance of rat laryngeal squamous metaplasia and concomitant effects at the various exposure levels resulted in identifying a LOAEL for laryngeal squamous hyperplasia and chronic inflammation that was much higher than the liver weight LOAEL identified. The RfC of 25 µg/m3 is considered health protective for the general population and can be used to evaluate the potential health effects of long-term environmental exposure of the general public (i.e., long-term, ambient air dispersion modelling or monitoring data).


Subject(s)
Ethanolamines/administration & dosage , Ethanolamines/chemistry , Animals , Ethanolamines/adverse effects , Female , Humans , Hyperplasia/chemically induced , Inflammation/chemically induced , Inhalation/drug effects , Laryngeal Diseases/chemically induced , Male , Metaplasia/chemically induced , Rats
9.
Dermatol Online J ; 23(9)2017 Sep 15.
Article in English | MEDLINE | ID: mdl-29469718

ABSTRACT

Eccrine squamous syringometaplasia (ESS) is a rare finding defined as metaplastic change of the cuboidal epithelial cells of eccrine glands into two or more layers of squamous epithelial cells. We present a patient who developed ESS after induction of CLAG chemotherapy [2-Chlorodeoxyadenosine (2-CdA) with cytarabine (Ara-C) and (granulocyte-colony stimulating factor) G-CSF] for management of the blast crisis of his chronic myelogenous leukemia (CML). Our patient's ESS eruption presented with a variety of morphologies, thus multiple skin biopsies were taken to determine the possible diagnosis(es). All skin biopsies showed ESS and the eruption resolved with topical corticosteroids after CLAG therapy was finished.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Eruptions/etiology , Eccrine Glands/pathology , Epithelial Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Adult , Cladribine/administration & dosage , Cytarabine/administration & dosage , Drug Eruptions/pathology , Fatal Outcome , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Male , Metaplasia/chemically induced , Stem Cell Transplantation , Transplantation, Homologous
10.
Toxicol Ind Health ; 32(6): 980-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-24958741

ABSTRACT

Nonsteroidal anti-inflammatory (NSAI) drugs are the most commonly used group of drugs today. Increase in the use of standard NSAI for treating pain and inflammation was restricted by the fact that these drugs were proven to possibly cause gastrointestinal and renal toxicity. Meloxicam is a NSAI that has anti-inflammatory, analgesic, and antipyretic effects. This study aims to investigate the effects of meloxicam on stomach, kidney, and liver of rats under light microscopy level. Based on the light microscopic observations, mononuclear cell infiltration and pseudolobular formation was established in liver samples of animals in the experimental group. Metaplasia in surface and glandular epithelia and atrophy were observed in stomach samples. Glomerular stasis-related hypertrophy and focal interstitial nephritis were found in kidneys. It was concluded in this study that meloxicam might cause hepatotoxicity, nephrotoxicity, and gastric metaplasia in rats at a used dose and duration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Kidney/drug effects , Liver/drug effects , Stomach/drug effects , Thiazines/pharmacology , Thiazoles/pharmacology , Animals , Chemical and Drug Induced Liver Injury/diagnosis , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation/drug therapy , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/diagnosis , Liver/pathology , Meloxicam , Metaplasia/chemically induced , Pain/drug therapy , Rats , Rats, Sprague-Dawley , Stomach/pathology , Stomach Diseases/chemically induced , Stomach Diseases/diagnosis , Toxicity Tests
11.
Thorax ; 70(7): 668-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25995156

