Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Dig Dis Sci ; 69(4): 1156-1168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448762

ABSTRACT

BACKGROUND/AIMS: We examined the involvement of cholecystokinin (CCK) in the exacerbation of indomethacin (IND)-induced gastric antral ulcers by gastroparesis caused by atropine or dopamine in mice. METHODS: Male mice were fed for 2 h (re-feeding) following a 22-h fast. Indomethacin (IND; 10 mg/kg, s.c.) was administered after re-feeding; gastric lesions were examined 24 h after IND treatment. In another experiment, mice were fed for 2 h after a 22-h fast, after which the stomachs were removed 1.5 h after the end of the feeding period. Antral lesions, the amount of gastric contents, and the gastric luminal bile acids concentration were measured with or without the administration of the pro- and antimotility drugs CCK-octapeptide (CCK-8), atropine, dopamine, SR57227 (5-HT3 receptor agonist), apomorphine, lorglumide (CCK1 receptor antagonist), ondansetron, and haloperidol alone and in combination. RESULTS: IND produced severe lesions only in the gastric antrum in re-fed mice. CCK-8, atropine, dopamine, SR57227 and apomorphine administered just after re-feeding increased bile reflux and worsened IND-induced antral lesions. These effects were significantly prevented by pretreatment with lorglumide. Although atropine and dopamine also increased the amount of gastric content, lorglumide had no effect on the delayed gastric emptying provoked by atropine and dopamine. Both ondansetron and haloperidol significantly inhibited the increase of bile reflux and the exacerbation of antral lesions induced by atropine and dopamine, respectively, but did not affect the effects of CCK-8. CONCLUSIONS: These results suggest that CCK-CCK1 receptor signal increases bile reflux during gastroparesis induced by atropine and dopamine, exacerbating IND-induced antral ulcers.


Subject(s)
Bile Reflux , Gastroparesis , Stomach Ulcer , Mice , Male , Animals , Indomethacin , Ulcer , Receptor, Cholecystokinin A , Sincalide/adverse effects , Apomorphine/adverse effects , Dopamine , Haloperidol/adverse effects , Ondansetron , Stomach Ulcer/chemically induced , Cholecystokinin/adverse effects , Receptors, Cholecystokinin , Atropine/adverse effects
2.
Dig Dis Sci ; 68(10): 3886-3901, 2023 10.
Article in English | MEDLINE | ID: mdl-37632663

ABSTRACT

BACKGROUND/AIMS: We examined the contributions of gastric emptying and duodenogastric bile reflux in the formation of gastric antral ulcers induced by NSAIDs in mice. METHODS: We used the murine re-fed indomethacin (IND) experimental ulcer model. Outcome measures included the appearance of gastric lesions 24 h after IND treatment and the assessment of gastric contents and the concentration of bile acids 1.5 h after re-feeding. The effects of atropine, dopamine, SR57227 (5-HT3 receptor agonist), apomorphine, ondansetron, haloperidol, and dietary taurocholate and cholestyramine were also examined. RESULTS: IND (10 mg/kg, s.c.) induced severe lesions only in the gastric antrum in the re-fed model. The antral lesion index and the amount of food intake during the 2-h refeeding period were positively correlated. Atropine and dopamine delayed gastric emptying, increased bile reflux, and worsened IND-induced antral lesions. SR57227 and apomorphine worsened antral lesions with increased bile reflux. These effects were prevented by the anti-emetic drugs ondansetron and haloperidol, respectively. The anti-emetic drugs markedly decreased the severity of antral lesions and the increase of bile reflux induced by atropine or dopamine without affecting delayed gastric emptying. Antral lesions induced by IND were increased by dietary taurocholate but decreased by the addition of the bile acid sequestrant cholestyramine. CONCLUSIONS: These results suggest that gastroparesis induced by atropine or dopamine worsens NSAID-induced gastric antral ulcers by increasing duodenogastric bile reflux via activation of 5-HT3 and dopamine D2 receptors.


Subject(s)
Antiemetics , Bile Reflux , Duodenogastric Reflux , Gastroparesis , Stomach Ulcer , Mice , Animals , Indomethacin , Dopamine , Ulcer , Gastroparesis/chemically induced , Serotonin , Apomorphine/adverse effects , Antiemetics/adverse effects , Ondansetron/pharmacology , Cholestyramine Resin/adverse effects , Haloperidol/adverse effects , Stomach Ulcer/chemically induced , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Atropine/adverse effects
3.
Cell Mol Gastroenterol Hepatol ; 16(4): 557-572, 2023.
Article in English | MEDLINE | ID: mdl-37369278

ABSTRACT

BACKGROUND & AIMS: Metabolic syndrome (MetS) is characterized by obesity, glucose intolerance, and hepatic steatosis. Alterations in the gut microbiome play important roles in the development of MetS. However, the mechanisms by which this occurs are poorly understood. Dual oxidase 2 (DUOX2) is an antimicrobial reduced nicotinamide adenine dinucleotide phosphate oxidase expressed in the gut epithelium. Here, we posit that epithelial DUOX2 activity provides a mechanistic link between the gut microbiome and the development of MetS. METHODS: Mice carrying an intestinal epithelial-specific deletion of dual oxidase maturation factor 1/2 (DA IEC-KO), and wild-type littermates were fed a standard diet and killed at 24 weeks. Metabolic alterations were determined by glucose tolerance, lipid tests, and body and organ weight measurements. DUOX2 activity was determined by Amplex Red. Intestinal permeability was determined by fluorescein isothiocyanate-dextran, microbial translocation assessments, and portal vein lipopolysaccharide measurements. Metagenomic analysis of the stool microbiome was performed. The role of the microbiome was assessed in antibiotic-treated mice. RESULTS: DA IEC-KO males showed increased body and organ weights accompanied by glucose intolerance and increased plasma lipid and liver enzyme levels, and increased adiposity in the liver and adipose tissue. Expression of F4/80, CD68, uncoupling protein 1, carbohydrate response element binding protein, leptin, and adiponectin was altered in the liver and adipose tissue of DA IEC-KO males. DA IEC-KO males produced less epithelial H2O2, had altered relative abundance of Akkermansiaceae and Lachnospiraceae in stool, and showed increased portal vein lipopolysaccharides and intestinal permeability. Females were protected from barrier defects and MetS, despite producing less H2O2. Antibiotic depletion abrogated all MetS phenotypes observed. CONCLUSIONS: Intestinal epithelial inactivity of DUOX2 promotes MetS in a microbiome-dependent manner.


Subject(s)
Gastrointestinal Microbiome , Glucose Intolerance , Metabolic Syndrome , Animals , Female , Male , Mice , Anti-Bacterial Agents , Dual Oxidases , Hydrogen Peroxide , Lipopolysaccharides , Obesity/metabolism
4.
Sci Rep ; 12(1): 20935, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36463312

ABSTRACT

The lactoperoxidase (LPO)-hydrogen peroxide-halides reaction (LPO system) converts iodide and thiocyanate (SCN-) into hypoiodous acid (HOI) and hypothiocyanite (OSCN-), respectively. Since this system has been implicated in defense of the airways and oropharynx from microbial invasion, in this proof-of-concept study we measured the concentrations of these analytes in human saliva from a convenience clinical sample of 40 qualifying subjects before and after acute iodine administration via the iodinated contrast medium used in coronary angiography to test the hypothesis that an iodide load increases salivary iodide and HOI concentrations. Saliva was collected and salivary iodide, SCN-, HOI and OSCN- were measured using standard methodology. The large iodine load delivered by the angiographic dye, several 100-fold in excess of the U.S. Recommended Daily Allowance for iodine (150 µg/day), significantly increased salivary iodide and HOI levels compared with baseline levels, whereas there was no significant change in salivary SCN- and OSCN- levels. Iodine load and changes of salivary iodide and HOI levels were positively correlated, suggesting that higher iodide in the circulation increases iodide output and salivary HOI production. This first of its kind study suggests that a sufficient but safe iodide supplementation less than the Tolerable Upper Limit for iodine set by the U.S. Institute of Medicine (1,100 µg/day) may augment the generation of antimicrobial HOI by the salivary LPO system in concentrations sufficient to at least in theory protect the host against susceptible airborne microbial pathogens, including enveloped viruses such as coronaviruses and influenza viruses.


Subject(s)
Anti-Infective Agents , Iodine , United States , Humans , Iodides , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents , Coronary Angiography
5.
Front Med (Lausanne) ; 9: 1033601, 2022.
Article in English | MEDLINE | ID: mdl-36530869

ABSTRACT

Background: Iodine and particularly its oxidated forms have long been recognized for its effective antiseptic properties. Limited in vitro and in vivo data suggest that iodine exposure may rapidly inactivate, reduce transmission, and reduce infectivity of SARS-CoV-2. We hypothesized that iodine exposure may be associated with decreased incident COVID-19 infection. Methods: A retrospective population-level cohort analysis was performed of the U.S. Veterans Health Administration between 1 March 2020 and 31 December 2020, before the widespread availability of vaccines against SARS-CoV-2. Multivariable logistic regression models estimated the adjusted odds ratios (OR) and 95% confidence intervals (CI) of the associations between iodinated contrast exposure and incident COVID-19 infection, adjusting for age, sex, race/ethnicity, place of residence, socioeconomic status, and insurance status. Results: 530,942 COVID-19 tests from 333,841 Veterans (mean ± SD age, 62.7 ± 15.2 years; 90.2% men; 61.9% non-Hispanic Whites) were analyzed, of whom 9% had received iodinated contrast ≤60 days of a COVID-19 test. Iodine exposure was associated with decreased incident COVID-19 test positivity (OR, 0.75 95% CI, 0.71-0.78). In stratified analyses, the associations between iodinated contrast use and decreased COVID-19 infection risk did not differ by age, sex, and race/ethnicity. Conclusion: Iodine exposure may be protective against incident COVID-19 infection. Weighed against the risks of supraphysiologic iodine intake, dietary, and supplemental iodine nutrition not to exceed its Tolerable Upper Limit may confer an antimicrobial benefit against SARS-CoV-2. A safe but antimicrobial level of iodine supplementation may be considered in susceptible individuals, particularly in geographic regions where effective COVID-19 vaccines are not yet readily available.

6.
Neurogastroenterol Motil ; 34(10): e14378, 2022 10.
Article in English | MEDLINE | ID: mdl-35388579

ABSTRACT

BACKGROUND: Low-grade duodenal inflammation has recently been identified in patients with functional dyspepsia (FD). Chemosensory tuft cells were reported to be associated with gastrointestinal diseases. We therefore assessed duodenal tuft cell density and microinflammation in patients with FD to determine whether these measures could serve as useful biomarkers, and also correlated tuft cell density and microinflammation in FD patients. METHODS: Duodenal biopsy specimens were obtained from patients with FD and from controls. Tuft cells, eosinophils, and mast cells were immunochemically stained with specific antibodies. Tuft cells were identified by immunostaining for doublecortin-like kinase 1 (DCLK1); cholinergic tuft cells were assessed by double staining for choline acetyltransferase (ChAT) and DCLK1. Immune-type tuft cells were assessed by IL-25 mRNA expression using real-time PCR. KEY RESULTS: The density of intramucosal eosinophils and mast cells was significantly higher in the duodenum of FD patients than in controls. The density of tuft cells was significantly higher in the duodenum of FD patients compared with controls, and significantly correlated with eosinophil density in the duodenum of FD patients and controls. Moreover, a fraction of ChAT-positive cells was DCLK1 positive; all duodenal DCLK1+ tuft cells were ChAT-immunoreactive in FD and in control subjects. CONCLUSIONS AND INFERENCES: Cholinergic tuft cell density was higher in the duodenum of patients with FD and significantly correlated with eosinophil density. Further studies are needed to investigate the pathophysiological significance of tuft cells in FD and may provide valuable clues to the pathophysiology of FD.


Subject(s)
Dyspepsia , Biomarkers/metabolism , Cell Count , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/metabolism , Doublecortin-Like Kinases , Duodenum/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism
7.
Eur J Pharmacol ; 906: 174292, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34216575

ABSTRACT

Short-chain fatty acids (SCFAs) produced by the microbial fermentation of carbohydrates are important energy substrates for mammals. Intestinal epithelia respond to these metabolites by stimulation of anion secretion via the release of epithelial acetylcholine. The present experiments were performed to discover which of the known receptors for SCFAs are expressed in rat caecum, the most important site of fermentation within the intestine of non-ruminant mammals. Using the increase in short-circuit current (Isc) induced by anion secretion as the readout, the order of efficiency of the tested SCFAs in rat caecum was propionate > butyrate > acetate. Both synthetic high-affinity selective free fatty acid (FFA) receptor agonists 4-CMTB (FFA2 receptor) and AR420626 (FFA3 receptor) partially mimicked the effect of propionate on Isc (IProp). IProp was concentration-dependently inhibited by the FFA3 receptor antagonist ß-OH-butyrate. Although no antagonist of rat FFA2 receptor is available, coadministration of the allosteric FFA2 receptor agonist 4-CMTB together with a low concentration of propionate potentiated IProp, suggesting that FFA2 receptor is involved in sensing of short-chain fatty acids as well. The expression of both receptor types was confirmed by qPCR (with FFA2 > FFA3 receptor). Immunohistochemical staining revealed the localization of FFA2 receptor in the surface epithelium and the FFA3 receptor expression predominantly in enteroendocrine cells and subepithelial nerve-like fibers. Taken together, the present results demonstrate that the anion secretion induced by the microbial metabolite propionate in rat caecum is enhanced by activation of FFA2 and FFA3 receptor expressed in different cell types within the caecal epithelium.


Subject(s)
Acetylcholine/metabolism , Cecum/metabolism , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cecum/drug effects , Fatty Acids, Volatile/metabolism , Female , Intestinal Mucosa/drug effects , Male , Models, Animal , Rats , Receptors, G-Protein-Coupled/agonists
8.
J Clin Invest ; 131(9)2021 05 03.
Article in English | MEDLINE | ID: mdl-33651715

ABSTRACT

A primordial gut-epithelial innate defense response is the release of hydrogen peroxide by dual NADPH oxidase (DUOX). In inflammatory bowel disease (IBD), a condition characterized by an imbalanced gut microbiota-immune homeostasis, DUOX2 isoenzyme is the highest induced gene. Performing multiomic analyses using 2872 human participants of a wellness program, we detected a substantial burden of rare protein-altering DUOX2 gene variants of unknown physiologic significance. We identified a significant association between these rare loss-of-function variants and increased plasma levels of interleukin-17C, which is induced also in mucosal biopsies of patients with IBD. DUOX2-deficient mice replicated increased IL-17C induction in the intestine, with outlier high Il17c expression linked to the mucosal expansion of specific Proteobacteria pathobionts. Integrated microbiota/host gene expression analyses in patients with IBD corroborated IL-17C as a marker for epithelial activation by gram-negative bacteria. Finally, the impact of DUOX2 variants on IL-17C induction provided a rationale for variant stratification in case control studies that substantiated DUOX2 as an IBD risk gene. Thus, our study identifies an association of deleterious DUOX2 variants with a preclinical hallmark of disturbed microbiota-immune homeostasis that appears to precede the manifestation of IBD.


Subject(s)
Dual Oxidases , Gastrointestinal Microbiome/immunology , Genetic Variation , Homeostasis , Inflammatory Bowel Diseases , Animals , Dual Oxidases/genetics , Dual Oxidases/immunology , Female , HEK293 Cells , Homeostasis/genetics , Homeostasis/immunology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Knockout
10.
Curr Opin Gastroenterol ; 36(6): 501-508, 2020 11.
Article in English | MEDLINE | ID: mdl-32925177

ABSTRACT

PURPOSE OF REVIEW: The gut barrier serves as the primary interface between the environment and host in terms of surface area and complexity. Luminal chemosensing is a term used to describe how small molecules in the gut lumen interact with the host through surface receptors or via transport into the subepithelial space. In this review, we have summarized recent advances in the understanding of the luminal chemosensory system in the gastroduodenal epithelium consisting of enterocytes, enteroendocrine, and tuft cells, with particular emphasis on how chemosensing affects mucosal protective responses and the metabolic syndrome. RECENT FINDINGS: Recent single-cell RNA sequencing provides detailed cell type-specific expression of chemosensory receptors and other bioactive molecules as well as cell lineages; some are similar to lingual taste cells whereas some are gut specific. Gut luminal chemosensing is not only important for the local or remote regulation of gut function, but also contributes to the systemic regulation of metabolism, energy balance, and food intake. We will discuss the chemosensory mechanisms of the proximal intestine, in particular to gastric acid, with a focus on the cell types and receptors involved in chemosensing, with emphasis on the rare chemosensory cells termed tuft cells. We will also discuss the chemosensory functions of intestinal ectoenzymes and bacterial components (e.g., lipopolysaccharide) as well as how they affect mucosal function through altering the gut-hormonal-neural axis. SUMMARY: Recent updates in luminal chemosensing by different chemosensory cells have provided new possibilities for identifying novel molecular targets for the treatment of mucosal injury, metabolic disorders, and abnormal visceral sensation.


Subject(s)
Enterocytes , Receptors, G-Protein-Coupled , Duodenum , Enteroendocrine Cells , Humans , Intestinal Mucosa , Taste
11.
Am J Physiol Gastrointest Liver Physiol ; 318(6): G1070-G1087, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32390462

ABSTRACT

Lipopolysaccharides (LPS) are potent pro-inflammatory molecules that enter the systemic circulation from the intestinal lumen by uncertain mechanisms. We investigated these mechanisms and the effect of exogenous glucagon-like peptide-2 (GLP-2) on LPS transport in the rodent small intestine. Transmucosal LPS transport was measured in Ussing-chambered rat jejunal mucosa. In anesthetized rats, the appearance of fluorescein isothiocyanate (FITC)-LPS into the portal vein (PV) and the mesenteric lymph was simultaneously monitored after intraduodenal perfusion of FITC-LPS with oleic acid and taurocholate (OA/TCA). In vitro, luminally applied LPS rapidly appeared in the serosal solution only with luminal OA/TCA present, inhibited by the lipid raft inhibitor methyl-ß-cyclodextrin (MßCD) and the CD36 inhibitor sulfosuccinimidyl oleate (SSO), or by serosal GLP-2. In vivo, perfusion of FITC-LPS with OA/TCA rapidly increased FITC-LPS appearance into the PV, followed by a gradual increase of FITC-LPS into the lymph. Rapid PV transport was inhibited by the addition of MßCD or by SSO, whereas transport into the lymph was inhibited by chylomicron synthesis inhibition. Intraveous injection of the stable GLP-2 analog teduglutide acutely inhibited FITC-LPS transport into the PV, yet accelerated FITC-LPS transport into the lymph via Nω-nitro-l-arginine methyl ester (l-NAME)- and PG97-269-sensitive mechanisms. In vivo confocal microscopy in mouse jejunum confirmed intracellular FITC-LPS uptake with no evidence of paracellular localization. This is the first direct demonstration in vivo that luminal LPS may cross the small intestinal barrier physiologically during fat absorption via lipid raft- and CD36-mediated mechanisms, followed by predominant transport into the PV, and that teduglutide inhibits LPS uptake into the PV in vivo.NEW & NOTEWORTHY We report direct in vivo confirmation of transcellular lipopolysaccharides (LPS) uptake from the intestine into the portal vein (PV) involving CD36 and lipid rafts, with minor uptake via the canonical chylomicron pathway. The gut hormone glucagon-like peptide-2 (GLP-2) inhibited uptake into the PV. These data suggest that the bulk of LPS absorption is via the PV to the liver, helping clarify the mechanism of LPS transport into the PV as part of the "gut-liver" axis. These data do not support the paracellular transport of LPS, which has been implicated in the pathogenesis of the "leaky gut" syndrome.


Subject(s)
Fats/metabolism , Intestine, Small/metabolism , Lipopolysaccharides/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Gastrointestinal Agents/pharmacology , HEK293 Cells , Humans , Intestine, Small/drug effects , Male , Mice , Mice, Inbred C57BL , Peptides/chemistry , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
Dig Dis Sci ; 65(9): 2580-2594, 2020 09.
Article in English | MEDLINE | ID: mdl-32140944

ABSTRACT

BACKGROUND/AIMS: We examined the effects of proton pump inhibitors (PPIs) on gastric antral ulcers induced by non-steroidal anti-inflammatory drugs in re-fed mice and the role of capsaicin-sensitive afferent nerves (CSANs) in the protective effects of PPIs on the antral mucosa. METHODS: Male mice were administered indomethacin after 2 h of re-feeding of diet after a 24-h fast, and gastric lesions were examined 24 h after indomethacin dosing. The effects of PPIs (lansoprazole and omeprazole), histamine H2-receptor antagonists (H2-RAs, famotidine, ranitidine), capsaicin and misoprostol on the formation of antral ulcers induced by indomethacin were examined. Functional ablation of CSANs was caused by pretreatment of mice with a high dose of capsaicin. RESULTS: Indomethacin produced lesions selectively in the gastric antrum in re-fed conditions. Formation of antral ulcers was not affected by H2-RAs, but inhibited by PPIs, capsaicin and misoprostol. The anti-ulcer effect of lansoprazole was 30 times stronger than that of omeprazole. Antral ulcers induced by indomethacin were markedly aggravated in mice with ablated CSANs. The effects of PPIs and capsaicin on ulcer formation were inhibited by ablation of CSANs, pretreatment with a capsaicin receptor antagonist (capsazepine/ruthenium red) and an inhibitor of nitric oxide synthesis (L-NAME). However, the inhibitory effect of misoprostol was not prevented by the ablation of CSANs or drugs. CONCLUSIONS: The results suggested that CSANs play an important role in protection of the antral mucosa and that both lansoprazole and omeprazole are capable of preventing NSAID-induced antral ulcers by activating CSANs.


Subject(s)
Capsaicin/pharmacology , Gastric Mucosa/innervation , Lansoprazole/pharmacology , Neurons, Afferent/drug effects , Omeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Pyloric Antrum/innervation , Stomach Ulcer/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal , Disease Models, Animal , Gastric Emptying/drug effects , Gastric Juice/metabolism , Gastric Mucosa/pathology , Histamine H2 Antagonists/pharmacology , Indomethacin , Male , Mice , Neurons, Afferent/pathology , Pyloric Antrum/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Stomach Ulcer/physiopathology
13.
Dig Dis Sci ; 65(9): 2605-2618, 2020 09.
Article in English | MEDLINE | ID: mdl-32006214

ABSTRACT

BACKGROUND: Circulating endotoxin (lipopolysaccharide, LPS) increases the gut paracellular permeability. We hypothesized that glucagon-like peptide-2 (GLP-2) acutely reduces LPS-related increased intestinal paracellular permeability by a mechanism unrelated to its intestinotrophic effect. METHODS: We assessed small intestinal paracellular permeability in vivo by measuring the appearance of intraduodenally perfused FITC-dextran 4000 (FD4) into the portal vein (PV) in rats 1-24 h after LPS treatment (5 mg/kg, ip). We also examined the effect of a stable GLP-2 analog teduglutide (TDG) on FD4 permeability. RESULTS: FD4 movement into the PV was increased 6 h, but not 1 or 3 h after LPS treatment, with increased PV GLP-2 levels and increased mRNA expressions of proinflammatory cytokines and proglucagon in the ileal mucosa. Co-treatment with a GLP-2 receptor antagonist enhanced PV FD4 concentrations. PV FD4 concentrations 24 h after LPS were higher than FD4 concentrations 6 h after LPS, reduced by exogenous GLP-2 treatment given 6 or 12 h after LPS treatment. FD4 uptake measured 6 h after LPS was reduced by TDG 3 or 6 h after LPS treatment. TDG-associated reduced FD4 uptake was reversed by the VPAC1 antagonist PG97-269 or L-NAME, not by EGF or IGF1 receptor inhibitors. CONCLUSIONS: Systemic LPS releases endogenous GLP-2, reducing LPS-related increased permeability. The therapeutic window of exogenous GLP-2 administration is at minimum within 6-12 h after LPS treatment. Exogenous GLP-2 treatment is of value in the prevention of increased paracellular permeability associated with endotoxemia.


Subject(s)
Endotoxemia/prevention & control , Glucagon-Like Peptide 2/metabolism , Glucagon-Like Peptide-2 Receptor/agonists , Intestinal Absorption/drug effects , Intestine, Small/drug effects , Peptides/pharmacology , Animals , Dextrans/blood , Disease Models, Animal , Endotoxemia/blood , Endotoxemia/chemically induced , Fluorescein-5-isothiocyanate/analogs & derivatives , Glucagon-Like Peptide-2 Receptor/metabolism , Inflammation Mediators/metabolism , Intestine, Small/metabolism , Lipopolysaccharides , Male , Permeability , Portal Vein , Rats, Sprague-Dawley , Time Factors
14.
Dig Dis Sci ; 65(2): 669, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31823102

ABSTRACT

The original version of the article unfortunately contained an error in one of the sentences. The sentence, "Eventually, Dan Drucker in Pat Brubaker's laboratory convincingly demonstrated that GLP-2 is the proglucagon product controlling intestinal proliferation [19]," should read "Eventually, Dan Drucker convincingly demonstrated that GLP-2 is the proglucagon product controlling intestinal proliferation [19]."

15.
Dig Dis Sci ; 65(1): 119-131, 2020 01.
Article in English | MEDLINE | ID: mdl-31515722

ABSTRACT

BACKGROUND: Peptic ulcers recur, suggesting that ulcer healing may leave tissue predisposed to subsequent damage. In mice, we have identified that the regenerated epithelium found after ulcer healing will remain abnormal for months after healing. AIM: To determine whether healed gastric mucosa has altered epithelial function, as measured by electrophysiologic parameters. METHOD: Ulcers were induced in mouse gastric corpus by serosal local application of acetic acid. Thirty days or 8 months after ulcer induction, tissue was mounted in an Ussing chamber. Transepithelial electrophysiologic parameters (short-circuit current, Isc. resistance, R) were compared between the regenerated healed ulcer region and the non-ulcerated contralateral region, in response to luminal hyperosmolar NaCl challenge (0.5 M). RESULTS: In unperturbed stomach, luminal application of hyperosmolar NaCl transiently dropped Isc followed by gradual recovery over 2 h. Compared to the starting baseline Isc, percent Isc recovery was reduced in 30-day healing mucosa, but not at 8 months. Prior to NaCl challenge, a lower baseline Isc was observed in trefoil factor 2 (TFF2) knockout (KO) versus wild type (WT), with no Isc recovery in either non-ulcerated or healing mucosa of KO. Inhibiting Na/H exchanger (NHE) transport in WT mucosa inhibited Isc recovery in response to luminal challenge. NHE2-KO baseline Isc was reduced versus NHE2-WT. In murine gastric organoids, NHE inhibition slowed recovery of intracellular pH and delayed the repair of photic induced damage. CONCLUSION: Healing gastric mucosa has deficient electrophysiological recovery in response to hypertonic NaCl. TFF2 and NHE2 contribute to Isc regulation, and the recovery and healing of transepithelial function.


Subject(s)
Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Sodium Chloride/metabolism , Sodium-Hydrogen Exchangers/deficiency , Stomach Ulcer/metabolism , Wound Healing , Acetic Acid , Animals , Disease Models, Animal , Electric Impedance , Epithelial Cells/pathology , Female , Gastric Mucosa/pathology , Hydrogen-Ion Concentration , Hypertonic Solutions , Male , Mice, Inbred C57BL , Mice, Knockout , Re-Epithelialization , Sodium-Hydrogen Exchangers/genetics , Stomach Ulcer/chemically induced , Stomach Ulcer/genetics , Stomach Ulcer/pathology , Time Factors , Trefoil Factor-2/deficiency , Trefoil Factor-2/genetics
16.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31559013

ABSTRACT

Vasoactive intestinal peptide (VIP), a gut peptide hormone originally reported as a vasodilator in 1970, has multiple physiological and pathological effects on development, growth, and the control of neuronal, epithelial, and endocrine cell functions that in turn regulate ion secretion, nutrient absorption, gut motility, glycemic control, carcinogenesis, immune responses, and circadian rhythms. Genetic ablation of this peptide and its receptors in mice also provides new insights into the contribution of VIP towards physiological signaling and the pathogenesis of related diseases. Here, we discuss the impact of VIP on gastrointestinal function and diseases based on recent findings, also providing insight into its possible therapeutic application to diabetes, autoimmune diseases and cancer.


Subject(s)
Gastrointestinal Diseases , Gastrointestinal Tract , Vasoactive Intestinal Peptide , Animals , Gastrointestinal Diseases/metabolism , Gastrointestinal Tract/physiology , Mice , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Signal Transduction , Vasoactive Intestinal Peptide/physiology
18.
Curr Gastroenterol Rep ; 21(8): 35, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31289927

ABSTRACT

PURPOSE OF REVIEW: Short-chain fatty acids (SCFAs), the main bacterial fermentation products in the hindgut of hindgut fermenters, are also present in the foregut lumen. We discuss the impact of SCFAs in the duodenal defense mechanisms and in the gastrointestinal (GI) pathogenesis. RECENT FINDINGS: Luminal SCFAs augment the duodenal mucosal defenses via release of serotonin (5-HT) and glucagon-like peptide-2 (GLP-2) from enteroendocrine cells. Released GLP-2 protects the small intestinal mucosa from nonsteroidal anti-inflammatory drug-induced enteropathy. SCFAs are also rapidly absorbed via SCFA transporters and interact with afferent and myenteric nerves. Excessive SCFA signals with 5-HT3 receptor overactivation may be implicated in the pathogenesis of irritable bowel syndrome symptoms. SCFA production exhibits diurnal rhythms with host physiological responses, suggesting that oral SCFA treatment may adjust the GI clocks. SCFAs are not only a source of energy but also signaling molecules for the local regulation of the GI tract and systemic regulation via release of gut hormones. Targeting SCFA signals may be a novel therapeutic for GI diseases and metabolic syndrome.


Subject(s)
Duodenum/metabolism , Fatty Acids, Volatile/physiology , Gastrointestinal Diseases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Circadian Rhythm/physiology , Duodenum/microbiology , Gastrointestinal Diseases/chemically induced , Gastrointestinal Microbiome/physiology , Humans , Intestinal Mucosa/metabolism
19.
J Infect Dis ; 220(11): 1719-1728, 2019 10 22.
Article in English | MEDLINE | ID: mdl-30624615

ABSTRACT

BACKGROUND: Cholera toxin (CT)-induced diarrhea is mediated by cyclic adenosine monophosphate (cAMP)-mediated active Cl- secretion via the cystic fibrosis transmembrane conductance regulator (CFTR). Although the constitutive activation of adenylyl cyclase (AC) in response to CT is due to adenosine diphosphate ribosylation of the small G protein α-subunit activating CFTR with consequent secretory diarrhea, the AC isoform(s) involved remain unknown. METHODS: We generated intestinal epithelial cell-specific adenylyl cyclase 6 (AC6) knockout mice to study its role in CT-induced diarrhea. RESULTS: AC6 messenger RNA levels were the highest of all 9 membrane-bound AC isoforms in mouse intestinal epithelial cells. Intestinal epithelial-specific AC6 knockout mice (AC6loxloxVillinCre) had undetectable AC6 levels in small intestinal and colonic epithelial cells. No significant differences in fluid and food intake, plasma electrolytes, intestinal/colon anatomy and morphology, or fecal water content were observed between genotypes. Nevertheless, CT-induced fluid accumulation in vivo was completely absent in AC6loxloxVillinCre mice, associated with a lack of forskolin- and CT-induced changes in the short-circuit current (ISC) of the intestinal mucosa, impaired cAMP generation in acutely isolated small intestinal epithelial cells, and significantly impaired apical CFTR levels in response to forskolin. CONCLUSIONS: AC6 is a novel target for the treatment of CT-induced diarrhea.


Subject(s)
Adenylyl Cyclases/metabolism , Cholera Toxin/toxicity , Cholera/physiopathology , Diarrhea/physiopathology , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Adenylyl Cyclases/deficiency , Animals , Colforsin/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout
20.
Curr Opin Gastroenterol ; 34(6): 422-427, 2018 11.
Article in English | MEDLINE | ID: mdl-30124472

ABSTRACT

PURPOSE OF REVIEW: Luminal chemosensing is a term used to describe how small molecules in the gut lumen interact with the host through surface receptors or via transport into the submucosa. In this review, we have summarized recent advances of understanding luminal chemosensing in the gastroduodenal mucosa, with a particular emphasis on how chemosensing affects mucosal protective responses and the metabolic syndrome. RECENT FINDINGS: In the past decade, data have supported the hypothesis that gut luminal chemosensing not only is important for the local or remote regulation of gut function but also contributes to the systemic regulation of metabolism, energy balance and food intake. We have provided examples of how luminal nutrients such as long-chain fatty acids (LCFAs), endogenous compounds such as bile acids, bacterial metabolites such as short-chain fatty acids (SCFAs) and bacterial components such as lipopolysaccharide (LPS) activate cognate receptors expressed on key effector cells such as enteroendocrine cells and inflammatory cells in order to profoundly affect organ function through the initiation or suppression of inflammatory pathways, altering gut barrier function and nutrient uptake, altering gut motility and visceral pain pathways, and preventing mucosal injury. SUMMARY: These recent discoveries in this area have provided new possibilities for identifying novel molecular targets for the treatment of mucosal injury, metabolic disorders and abnormal visceral sensation. Understanding luminal chemosensory mechanisms may help to identify novel molecular targets for the treatment and prevention of mucosal injury, metabolic disorders and abnormal visceral sensation.


Subject(s)
Chemoreceptor Cells/physiology , Duodenum/innervation , Alkaline Phosphatase/physiology , Duodenum/metabolism , Fatty Acids/metabolism , GPI-Linked Proteins/physiology , Humans , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Lipopolysaccharides/metabolism , Nutrients/metabolism , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL