Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
2.
Nutrients ; 14(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35057580

ABSTRACT

The objective of the present research was to review the state of the art on the consequences of drinking coffee at the different levels of the gastrointestinal tract. At some steps of the digestive process, the effects of coffee consumption seem rather clear. This is the case for the stimulation of gastric acid secretion, the stimulation of biliary and pancreatic secretion, the reduction of gallstone risk, the stimulation of colic motility, and changes in the composition of gut microbiota. Other aspects are still controversial, such as the possibility for coffee to affect gastro-esophageal reflux, peptic ulcers, and intestinal inflammatory diseases. This review also includes a brief summary on the lack of association between coffee consumption and cancer of the different digestive organs, and points to the powerful protective effect of coffee against the risk of hepatocellular carcinoma. This review reports the available evidence on different topics and identifies the areas that would most benefit from additional studies.


Subject(s)
Coffee , Gastrointestinal Tract , Bile/physiology , Caffeine/administration & dosage , Coffee/adverse effects , Female , Gallstones/prevention & control , Gastric Acid/physiology , Gastroesophageal Reflux , Gastrointestinal Microbiome , Gastrointestinal Motility , Gastrointestinal Neoplasms , Gastrointestinal Tract/drug effects , Humans , Inflammatory Bowel Diseases , Male , Pancreas/physiology , Peptic Ulcer , Saliva/enzymology
3.
Clin Drug Investig ; 40(10): 897-899, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32779119

ABSTRACT

During the ongoing pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more attention should be paid to the balance of risks and benefits associated with proton pump inhibitors for the following reasons. One of the main functions of gastric juice is to inactivate swallowed microorganisms, thereby inhibiting infectious agents from reaching the intestine. Studies have documented that proton pump inhibitors are a risk factor for rotavirus, influenza virus, norovirus, and Middle East respiratory syndrome coronavirus infections, and are associated with an increased risk of acute gastroenteritis during periods of highest circulation of enteric viruses. In light of the evidence for gastrointestinal infection implying a fecal-oral transmission of SARS-CoV-2 and given the magnitude of the SARS-CoV-2/coronavirus disease 2019 pandemic, associated with the widespread misuse of proton pump inhibitors, this suggests that we should not rule out the hypothesis that patients treated with proton pump inhibitors may be more at risk of being infected by SARS-CoV-2.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , Proton Pump Inhibitors/adverse effects , Betacoronavirus/physiology , COVID-19 , Coronavirus Infections/chemically induced , Gastric Acid/physiology , Humans , Middle East Respiratory Syndrome Coronavirus/drug effects , Middle East Respiratory Syndrome Coronavirus/physiology , Pandemics , Pneumonia, Viral/chemically induced , Risk Factors , SARS-CoV-2
4.
Am J Perinatol ; 37(4): 415-420, 2020 03.
Article in English | MEDLINE | ID: mdl-30780184

ABSTRACT

OBJECTIVE: The use of acid suppression therapies in newborns lacks efficacy and is associated with adverse effects. Point-of-care (POC) assessment of gastric aspirate pH may provide an objective, noninvasive measure of gastric acidity in tube fed infants. We conducted the present study to characterize the POC gastric pH levels in gastric tube fed infants before and after initiation of enteral omeprazole or ranitidine. STUDY DESIGN: Retrospective cohort study of infants with gastric aspirate pH levels determined by POC pH strips. Gastric pH levels recorded during 7 days before and 14 days after medication initiation were compared using Wilcoxon's sign-rank tests. RESULTS: Among 307 evaluated infants, 284 (92%) had a median gastric pH level ≥4 in 7 days prior to ranitidine or omeprazole. In 14 days after medication initiation, the median gastric pH of infants with pretreatment median gastric pH < 4 increased to 4.5 and 5 (p < 0.01) in the ranitidine and omeprazole groups, respectively. There was no change in infants with pretreatment median gastric pH ≥4. CONCLUSION: Among infants receiving gastric tube feedings and enteral omeprazole or ranitidine, only those with a pretreatment gastric pH level <4 demonstrated a significant increase in gastric pH. Validation of our findings against esophageal pH multichannel intraluminal impedance testing is needed.


Subject(s)
Anti-Ulcer Agents/pharmacology , Enteral Nutrition , Gastric Acidity Determination , Hydrogen-Ion Concentration/drug effects , Omeprazole/pharmacology , Point-of-Care Testing , Ranitidine/pharmacology , Anti-Ulcer Agents/therapeutic use , Critical Care , Female , Gastric Acid/physiology , Humans , Infant, Newborn , Infant, Premature , Intensive Care Units, Neonatal , Intubation, Gastrointestinal , Male , Omeprazole/therapeutic use , Ranitidine/therapeutic use , Retrospective Studies
5.
Med Sci Monit ; 25: 6153-6164, 2019 Aug 16.
Article in English | MEDLINE | ID: mdl-31419218

ABSTRACT

BACKGROUND The clinical association between gastroesophageal reflux disease (GERD) and idiopathic pulmonary fibrosis (IPF) has been known for many years, but it is still unclear. The present study investigated the association between experimentally simulated aspiration and pulmonary fibrosis. MATERIAL AND METHODS A total of 120 male Sprague-Dawley rats were randomly divided into a negative control group, a bleomycin group, and 3 simulated aspiration groups. The bleomycin group was administered a one-time intratracheal injection of bleomycin, whereas the 3 simulated aspiration groups were treated either with an intratracheal instillation of gastric fluid combined with pepsin, with pepsin alone, or with hydrochloric acid, all twice a week, and the negative control group was administered normal saline twice a week. Lung tissues were collected to evaluate pathological changes and the mRNA expression levels of connective tissue growth factor (CTGF), type I collagen, and transforming growth factor. RESULTS The results demonstrated that the degree of fibrosis in the early stage was low in each of the 3 simulated aspiration groups, but gradually increased over time. The expression levels of the downstream factor of fibrosis, CTGF, and type I collagen also reflected this trend. CONCLUSIONS The study demonstrates that aspiration of gastric contents can cause pulmonary fibrosis, and mixed aspiration of pepsin and gastric fluid can accelerate this process. This study provides strong evidence in support of a potential association between human GERD and IPF.


Subject(s)
Gastric Acid/metabolism , Pepsin A/metabolism , Pulmonary Fibrosis/metabolism , Administration, Inhalation , Animals , Bleomycin/pharmacology , Cysteine-Rich Protein 61/genetics , Gastric Acid/physiology , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/physiopathology , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/metabolism , Lung/pathology , Male , Pepsin A/physiology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/physiopathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factors/genetics
6.
Chest ; 154(4): 963-971, 2018 10.
Article in English | MEDLINE | ID: mdl-29859888

ABSTRACT

Nocturnal gastroesophageal reflux has been associated with poor sleep quality. Normal physiological adaptations of the aerodigestive system to sleep prolong and intensify nocturnal reflux events. This occurrence leads to sleep disruption, as well as to esophageal, laryngeal, and laryngopharyngeal reflux. Controversy exists on whether OSA and nocturnal reflux are causally linked or merely associated because of shared risk factors. Advances in diagnostic technology have provided new insights into gastroesophageal reflux and the mechanisms of nocturnal reflux during sleep. This update reviews new data on causal links between sleep and gastroesophageal reflux disease.


Subject(s)
Gastroesophageal Reflux/complications , Sleep Apnea, Obstructive/complications , Esophageal Motility Disorders/physiopathology , Esophageal Sphincter, Lower/physiology , Esophageal Sphincter, Upper/physiology , Gastric Acid/physiology , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/physiopathology , Humans , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/physiopathology
7.
Nat Commun ; 8(1): 837, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038503

ABSTRACT

Chronic liver disease is rising in western countries and liver cirrhosis is the 12th leading cause of death worldwide. Simultaneously, use of gastric acid suppressive medications is increasing. Here, we show that proton pump inhibitors promote progression of alcoholic liver disease, non-alcoholic fatty liver disease, and non-alcoholic steatohepatitis in mice by increasing numbers of intestinal Enterococcus spp. Translocating enterococci lead to hepatic inflammation and hepatocyte death. Expansion of intestinal Enterococcus faecalis is sufficient to exacerbate ethanol-induced liver disease in mice. Proton pump inhibitor use increases the risk of developing alcoholic liver disease among alcohol-dependent patients. Reduction of gastric acid secretion therefore appears to promote overgrowth of intestinal Enterococcus, which promotes liver disease, based on data from mouse models and humans. Recent increases in the use of gastric acid-suppressive medications might contribute to the increasing incidence of chronic liver disease.Proton pump inhibitors (PPIs) reduce gastric acid secretion and modulate gut microbiota composition. Here Llorente et al. show that PPIs induce bacterial overgrowth of enterococci, which, in turn, exacerbate ethanol-induced liver disease both in mice and humans.


Subject(s)
Gastric Acid/physiology , Liver Diseases, Alcoholic/pathology , Microbiota/drug effects , Non-alcoholic Fatty Liver Disease/pathology , Proton Pump Inhibitors/adverse effects , Animals , Disease Progression , Enterococcus/drug effects , Enterococcus/growth & development , Female , Humans , Incidence , Liver Diseases, Alcoholic/epidemiology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/epidemiology
8.
Curr Opin Gastroenterol ; 33(6): 430-438, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28787289

ABSTRACT

PURPOSE OF REVIEW: The present review summarizes the past year's literature, both clinical and basic science, regarding physiologic and pharmacologic regulation of gastric acid secretion in health and disease. RECENT FINDINGS: Gastric acid kills microorganisms, assists digestion, and facilitates absorption of iron, calcium, and vitamin B12. The main stimulants of acid secretion are the hormone gastrin, released from antral G cells; paracrine agent histamine, released from oxyntic enterochromaffin-like cells; and neuropeptide acetylcholine, released from antral and oxyntic intramural neurons. Gastrin is also a trophic hormone that participates in carcinogenesis. Helicobacter pylori may increase or decrease acid secretion depending upon the acuity and predominant anatomic focus of infection; most patients manifest hypochlorhydria. Despite the fact that proton pump inhibitors (PPIs) are amongst the most widely prescribed drugs, they are underutilized in patients at high risk for UGI bleeding. Although generally considered well tolerated, concerns have been raised regarding associations between PPI use and dementia, kidney disease, myocardial infarction, pneumonia, osteoporosis, dysbiosis, small bowel injury, micronutrient deficiency, and fundic gland polyps. SUMMARY: Our understanding of the physiologic, pathophysiologic, and pharmacologic regulation of gastric secretion continues to advance. Such knowledge is crucial for improved and safe management of acid-peptic disorders.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Acetylcholine/physiology , Gastric Acid/physiology , Gastrins/physiology , Helicobacter Infections/metabolism , Helicobacter pylori , Histamine/physiology , Humans , Proton Pump Inhibitors/pharmacology
9.
Caries Res ; 50(2): 117-23, 2016.
Article in English | MEDLINE | ID: mdl-26981853

ABSTRACT

Studies of wine tasters and patients with self-induced vomiting have revealed that 30-50% of individuals at high risk do not develop erosive lesions. The aim was to investigate this apparent individual susceptibility to enamel erosion. Two enamel specimens were made from each of 3 premolars from 8 persons (donors). Six acrylic mouth appliances were worn by 6 volunteers (carriers). One specimen from each donor was mounted on each appliance. The carriers wore the appliances for 9 days. The appliances were immersed in 0.01 M HCl for 3 min twice per day to imitate a vomiting/reflux situation. The enamel specimens were analysed by a white-light interferometer to measure enamel loss (in micrometres). The enamel loss varied significantly both between the donor teeth (p = 0.009) and the carriers (p = 0.004). The lesion in the specimen with the largest amount of enamel loss was 4 times as deep as in the specimen with the lowest. In 1 carrier, all specimens displayed enamel loss above the mean, including the specimen from the donor with the most resistant enamel. The variation in susceptibility to erosion among individuals appears to be influenced both by the sustainability of the enamel and by factors in the oral environment. This could explain the variation in prevalence and severity of dental erosions among patients exposed to similar acidic challenges. The results suggest that for certain individuals, only minimal acidic challenges may be sufficient to cause damage to the teeth, while others may never develop dental erosions despite extensive exposure to acid.


Subject(s)
Dental Caries Susceptibility , Dental Enamel , Tooth Erosion , Acids/adverse effects , Adult , Dental Enamel/drug effects , Dental Enamel/pathology , Dental Pellicle/physiology , Disease Susceptibility , Female , Fluorides/therapeutic use , Gastric Acid/physiology , Humans , Hydrogen-Ion Concentration , Interferometry , Saliva/metabolism , Time Factors , Tooth Erosion/chemically induced
10.
Dis Esophagus ; 29(8): 1054-1063, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26541138

ABSTRACT

Postprandial gastroesophageal reflux (PGER) in the distal esophagus (DE) is associated with a gastric juice 'acid pocket' (AP). Baclofen reduces AP extension into the DE in healthy volunteers, in part through increased lower esophageal sphincter (LES) pressure. We aimed to verify whether baclofen also affects postprandial AP location and extent in gastroesophageal reflux disease (GERD) patients. Thirteen treatment-naive heartburn-prevalent GERD patients underwent two AP studies, after pretreatment with baclofen 40 mg or placebo 30 minutes preprandially. We performed pH-probe stepwise pull-throughs (PT) (1 cm/min, LES -10 to +5 cm) before and every 30 minutes from 30 minutes before up to 150 minutes after a test meal. After the meal, both after placebo and baclofen, gastric pH significantly dropped at 30, 60, 90 minutes postprandially (P: nadir pHs of 3.9 ± 0.6, 2.3 ± 0.6, 2.1 ± 0.4; B: nadir pHs of 2.5 ± 0.4, 2.8 ± 0.4, 2.5 ± 0.3; all P < 0.05). After placebo, LES pressure decreased at 60, 90 and 120 minutes postprandially (32.7 ± 6.1 vs. 24.5 ± 3.1, 27.3 ± 5.9, 27.3 ± 6.0 mmHg; analysis of variance [ANOVA], P = 0.037), but this was prevented by baclofen (25.4 ± 3.4 vs. 29.4 ± 2, 32.2 ± 1.4, 35.5 ± 1.7 mmHg, ANOVA, P = not significant (NS)). Baclofen did not significantly decrease the postprandial AP extent above the LES but prevented the postprandial increase in transient lower esophageal sphincter relaxations (TLESRs) (preprandial vs. postprandial, placebo: 1.1 ± 0.3 vs. 3.7 ± 0.7, P < 0.05; baclofen: 1.4 ± 0.4 vs. 2 ± 0.5, P = NS). In GERD patients, baclofen significantly increases postprandial LES pressure, prevents the increase TLESRs but, unlike in healthy volunteers, does not affect AP extension into the DE.


Subject(s)
Baclofen/therapeutic use , Gastroesophageal Reflux/drug therapy , Heartburn/drug therapy , Muscle Relaxants, Central/therapeutic use , Adult , Double-Blind Method , Drug Administration Schedule , Esophageal Sphincter, Lower/drug effects , Esophageal Sphincter, Lower/physiopathology , Esophagogastric Junction/drug effects , Esophagogastric Junction/physiopathology , Female , Gastric Acid/physiology , Gastroesophageal Reflux/physiopathology , Heartburn/physiopathology , Humans , Hydrogen-Ion Concentration , Male , Manometry , Middle Aged , Postprandial Period/drug effects , Pressure , Time Factors , Young Adult
11.
J Physiol Sci ; 65(6): 545-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26319795

ABSTRACT

It is well known that hydrogen sulfide (H2S) protects the gastric mucosa against gastric acid and other noxious stimulants by several mechanisms but until now the effect of gastric acid on H2S production has not been evaluated. This study was performed to determine the effect of basal and stimulated gastric acid secretion on mRNA and protein expression of cystathionine gamma lyase (CSE) and cystathionine beta synthase (CBS), and on mucosal release of H2S in rats. Seventy-two male rats were randomly assigned into 9 groups (8 in each)-control, distention, and pentagastrin-induced gastric acid secretion groups. The effects of 15% alcohol solution, propargylglycine (PAG), L-NAME, and pantoprazole were also investigated. Under anesthesia, animals underwent tracheostomy and midline laparotomy. A catheter was inserted into the stomach through the duodenum for gastric washout. At the end of the experiments, the animals were killed and the gastric mucosa was collected to measure H2S concentration and to quantify mRNA expression of CSE and CBS by quantitative real-time PCR, and expression of their proteins by western blot. Basal and stimulated gastric acid secretion increased mucosal levels of H2S, and mRNA and protein expression of CSE. Pantoprazole and L-NAME reversed H2S release and restored protein expression of CSE to the control level. Pantoprazole, but not propargylglycine, pretreatment inhibited the elevated level of protein expression of eNOS in response to distention-induced gastric acid secretion. Our findings indicated that NO mediated the stimulatory effect of gastric acid on H2S release and protein expression of CSE.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Gastric Acid/physiology , Gene Expression Regulation/physiology , Hydrogen Sulfide/metabolism , RNA, Messenger/metabolism , Up-Regulation/physiology , 2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Alcohols/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/genetics , Gastric Mucosa/metabolism , Gene Expression Regulation, Enzymologic/physiology , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Pantoprazole , Pentagastrin/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
12.
PLoS One ; 10(7): e0134116, 2015.
Article in English | MEDLINE | ID: mdl-26222383

ABSTRACT

Gastric acidity is likely a key factor shaping the diversity and composition of microbial communities found in the vertebrate gut. We conducted a systematic review to test the hypothesis that a key role of the vertebrate stomach is to maintain the gut microbial community by filtering out novel microbial taxa before they pass into the intestines. We propose that species feeding either on carrion or on organisms that are close phylogenetic relatives should require the most restrictive filter (measured as high stomach acidity) as protection from foreign microbes. Conversely, species feeding on a lower trophic level or on food that is distantly related to them (e.g. herbivores) should require the least restrictive filter, as the risk of pathogen exposure is lower. Comparisons of stomach acidity across trophic groups in mammal and bird taxa show that scavengers and carnivores have significantly higher stomach acidities compared to herbivores or carnivores feeding on phylogenetically distant prey such as insects or fish. In addition, we find when stomach acidity varies within species either naturally (with age) or in treatments such as bariatric surgery, the effects on gut bacterial pathogens and communities are in line with our hypothesis that the stomach acts as an ecological filter. Together these results highlight the importance of including measurements of gastric pH when investigating gut microbial dynamics within and across species.


Subject(s)
Biological Evolution , Gastric Acid/physiology , Gastrointestinal Microbiome , Animals , Birds , Food Microbiology , Herbivory , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Mammals , Phylogeny , Stomach/microbiology , Stomach/physiology , Symbiosis
13.
J Dig Dis ; 16(3): 109-17, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25624012

ABSTRACT

The discovery of Helicobacter pylori (H. pylori) changed the dogma of the stomach as a sterile organ. H. pylori is an obligate pathogen in the human stomach and recognized as a definite carcinogen. Extensive research on the interaction of this bacterium with the gastric mucosa has been performed over the past three decades. The development of new nucleotide sequencing techniques and new biocomputational tools has opened the field for studying the diversity and complexity of the microbiome in the gastrointestinal tract independently of cultural methods. These techniques allow to better characterize further gastric bacteria. However, the differentiation of alive resident and transient microbes requires an analysis beyond the pure detection of bacterial genomic material applying a combination with metabolomic analyses. Currently, the interaction of gastric microbiota with each other, with H. pylori and with the host is addressed by extensive research. This review gives a concise overview on current knowledge on this topic.


Subject(s)
Gastrointestinal Microbiome/physiology , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Bacteria/isolation & purification , Biomedical Research/methods , Gastric Acid/physiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Tract/microbiology , Helicobacter Infections/drug therapy , Host-Pathogen Interactions , Humans , Probiotics/therapeutic use
14.
J Pharm Pharmacol ; 67(1): 133-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25213103

ABSTRACT

OBJECTIVES: The aim of this study was to extend our knowledge about the mechanism involved in the gastroprotective effect of P1G10, a proteolytic fraction rich in cysteine proteinases from Vasconcellea cundinamarcensis (syn. Carica candamarcensis) latex, which demonstrated gastric healing and protection activities in rats. METHODS: Wistar rats were submitted to gastric lesions by indomethacin and treated with P1G10 (10 mg/kg). Free thiol groups and prostaglandin E2 content were measured in gastric mucosal and gastrin levels in blood samples. To evaluate the participation of nitric oxide (NO) or proteolytic activity of P1G10 on its gastroprotective effect, animals were treated with an inhibitor of NO production (L-NAME) or the fraction inhibited by iodoacetamide, respectively. Gastric secretion study (acidity and pepsin activity) was also performed. KEY FINDINGS: P1G10 (10 mg/kg) inhibited the occurrence of gastric lesions by indomethacin, restored the free thiol groups content on gastric mucosa and increased moderately prostaglandin E2 levels (34%). Furthermore, the treatment decreased the gastrin levels (95%), suggesting a possible modulation of secretory activity. This effect was accordant with attenuation of gastric acidity (42%) and pepsin activity (69%) seen in animals subjected to pyloric ligation. The inhibition of NO production or the proteolytic activity of P1G10 does not affect the gastroprotective effect. CONCLUSIONS: These results can explain the gastroprotective activity of P1G10 and serve a basis for further studies of this active principle.


Subject(s)
Carica , Cysteine Proteases/pharmacology , Dinoprostone/metabolism , Gastric Acid/metabolism , Plant Extracts/pharmacology , Sulfhydryl Compounds/metabolism , Animals , Female , Gastric Acid/chemistry , Gastric Acid/physiology , Gastric Mucosa , Gastrins/biosynthesis , Gastrins/blood , Indomethacin/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Rats , Rats, Wistar
16.
Dis Esophagus ; 28(6): 520-3, 2015.
Article in English | MEDLINE | ID: mdl-24898795

ABSTRACT

Multiple water swallows (MWS) stimulates neural inhibition, resulting in abolition of contractions in the esophageal body and complete lower esophageal sphincter relaxation, which is followed by peristalsis and the lower esophageal sphincter contraction. We assessed the relationship between MWS and gastroesophageal reflux in patients with esophageal symptoms and with normal findings by high-resolution manometry (HRM). We retrospectively reviewed the clinical records of patients who underwent HRM and a 24-hour ambulatory impedance-pH study. Correlation between the findings of the impedance-pH study and abnormal MWS responses without motility disorders was evaluated. Independent t-tests were used for statistical analysis. Of 28 patients, 20 (71%) had abnormal MWS responses: four (20%) had abnormal responses during MWS, six (30%) had abnormal responses after MWS, and 10 (50%) had abnormal responses both during and after MWS. Total acid exposure times were significantly longer in patients with abnormal MWS responses than in patients with normal MWS responses. In particular, upright acid exposure time and all reflux percent times were significantly longer in patients with abnormal MWS responses. However, bolus clearance time and longest reflux episode were not different between the two groups. Abnormal MWS responses predicted increased acid exposure times in patients with normal findings of HRM by the Chicago classification.


Subject(s)
Deglutition/physiology , Gastroesophageal Reflux/physiopathology , Electric Impedance , Esophageal pH Monitoring , Female , Gastric Acid/physiology , Humans , Male , Manometry/methods , Patient Positioning , Peristalsis/physiology , Retrospective Studies , Water/administration & dosage
17.
J Gastroenterol Hepatol ; 29 Suppl 4: 93-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521740

ABSTRACT

Acid-sensing pathways, which trigger mucosal defense mechanisms in response to luminal acid, involve the rapid afferent-mediated "capsaicin pathway" and the sustained "prostaglandin (PG) pathway." Luminal acid quickly increases protective PG synthesis and release from epithelia, although the mechanism by which luminal acid induces PG synthesis is still mostly unknown. Acid exposure augments purinergic ATP-P2Y signaling by inhibition of intestinal alkaline phosphatase activity. Since P2Y activation increases intracellular Ca2+, we further hypothesized that ATP-P2Y signals increase the generation of H2O2 derived from dual oxidase, a member of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family activated by Ca2+. Our recent studies suggest that acid exposure increases H2O2 output, followed by phospholipase A2 and cyclooxygenase activation, increasing PG synthesis. Released prostaglandin E2 augments protective HCO3- and mucus secretion via EP4 receptor activation. Thus, the PG pathway as a component of duodenal acid sensing consists of acid-related intestinal alkaline phosphatase inhibition, ATP-P2Y signals, dual oxidase 2-derived H2O2 production, phospholipase A2 activation, prostaglandin E2 synthesis, and EP4 receptor activation. The PG pathway is also involved in luminal bacterial sensing in the duodenum via activation of pattern recognition receptors, including Toll-like receptors and nucleotide-binding oligomerization domain 2. The presence of acute mucosal responses to luminal bacteria suggests that the duodenum is important for host defenses and may reduce bacterial loading to the hindgut using H2O2, complementing gastric acidity and anti-bacterial bile acids.


Subject(s)
Dinoprostone/metabolism , Duodenum/physiology , Gastric Acid/physiology , Intestinal Mucosa/physiology , Signal Transduction/physiology , Adenosine Triphosphate/physiology , Bacterial Adhesion , Bicarbonates/metabolism , Calcium/metabolism , Duodenum/metabolism , Humans , Hydrogen Peroxide , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , NADPH Oxidases/metabolism , Phospholipases A2/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Pattern Recognition/physiology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Purinergic P2Y/physiology
18.
World J Surg Oncol ; 12: 375, 2014 Dec 06.
Article in English | MEDLINE | ID: mdl-25479911

ABSTRACT

BACKGROUND: The nature of the relationship between Helicobacter pylori and reflux esophagitis (RE) is not fully understood. In addition, the effect of H. pylori eradication on RE and gastroesophageal reflux disease (GERD) is unclear. This study was designed to investigate the relationship between H. pylori infection and the grade of GERD in patients with reflux symptoms. METHODS: Between January 2010 and July 2013, 184 consecutive patients with daily reflux symptoms for at least one year were evaluated at the ambulatory for functional esophageal disease, Tor Vergata University Hospital, Rome, Italy. All patients underwent a pretreatment evaluation, which included anamnesis, clinical examination, Esophagogastroduodenoscopy (EGDS) with biopsy, esophageal manometry and 24-hour pH-metry. All statistical elaborations were obtained using Statigraphies 5 plus for Window XP. RESULTS: There was no statistical difference regarding Lower Esophageal Sphincter (LES) pressure between patients who were H. pylori-positive and H. Pylori-negative (19.2 ± 9.5 (range: 3.7 to 46.2) and 19.7 ± 11.0 (range: 2.6 to 61), respectively). Further, no significant difference was evidenced in esophageal wave length (mean value: 3.1 seconds in H. pylori-negative patients versus 3.2 seconds in H. pylori-positive patients) or in esophageal wave height (mean value: 72.2 ± 39.3 in H. pylori-negative patients versus 67.7 ± 28.4 in H. pylori-positive patients). We observed that hiatal hernia (P = 0.01), LES opening (P = 0.05), esophageal wave length (P = 0.01) and pathological reflux number (P = 0.05) were significantly related to the presence of esophagitis. However, H. pylori infection was not significantly related to the presence of reflux esophagitis. CONCLUSIONS: Our clinical, endoscopic, manometric and pH-metric data shows no significant role of H. pylori infection in the development of GERD or in the pathogenesis of reflux esophagitis. However, current data do not provide sufficient evidence to define this relationship and further prospective large studies are needed.


Subject(s)
Esophagitis, Peptic/physiopathology , Gastric Acid/physiology , Gastroesophageal Reflux/physiopathology , Helicobacter Infections/physiopathology , Helicobacter pylori/pathogenicity , Adult , Aged , Aged, 80 and over , Endoscopy, Digestive System , Esophagitis, Peptic/microbiology , Female , Gastric Acidity Determination , Gastroesophageal Reflux/microbiology , Humans , Hydrogen-Ion Concentration , Male , Manometry , Middle Aged , Young Adult
19.
Monogr Oral Sci ; 25: 180-96, 2014.
Article in English | MEDLINE | ID: mdl-24993266

ABSTRACT

Gastric juice entering the mouth causes dental erosion. Common causes for the migration of gastric juice through the lower and upper oesophageal sphincters are reflux disease, laryngopharyngeal reflux, eating disorders, chronic alcoholism and pregnancy. Gastroesophageal reflux is a common condition affecting up to 65% of the Western population at some point in their lifetime. A typical clinical sign of acidic gastric juice entering the mouth is palatal dental erosion. As the condition becomes more chronic it becomes more widespread. There have been relatively few randomised studies investigating the aetiology of acids causing erosion. Of the few that have reported their findings, it appears that gastric acids have the potential to induce moderate-to-severe erosion. This literature review reports the conditions associated with the movement of gastric juice and dental erosion using medical and dental sources.


Subject(s)
Tooth Erosion/etiology , Alcoholism/complications , Esophageal pH Monitoring , Feeding and Eating Disorders/complications , Gastric Acid/physiology , Gastric Juice/physiology , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/diagnosis , Humans , Laryngopharyngeal Reflux/complications
20.
Antimicrob Agents Chemother ; 58(7): 4020-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798274

ABSTRACT

Posaconazole oral suspension is an extended-spectrum triazole that should be taken with food to maximize absorption. A new posaconazole tablet formulation has demonstrated improved bioavailability over the oral suspension in healthy adults in a fasting state. This study evaluated the effects of concomitant medications altering gastric pH (antacid, ranitidine, and esomeprazole) and gastric motility (metoclopramide) on the pharmacokinetics of posaconazole tablets. This was a prospective open-label 5-way crossover study in 20 healthy volunteers. In each treatment period, a single 400-mg dose (4 100-mg tablets) of posaconazole was administered alone or with 20 ml antacid (2 g of aluminum hydroxide and 2 g of magnesium hydroxide), ranitidine (150 mg), esomeprazole (40 mg), or metoclopramide (15 mg). There was a ≥ 10-day washout between treatment periods. Posaconazole exposure, time to maximum concentration of drug in serum (Tmax), and apparent terminal half-life (t1/2) were similar when posaconazole was administered alone or with medications affecting gastric pH and gastric motility. Geometric mean ratios (90% confidence intervals [CIs]) of the area under the concentration-time curve from time zero to infinity (AUC0-inf) (posaconazole with medications affecting gastric pH and gastric motility versus posaconazole alone) were 1.03 (0.88-1.20) with antacid, 0.97 (0.84-1.12) with ranitidine, 1.01 (0.87-1.17) with esomeprazole, and 0.93 (0.79-1.09) with metoclopramide. Geometric mean ratios (90% CIs) of the maximum concentration of drug in serum (Cmax) were 1.06 (0.90-1.26) with antacid, 1.04 (0.88-1.23) with ranitidine, 1.05 (0.89-1.24) with esomeprazole, and 0.86 (0.73-1.02) with metoclopramide. In summary, in healthy volunteers, the pharmacokinetics of a single 400-mg dose of posaconazole tablets was not altered to a clinically meaningful extent when posaconazole was administered alone or with medications affecting gastric pH or gastric motility.


Subject(s)
Antifungal Agents/pharmacokinetics , Gastric Acid/physiology , Gastrointestinal Motility/physiology , Triazoles/pharmacokinetics , Adult , Antacids/pharmacology , Antifungal Agents/administration & dosage , Antifungal Agents/adverse effects , Area Under Curve , Drug Interactions , Female , Healthy Volunteers , Histamine H1 Antagonists/pharmacology , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Proton Pump Inhibitors/pharmacology , Tablets , Triazoles/administration & dosage , Triazoles/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...