ABSTRACT

BACKGROUND: It has been shown that acetylcholine is both a neurotransmitter and acts as a local mediator, produced by airway cells including epithelial cells. In vivo studies have demonstrated an indirect role for acetylcholine in epithelial cell differentiation. Here, we aimed to investigate direct effects of endogenous non-neuronal acetylcholine on epithelial cell differentiation. METHODS: Human airway epithelial cells from healthy donors were cultured at an air-liquid interface (ALI). Cells were exposed to the muscarinic antagonist tiotropium (10 nM), interleukin (IL)-13 (1, 2 and 5 ng/mL), or a combination of IL-13 and tiotropium, during or after differentiation at the ALI. RESULTS: Human airway epithelial cells expressed all components of the non-neuronal cholinergic system, suggesting acetylcholine production. Tiotropium had no effects on epithelial cell differentiation after air exposure. Differentiation into goblet cells was barely induced after air exposure. Therefore, IL-13 (1 ng/mL) was used to induce goblet cell metaplasia. IL-13 induced MUC5AC-positive cells (5-fold) and goblet cells (14-fold), as assessed by histochemistry, and MUC5AC gene expression (105-fold). These effects were partly prevented by tiotropium (47-92%). Goblet cell metaplasia was induced by IL-13 in a dose-dependent manner, which was inhibited by tiotropium. In addition, tiotropium reversed goblet cell metaplasia induced by 2 weeks of IL-13 exposure. IL-13 decreased forkhead box protein A2 (FoxA2) expression (1.6-fold) and increased FoxA3 (3.6-fold) and SAM-pointed domain-containing ETS transcription factor (SPDEF) (5.2-fold) expression. Tiotropium prevented the effects on FoxA2 and FoxA3, but not on SPDEF. CONCLUSIONS: We demonstrate that tiotropium has no effects on epithelial cell differentiation after air exposure, but inhibits and reverses IL-13-induced goblet cell metaplasia, possibly via FoxA2 and FoxA3. This indicates that non-neuronal acetylcholine contributes to goblet cell differentiation by a direct effect on epithelial cells.


Subject(s)
Goblet Cells/drug effects , Interleukin-13/antagonists & inhibitors , Respiratory Mucosa/drug effects , Scopolamine Derivatives/pharmacology , Acetylcholine/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cholinergic Antagonists/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Goblet Cells/pathology , Humans , Interleukin-13/administration & dosage , Interleukin-13/pharmacology , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/pathology , Mucin 5AC/biosynthesis , Mucin 5AC/genetics , Respiratory Mucosa/pathology , Tiotropium Bromide , Transcription Factors/metabolism
13.
Cochrane Database Syst Rev ; (12): CD010623, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25464111

ABSTRACT

BACKGROUND: Proton pump inhibitors (PPIs) are the most effective drugs to reduce gastric acid secretion. PPIs are one of the most commonly prescribed classes of medications worldwide. Apart from short-term application, maintenance therapy with PPIs is recommended and increasingly used in certain diseases, such as Zollinger-Ellison syndrome and gastro-oesophageal reflux disease, especially for people with erosive oesophagitis or Barrett's oesophagus. Although PPIs are generally safe, their efficacy and safety of long-term use remains unclear. The question of whether the long-term use of PPIs could promote the development of gastric pre-malignant lesions has been widely investigated, but results are inconsistent. Limited insight on this problem leads to a dilemma in decision making for long-term PPI prescription. OBJECTIVES: To compare the development or progression of gastric pre-malignant lesions, such as atrophic gastritis, intestinal metaplasia, enterochromaffin-like (ECL) cell hyperplasia, and dysplasia, in people taking long-term (six months or greater) PPI maintenance therapy. SEARCH METHODS: We searched the following databases (from inception to 6 August 2013): the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, and CINAHL. In addition, we searched the reference lists of included trials and contacted experts in the field. SELECTION CRITERIA: We searched for randomised controlled trials (RCTs) in adults (aged 18 years or greater) concerning the effects of long-term (six months or greater) PPI use on gastric mucosa changes, confirmed by endoscopy or biopsy sampling (or both). DATA COLLECTION AND ANALYSIS: Two review authors independently performed selection of eligible trials, assessment of trial quality, and data extraction. We calculated odds ratios (OR) for analysis of dichotomous data and mean differences for continuous data, with 95% confidence intervals (CI). MAIN RESULTS: We included seven trials (1789 participants). Four studies had high risk of bias and the risk of bias in the other three trials was unclear. In addition, it was difficult to assess possible reporting bias. We pooled 1070 participants from four RCTs to evaluate corporal atrophy development revealing an insignificantly increased OR of 1.50 (95% CI 0.59 to 3.80; P value = 0.39; low-quality evidence) for long-term PPI users relative to non-PPI users. In five eligible trials, corporal intestinal metaplasia was assessed among 1408 participants, also with uncertain results (OR 1.46; 95% CI 0.43 to 5.03; P value = 0.55; low-quality evidence). However, by pooling data of 1705 participants from six RCTs, our meta-analysis showed that participants with PPI maintenance treatment were more likely to experience either diffuse (simple) (OR 5.01; 95% CI 1.54 to 16.26; P value = 0.007; very-low-quality evidence) or linear/micronodular (focal) ECL hyperplasia (OR 3.98; 95% CI 1.31 to 12.16; P value = 0.02; low-quality evidence) than controls. No participant showed any dysplastic or neoplastic change in any included studies. AUTHORS' CONCLUSIONS: There is presently no clear evidence that the long-term use of PPIs can cause or accelerate the progression of corpus gastric atrophy or intestinal metaplasia, although results were imprecise. People with PPI maintenance treatment may have a higher possibility of experiencing either diffuse (simple) or linear/micronodular (focal) ECL cell hyperplasia. However, the clinical importance of this outcome is currently uncertain.


Subject(s)
Precancerous Conditions/chemically induced , Proton Pump Inhibitors/adverse effects , Stomach Neoplasms/chemically induced , Enterochromaffin-like Cells/pathology , Gastritis, Atrophic/chemically induced , Humans , Hyperplasia/chemically induced , Intestines/pathology , Maintenance Chemotherapy/adverse effects , Metaplasia/chemically induced , Precancerous Conditions/pathology , Randomized Controlled Trials as Topic , Stomach Neoplasms/pathology , Time Factors
14.
Dermatology ; 226(4): 362-4, 2013.
Article in English | MEDLINE | ID: mdl-23860306

ABSTRACT

We report herein a patient treated with vemurafenib for a stage IV melanoma who developed an eruption related to eccrine squamous syringometaplasia (ESS). This entity is a well-described side effect of cytostatic therapies used for malignant neoplasia and is clinically characterized by a symmetric cutaneous eruption composed of papules and vesicles preferentially located on fold and intertriginous areas. It is histologically defined by a squamous metaplasia of eccrine ductal epithelium. ESS represents another skin eruption to be added to the list of cutaneous adverse events associated with vemurafenib, a selective BRAF inhibitor used to treat patients with metastatic melanoma harboring the V600E mutation. The discussion focuses on the pathogenesis of ESS secondary to vemurafenib and on alternative diagnoses.


Subject(s)
Antineoplastic Agents/adverse effects , Drug Eruptions/etiology , Eccrine Glands/pathology , Epithelial Cells/pathology , Indoles/adverse effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Sulfonamides/adverse effects , Adult , Drug Eruptions/pathology , Humans , Male , Melanoma/secondary , Metaplasia/chemically induced , Metaplasia/pathology , Skin Neoplasms/pathology , Vemurafenib
15.
Klin Oczna ; 115(1): 34-9, 2013.
Article in Polish | MEDLINE | ID: mdl-23882737

ABSTRACT

PURPOSE: Evaluation of different types of chemotherapy schemes administered in lung, breast and bowel cancer on conjunctival epithelium and goblet cells morphology. MATERIAL AND METHODS: 36 patients (72 eyes) were enrolled to the study. Patients were divided into three groups depending on type of cancer and chemotherapy: group I - patients diagnosed with non- small cells lung cancer treated with PE schema (cisplatin, etoposide), group II - with breast cancer treated with FAC schema (fluorouracil, doxorubicin, cyclophosphamide), group Ill - bowel cancer treated with FU/LV schema (fluorouracil, leucovorin). Examinations were performed before chemotherapy and after Il'th, IV'th, VI'th chemotherapy cycle. Conjuntival specimen were obtained with exfoliative cytology, stained with PAS and hematoxyline. RESULTS: Statistically significant deterioration of conjunctival epithelium and goblet cells in all the groups in each time of examination (p<0.001) was observed. Alterations were aggravated with duration of chemotherapy. Before chemotherapy all the patients had normal epithelium and goblet cells (grade 0 or 1 according to the Nelson's scale). Conjunctival cells status gradually deteriorated and altered from the normal glandular epithelium to the squamous cells epithelium through the process of squamous metaplasia. In further chemotherapy cycles each patient (1,0 fraction) had abnormal morphology of epithelium and goblet cells (grade 2 or 3 of Nelson's scale). CONCLUSIONS: Chemotherapy induces squamous metaplasia of epithelium and the reduction of number of conjunctival goblet cells. This abnormalities were time dependent and increased with duration of chemotherapy and were not depended on type of chemotherapy scheme.


Subject(s)
Antineoplastic Agents/adverse effects , Conjunctiva/drug effects , Conjunctival Diseases/chemically induced , Epithelial Cells/drug effects , Goblet Cells/drug effects , Adult , Aged , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Colonic Neoplasms/drug therapy , Female , Humans , Male , Metaplasia/chemically induced , Middle Aged , Risk Factors
16.
Gut ; 60(5): 624-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21097844

ABSTRACT

BACKGROUND: We investigated whether corpus atrophic gastritis worsens in Mongolian gerbils (MGs) after long-term administration of proton pump inhibitor (PPI). MGs are an excellent model for studying Helicobacter pylori-related gastritis and adenocarcinoma. METHODS: MGs were separated into four groups (n =15/group); H pylori (ATCC43504) was inoculated into the OPZ(omeprazole)+Hp (H pylori) and Hp groups, a PPI (OPZ) was administered to the OPZ+Hp and OPZ groups and the control group received no treatment. MGs had access to food containing omeprazole (100 mg/kg body weight/day) for 6 months, after which their stomachs were removed and cut into nine sections (six sections in the fundus and three sections in the antrum). Corpus atrophy was evaluated by the absence of parietal cells in the six sections in the fundus. First, we calculated a percentage of the area devoid of parietal cells in each haematoxylin and eosin-stained section, and then we scored the degree of atrophy by adding the percentages of the six sections. A full score was 600. RESULTS: Neutrophilic and lymphoid infiltrates were greater in the OPZ+Hp group than in the other groups. The corpus atrophy score in the OPZ+Hp group was significantly higher than that in the Hp group (p < 0.0048, Student t test). Significantly more adenocarcinomas were found in the OPZ+Hp (60%) than in the Hp (7%) group animals. CONCLUSION: Long-term PPI administration promotes development of adenocarcinoma, which is associated with the progression of atrophic corpus gastritis in MGs infected with H pylori.


Subject(s)
Adenocarcinoma/chemically induced , Helicobacter Infections/complications , Helicobacter pylori , Proton Pump Inhibitors/toxicity , Stomach Neoplasms/chemically induced , Adenocarcinoma/microbiology , Animals , Body Weight , Cocarcinogenesis , Disease Models, Animal , Disease Progression , Drug Administration Schedule , Gastrins/blood , Gastritis, Atrophic/chemically induced , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Gerbillinae , Male , Metaplasia/chemically induced , Metaplasia/microbiology , Proton Pump Inhibitors/administration & dosage , Stomach/pathology , Stomach Neoplasms/microbiology
17.
Intern Med ; 61(24): 3675-3682, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35527030

ABSTRACT

The histopathological findings of immune checkpoint inhibitor (ICI)-induced pancreatitis have rarely been reported. A 56-year-old man with squamous cell carcinoma of the lung with bone metastasis was being treated with pembrolizumab, an anti-programmed cell death protein-1 antibody. After 13 doses, he was referred to our department due to pancreatitis. Despite characteristic symptoms of acute pancreatitis, imaging findings were similar to those of autoimmune pancreatitis. However, a histological examination showed neutrophil-based inflammatory cell infiltration and acinar-ductal metaplasia. Immunostaining showed CD8-positive T lymphocyte infiltration. This case revealed the characteristic histopathology of pembrolizumab-induced pancreatitis, which was previously poorly understood.


Subject(s)
Pancreatitis , Male , Humans , Middle Aged , Pancreatitis/chemically induced , Pancreatitis/pathology , Neutrophil Infiltration , Acute Disease , Metaplasia/chemically induced
18.
Gut Microbes ; 14(1): 2120744, 2022.
Article in English | MEDLINE | ID: mdl-36067404

ABSTRACT

Intestinal metaplasia (IM) is the inevitable precancerous stage to develop intestinal-type gastric cancer (GC). Deoxycholic acid (DCA) is the main bile acid (BA) component of duodenogastric reflux and has shown an increased concentration during the transition from chronic gastritis to IM associated with continued STAT3 activation. However, the mechanisms underlying how DCA facilitates IM in the gastric epithelium need exploration. We evaluated IM and bile reflux in corpus tissues from 161 subjects undergoing GC screening. Cell survival and proliferation, proinflammatory cytokine expression and TGR5/STAT3/KLF5 axis activity were measured in normal human gastric cells, cancer cells, and organoid lines derived from C57BL/6, FVB/N and insulin-gastrin (INS-GAS) mice treated with DCA. The effects of DCA on IM development were determined in INS-GAS mice with long-term DCA supplementation, after which the gastric bacterial and BA metabolic profiles were measured by 16S rRNA gene sequencing and LC-MS. We revealed a BA-triggered TGR5/STAT3/KLF5 pathway in human gastric IM tissues. In gastric epithelial cells, DCA promoted proliferation and apoptotic resistance, upregulated proinflammatory cytokines and IM markers, and facilitated STAT3 phosphorylation, nuclear accumulation and DNA binding to the KLF5 promoter. DCA triggered STAT3 signaling and the downstream IM marker KLF5 in mouse gastric organoids in vitro and in vivo. In INS-GAS mice, DCA promoted the accumulation of serum total BAs and accelerated the stepwise development of gastric IM and dysplasia. DCA induced gastric environmental alterations involving abnormal BA metabolism and microbial dysbiosis, in which the Gemmobacter and Lactobacillus genera were specifically enriched. Lactobacillus genus enrichment was positively correlated with increased levels of GCA, CA, T-α-MCA, TCA and ß-MCA in DCA-administrated INS-GAS mice. DCA promotes nuclear STAT3 phosphorylation, which mediates KLF5 upregulation associated with gastric inflammation and IM development. DCA disturbs the gastric microbiome and BA metabolism homeostasis during IM induction.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Precancerous Conditions , Animals , Bile Acids and Salts , Deoxycholic Acid/toxicity , Humans , Metaplasia/chemically induced , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
19.
Trials ; 23(1): 437, 2022 May 24.
Article in English | MEDLINE | ID: mdl-35610625

ABSTRACT

BACKGROUND: Multifocal atrophic gastritis and intestinal metaplasia are considered to be important links in the gastric precancerous cascade. However, there are no specific drugs for these conditions. Although many studies have shown that traditional Chinese medicine is effective with no serious side effects, these studies have not been scientifically rigorous trials. Our aim is to design a high-quality trial for a Chinese patent medicine, Elian Granules, to investigate its efficacy and safety in treating patients with chronic atrophic gastritis with or without intestinal metaplasia. METHODS: This is a phase II, randomized, double-blind, placebo-controlled, multicenter clinical trial. A total of 240 participants will be assigned to a treatment or placebo control group in a 1:1 ratio. The experimental drug or placebo will be taken with boiling water, two small bags (24.2 g) each time, twice a day, half an hour after a meal, for 24 weeks. The primary outcome is the observation of histological changes in the gastric mucosa of patients with atrophic gastritis with or without intestinal metaplasia after 6 months based on the OLGA/OLGIM staging systems. The secondary outcomes include the assessment of dyspepsia and quality of life based on the dyspepsia symptom score and the quality-of-life scale. DISCUSSION: This study is designed to evaluate the efficacy and safety of Elian Granules in a randomized, double-blind, placebo-controlled, multicenter manner. This trial may not only provide evidence for a phase III clinical trial, but also an alternative option for the treatment of chronic atrophic gastritis (CAG). TRIAL REGISTRATION: Registry Platform For Evidence-Based Traditional Chinese Medicine ChiMCTR2000003929 . Registered on 13 September 2020.


Subject(s)
Drugs, Chinese Herbal , Dyspepsia , Gastritis, Atrophic , Clinical Trials, Phase II as Topic , Double-Blind Method , Drugs, Chinese Herbal/adverse effects , Dyspepsia/drug therapy , Gastritis, Atrophic/complications , Gastritis, Atrophic/diagnosis , Gastritis, Atrophic/drug therapy , Humans , Medicine, Chinese Traditional , Metaplasia/chemically induced , Metaplasia/complications , Metaplasia/drug therapy , Multicenter Studies as Topic , Quality of Life , Randomized Controlled Trials as Topic , Treatment Outcome
20.
Pharmacology ; 87(1-2): 56-62, 2011.
Article in English | MEDLINE | ID: mdl-21228611

ABSTRACT

OBJECTIVES: The purpose of this study was to develop an animal model for the study of mucus overproduction and to assess the effect of a 14-membered macrolide antibiotic and a glucocorticoid on lipopolysaccharide (LPS)-induced mucus production. METHODS: Tracheas from donor rats were homografted to recipient rats for 4 weeks, and the usefulness of this tracheal homograft model in the study of mucus production was examined. RESULTS: Oral administration of clarithromycin (CAM) to recipient rats for 4 weeks significantly reduced LPS-induced mucus production in the homografted trachea. Dexamethasone administered for 4 weeks also significantly reduced the mucus volume in LPS-treated homografted trachea compared with that in the control rats. The implanted trachea containing control medium was not histologically different from normal trachea. When the medium instilled into the implanted trachea contained 1 µg/ml LPS, the volume and spinability of mucus produced in the tracheal lumen were significantly increased compared to those in the trachea instilled with control medium. Goblet cell metaplasia was also observed in the implanted trachea containing LPS. CONCLUSIONS: The present study shows that LPS-administered homografted trachea is a good animal model of chronic hypersecretory diseases of the upper and lower airways. CAM and dexamethasone could be treatment choices in such hypersecretory diseases.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Clarithromycin/therapeutic use , Dexamethasone/analogs & derivatives , Disease Models, Animal , Glucocorticoids/therapeutic use , Mucus/metabolism , Trachea/drug effects , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacokinetics , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Clarithromycin/metabolism , Clarithromycin/pharmacokinetics , Dexamethasone/therapeutic use , Goblet Cells/drug effects , Goblet Cells/metabolism , Goblet Cells/pathology , Lipopolysaccharides/toxicity , Male , Metaplasia/chemically induced , Mucus/chemistry , Rats , Rats, Inbred F344 , Secretory Pathway/drug effects , Severity of Illness Index , Trachea/metabolism , Trachea/pathology , Trachea/transplantation , Tracheitis/drug therapy , Tracheitis/metabolism , Tracheitis/pathology , Tracheitis/physiopathology , Transplantation, Isogeneic , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL