1
|
Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther 2023; 8:386. [PMID: 37806986 PMCID: PMC10560686 DOI: 10.1038/s41392-023-01619-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 07/20/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
Individual variability in drug response (IVDR) can be a major cause of adverse drug reactions (ADRs) and prolonged therapy, resulting in a substantial health and economic burden. Despite extensive research in pharmacogenomics regarding the impact of individual genetic background on pharmacokinetics (PK) and pharmacodynamics (PD), genetic diversity explains only a limited proportion of IVDR. The role of gut microbiota, also known as the second genome, and its metabolites in modulating therapeutic outcomes in human diseases have been highlighted by recent studies. Consequently, the burgeoning field of pharmacomicrobiomics aims to explore the correlation between microbiota variation and IVDR or ADRs. This review presents an up-to-date overview of the intricate interactions between gut microbiota and classical therapeutic agents for human systemic diseases, including cancer, cardiovascular diseases (CVDs), endocrine diseases, and others. We summarise how microbiota, directly and indirectly, modify the absorption, distribution, metabolism, and excretion (ADME) of drugs. Conversely, drugs can also modulate the composition and function of gut microbiota, leading to changes in microbial metabolism and immune response. We also discuss the practical challenges, strategies, and opportunities in this field, emphasizing the critical need to develop an innovative approach to multi-omics, integrate various data types, including human and microbiota genomic data, as well as translate lab data into clinical practice. To sum up, pharmacomicrobiomics represents a promising avenue to address IVDR and improve patient outcomes, and further research in this field is imperative to unlock its full potential for precision medicine.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China.
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, PR China.
- Central Laboratory of Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, 410013, PR China.
| |
Collapse
|
2
|
Matoshina IV, Livzan MA, Fedorin MM, Lapteva IV. Clinically significant stigmas of the severity of GERD and dissociation with the degree of damage to the esophageal mucosa — the obligatory esophagoprotection? EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2021; 1:5-11. [DOI: 10.31146/1682-8658-ecg-190-6-5-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Study goal: Identify clinically significant stigmas of reduced esophageal mucosal resistance.Materials and methods: The study included 181 patients aged 18–70 years with gastroesophageal refl ux disease (GERD).Complaints, age, sex, history of the disease, life history, physical examination data, results of laboratory and instrumental examinations, data on medication intake at the time of enrollment of the patient for the main disease and concomitant pathology were noted in the individual registration card (IRC).Results: The study demonstrated the presence of features suggesting with a higher degree of probability a reduced esophageal mucosal resistance in a certain group of patients with GERD. Individuals with early- onset GERD, males, young adults, smokers, and individuals with a high body mass index (BMI) have a higher risk of esophageal mucosal erosions. The presence of esophageal hernia of the diaphragm (EVD), intake of acetylsalicylic acid (ASA), calcium channel blockers (CCBs), or nonsteroidal anti-inflammatory drugs (NSAIDs) are factors that reduce esophageal mucosal resistance. The formation of erosive esophagitis is not associated with the subjective perception of GERD symptoms and does not affect the patient’s complaints.Conclusion: Clinical manifestations of GERD are not associated with the degree of esophageal mucosal damage and cannot be a phenotypic sign of erosive esophagitis requiring mandatory prescription of an esophagoprotector. Clinically significant stigmas of decreased esophageal mucosal resistance were revealed. The need in research of expediency and efficacy of prescription of esophagoprotector in patients with GERD, having GVHD or regularly taking BCA, ASA and other NSAIDs, including obligatory estimation of therapy effi cacy taking into account patient gender and age, has been established.
Collapse
Affiliation(s)
- I. V. Matoshina
- Federal State Educational Establishment of Higher Education Omsk State Medical University of the Ministry of Health of the Russian Federation
| | - M. A. Livzan
- Federal State Educational Establishment of Higher Education Omsk State Medical University of the Ministry of Health of the Russian Federation
| | - M. M. Fedorin
- Federal State Educational Establishment of Higher Education Omsk State Medical University of the Ministry of Health of the Russian Federation
| | - I. V. Lapteva
- Healthcare institution of the Omsk region City Hospital No. 3
| |
Collapse
|
3
|
Hutka B, Lázár B, Tóth AS, Ágg B, László SB, Makra N, Ligeti B, Scheich B, Király K, Al-Khrasani M, Szabó D, Ferdinandy P, Gyires K, Zádori ZS. The Nonsteroidal Anti-Inflammatory Drug Ketorolac Alters the Small Intestinal Microbiota and Bile Acids Without Inducing Intestinal Damage or Delaying Peristalsis in the Rat. Front Pharmacol 2021; 12:664177. [PMID: 34149417 PMCID: PMC8213092 DOI: 10.3389/fphar.2021.664177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/19/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Nonsteroidal anti-inflammatory drugs (NSAIDs) induce significant damage to the small intestine, which is accompanied by changes in intestinal bacteria (dysbiosis) and bile acids. However, it is still a question of debate whether besides mucosal inflammation also other factors, such as direct antibacterial effects or delayed peristalsis, contribute to NSAID-induced dysbiosis. Here we aimed to assess whether ketorolac, an NSAID lacking direct effects on gut bacteria, has any significant impact on intestinal microbiota and bile acids in the absence of mucosal inflammation. We also addressed the possibility that ketorolac-induced bacterial and bile acid alterations are due to a delay in gastrointestinal (GI) transit. Methods: Vehicle or ketorolac (1, 3 and 10 mg/kg) were given to rats by oral gavage once daily for four weeks, and the severity of mucosal inflammation was evaluated macroscopically, histologically, and by measuring the levels of inflammatory proteins and claudin-1 in the distal jejunal tissue. The luminal amount of bile acids was measured by liquid chromatography-tandem mass spectrometry, whereas the composition of microbiota by sequencing of bacterial 16S rRNA. GI transit was assessed by the charcoal meal method. Results: Ketorolac up to 3 mg/kg did not cause any signs of mucosal damage to the small intestine. However, 3 mg/kg of ketorolac induced dysbiosis, which was characterized by a loss of families belonging to Firmicutes (Paenibacillaceae, Clostridiales Family XIII, Christensenellaceae) and bloom of Enterobacteriaceae. Ketorolac also changed the composition of small intestinal bile by decreasing the concentration of conjugated bile acids and by increasing the amount of hyodeoxycholic acid (HDCA). The level of conjugated bile acids correlated negatively with the abundance of Erysipelotrichaceae, Ruminococcaceae, Clostridiaceae 1, Muribaculaceae, Bacteroidaceae, Burkholderiaceae and Bifidobacteriaceae. Ketorolac, under the present experimental conditions, did not change the GI transit. Conclusion: This is the first demonstration that low-dose ketorolac disturbed the delicate balance between small intestinal bacteria and bile acids, despite having no significant effect on intestinal mucosal integrity and peristalsis. Other, yet unidentified, factors may contribute to ketorolac-induced dysbiosis and bile dysmetabolism.
Collapse
Affiliation(s)
- Barbara Hutka
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bernadette Lázár
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András S Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Szilvia B László
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Nóra Makra
- Department of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Balázs Ligeti
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Bálint Scheich
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornél Király
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dóra Szabó
- Department of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Klára Gyires
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
4
|
Aleksić Sabo V, Škorić D, Jovanović-Šanta S, Nikolić I, János C, Knežević P. Synergistic activity of bile salts and their derivatives in combination with conventional antimicrobial agents against Acinetobacter baumannii. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113266. [PMID: 32810621 DOI: 10.1016/j.jep.2020.113266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bile traditionally was used in wound healing, having erodent, antioxidant and antimicrobial potential. Acinetobacter baumannii is a frequent etiological agent of wound infections, exhibiting high level of resistance to conventional antibiotics. AIM OF THE STUDY To determine the effect of selected bile acid sodium salts and their 3-dehydro (i.e. 3-oxo) derivatives, as well as their combinations with commercial antibiotics against A. baumanniia, to confirm bile ethnopharmacological application in wound healing from aspect of microbiology. MATERIALS AND METHODS The sensitivity of reference and multidrug resistant (MDR) A. baumannii strains to bile salts, their derivatives and conventional antibiotics were examined by a microtiter plate method. The interaction of bile salts/derivatives and antibiotics was examined by a checkerboard method and time kill curve method. The interaction of bile salts with ciprofloxacin in terms of micelles formation was examined by DOSY NMR technique. RESULTS The bile salts sodium deoxycholate (Na-DCA) and sodium chenodeoxycholate (Na-CDCA), as well as their derivatives sodium 3-dehydro-deoxycholate (Na-3DH-DCA) and sodium 3-dehydro-chenodeoxycholate (Na-3DH-CDCA), potentiate antibiotic activity and resensitize A. baumannii. The bile salts and their derivatives enhance A. baumannii sensitivity to antibiotics, particularly those that should penetrate cell to exhibit activity. The sodium salts of bile acid derivatives, namely Na-3DH-DCA and Na-3DH-CDCA, showed synergy against both reference and MDR strain in combination with ciprofloxacin or gentamicin, while synergy with gentamicin was obtained in all combinations, regardless of bile salt type and bacterial strains. The synergy with Na-3DH-CDCA was further confirmed by the time-kill curve method, as bacterial number decreased after 12 h. NMR experiment revealed that this bile salt derivative and ciprofloxacin form co-aggregates when bile salts concentration was higher than critical micelle concentrations (CMC), which indicate the possibility that bile salts enhance ciprofloxacin cell penetration by membrane destabilization, contributing to the synergy. CONCLUSION The synergistic interactions between bile salts or derivatives with ciprofloxacin and particularly gentamicin represent a promising strategy for the treatment of A. baumannii wound infections.
Collapse
Affiliation(s)
- Verica Aleksić Sabo
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia
| | - Dušan Škorić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia
| | - Suzana Jovanović-Šanta
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia
| | - Isidora Nikolić
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia
| | - Csanádi János
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia
| | - Petar Knežević
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovica 3, 21 000, Novi Sad, Vojvodina, Serbia.
| |
Collapse
|
5
|
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are one of the most commonly used drugs in the world, and their side effects are very high. First of all, these are NSAID gastropathy, but in the long term, 5070% of NSAIDs cause damage to the small intestine (NSAID enteropathy), sometimes with serious consequences. To date, no drugs have been proposed with proven effectiveness to prevent this side effect. Apparently, this is not due to the fully clarified mechanism of pathogenesis. The most promising is the hypothesis of the participation of individual representatives of microflora in the development of enteropathy. Therefore, modulating the intestinal flora with the help of probiotics can be the basic therapeutic strategy for the prevention and treatment of such damage.
Collapse
Affiliation(s)
- E N Kareva
- Sechenov First Moscow State Medical University (Sechenov University).,Pirogov Russian National Research Medical University
| |
Collapse
|
6
|
Study of Melatonin as Preventive Agent of Gastrointestinal Damage Induced by Sodium Diclofenac. Cells 2020; 9:cells9010180. [PMID: 31936877 PMCID: PMC7016873 DOI: 10.3390/cells9010180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Safety profile of nonsteroidal anti-inflammatory drugs (NSAIDs) has been widely studied and both therapeutic and side effects at the gastric and cardiovascular level have been generally associated with the inhibitory effect of isoform 1 (COX-1) and 2 (COX-2) cyclooxygenase enzymes. Now there are evidences of the involvement of multiple cellular pathways in the NSAIDs-mediated-gastrointestinal (GI) damage related to enterocyte redox state. In a previous review we summarized the key role of melatonin (MLT), as an antioxidant, in the inhibition of inflammation pathways mediated by oxidative stress in several diseases, which makes us wonder if MLT could minimize GI NSAIDs side effects. So, the aim of this work is to study the effect of MLT as preventive agent of GI injury caused by NSAIDs. With this objective sodium diclofenac (SD) was administered alone and together with MLT in two experimental models, ex vivo studies in pig intestine, using Franz cells, and in vivo studies in mice where stomach and intestine were studied. The histological evaluation of pig intestine samples showed that SD induced the villi alteration, which was prevented by MLT. In vivo experiments showed that SD altered the mice stomach mucosa and induced tissue damage that was prevented by MLT. The evaluation by quantitative reverse transcription PCR (RT-qPCR) of two biochemical markers, COX-2 and iNOS, showed an increase of both molecules in less injured tissues, suggesting that MLT promotes tissue healing by improving redox state and by increasing iNOS/NO that under non-oxidative condition is responsible for the maintenance of GI-epithelium integrity, increasing blood flow and promoting angiogenesis and that in presence of MLT, COX-2 may be responsible for wound healing in enterocyte. Therefore, we found that MLT may be a preventive agent of GI damages induced by NSAIDs.
Collapse
|
7
|
Scarpignato C, Bjarnason I. Drug-Induced Small Bowel Injury: a Challenging and Often Forgotten Clinical Condition. Curr Gastroenterol Rep 2019; 21:55. [PMID: 31720893 DOI: 10.1007/s11894-019-0726-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Most drugs are given by the oral route. Oral intake allows direct contact between the drug and the entire GI tract mucosa, exposing it to potential topical damage until absorption. Medication-induced GI symptoms and lesions are therefore commonly encountered in clinical practice. This review will examine the most common drugs or classes of drugs affecting small bowel function and/or structure. RECENT FINDINGS Since non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely used medicines, NSAID enteropathy is highly prevalent and brings about considerable morbidity. Antimicrobials and proton-pump inhibitors profoundly modify intestinal microbiota, affecting gut sensory and motor functions, while other drugs (like iron and gold derivatives) impair intestinal permeability. Olmesartan (and likely ACE inhibitors) induce villous atrophy and consequent malabsorption. Mycophenolate mofetil, cancer chemotherapeutic agents, and immune checkpoint inhibitors cause intestinal inflammation, abdominal pain, and diarrhea. Potassium chloride supplements may induce small bowel ulceration, stenosis, and perforation while the cotraceptive pill and anticoagulants are associated with intestinal ischemia and spontaneous intramural hematoma, respectively. In clinical practice, a deep knowledge of clinical pharmacology and toxicology and a high degree of suspicion of drug-related adverse events are mandatory. Only then, the practicing physician will be able to diagnose medication-induced small bowel lesions correctly and will implement the best strategies to treat them.
Collapse
Affiliation(s)
- Carmelo Scarpignato
- LUdeS Lugano Campus, Lugano, Switzerland.
- United Campus of Malta, Gzira, Malta.
- Faculty of Medicine, Chinese University of Hong Kong, ShaTin, Hong Kong.
- Department of Medicine & Surgery, University of Parma, Parma, Italy.
| | - Ingvar Bjarnason
- Department of Gastroenterology, King's College Hospital, London, UK
| |
Collapse
|
8
|
Contaldo A, Losurdo G, Albano F, Iannone A, Barone M, Ierardi E, Di Leo A, Principi M. The Spectrum of Small Intestinal Lesions in Patients with Unexplained Iron Deficiency Anemia Detected by Video Capsule Endoscopy. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:59. [PMID: 30818850 PMCID: PMC6473446 DOI: 10.3390/medicina55030059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/16/2019] [Accepted: 02/21/2019] [Indexed: 02/07/2023]
Abstract
Background and objectives: Video-capsule endoscopy (VCE) has shown a large range (38⁻83%) of diagnostic yield in unexplained iron deficiency anemia (IDA) and obscure-occult bleeding. Therefore, we retrospectively investigated the VCE-detected spectrum and the prevalence of small bowel injuries and associated risk factors in inpatients with both of the above reported conditions. Methods: We selected inpatients with IDA (hemoglobin <12 g/dL in women, <13 g/dL in men) and obscure-occult bleeding. We excluded VCE indications other than IDA. Complete medical histories and laboratory tests were collected. All subjects underwent PillCam SB2/SB3. The VCE feature Lewis score was calculated when appropriate. We used the t-test and Fisher's exact test for continuous and categorical variables, respectively, in univariate analysis. For multivariate analysis, we used binomial logistic regression. Results: We retrieved 109 patients (female:male ratio of 53:56; age 63.4 ± 18.9 years). Eighty patients (73.4%) showed ≥1 small bowel lesions. The Lewis score was calculated in 41 patients: 13 (31.7%) showed a mild (<135) and 28 (68.3%) a moderate-severe (135⁻790 and >790, respectively) score. In univariate analysis, the small bowel transit time (6.2 ± 2.9 versus 5.2 ± 2.1 h; p = 0.049) and non-steroidal anti-inflammatory drug use for at least two weeks (17.5% versus 0%; p = 0.01) were significantly higher in subjects with injuries. These associations were not confirmed at multivariate analysis. The severity of a lesion directly correlated with proton pump inhibitor (PPI) use and duration (not confirmed in multivariate analysis). VCE can reveal the source of obscure-occult bleeding in a high percentage of unexplained IDAs. A wide spectrum of endoscopic pictures may be found. Known as well as supposed risk factors for small bowel lesions may be detected.
Collapse
Affiliation(s)
- Antonella Contaldo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Giuseppe Losurdo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Francesca Albano
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Andrea Iannone
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Michele Barone
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Enzo Ierardi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Alfredo Di Leo
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Mariabeatrice Principi
- Section of Gastroenterology, Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| |
Collapse
|
9
|
Colucci R, Pellegrini C, Fornai M, Tirotta E, Antonioli L, Renzulli C, Ghelardi E, Piccoli E, Gentile D, Benvenuti L, Natale G, Fulceri F, Palazón-Riquelme P, López-Castejón G, Blandizzi C, Scarpignato C. Pathophysiology of NSAID-Associated Intestinal Lesions in the Rat: Luminal Bacteria and Mucosal Inflammation as Targets for Prevention. Front Pharmacol 2018; 9:1340. [PMID: 30555323 PMCID: PMC6281992 DOI: 10.3389/fphar.2018.01340] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 10/31/2018] [Indexed: 12/11/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) can damage the small intestine, mainly through an involvement of enteric bacteria. This study examined the pathophysiology of NSAID-associated intestinal lesions in a rat model of diclofenac-enteropathy and evaluated the effect of rifaximin on small bowel damage. Enteropathy was induced in 40-week old male rats by intragastric diclofenac (4 mg/kg BID, 14 days). Rifaximin (delayed release formulation) was administered (50 mg/kg BID) 1 h before the NSAID. At the end of treatments, parameters dealing with ileal damage, inflammation, barrier integrity, microbiota composition, and TLR-NF-κB-inflammasome pathway were evaluated. In addition, the modulating effect of rifaximin on NLRP3 inflammasome was tested in an in vitro cell system. Diclofenac induced intestinal damage and inflammation, triggering an increase in tissue concentrations of tumor necrosis factor and interleukin-1β, higher expression of TLR-2 and TLR-4, MyD88, NF-κB and activation of caspase-1. In addition, the NSAID decreased ileal occludin expression and provoked a shift of bacterial phyla toward an increase in Proteobacteria and Bacteroidetes abundance. All these changes were counterbalanced by rifaximin co-administration. This drug was also capable of increasing the proportion of Lactobacilli, a genus depleted by the NSAID. In LPS-primed THP-1 cells stimulated by nigericin (a model to study the NLRP3 inflammasome), rifaximin reduced IL-1β production in a concentration-dependent fashion, this effect being associated with inhibition of the up-stream caspase-1 activation. In conclusion, diclofenac induced ileal mucosal lesions, driving inflammatory pathways and microbiota changes. In conclusion, rifaximin prevents diclofenac-induced enteropathy through both anti-bacterial and anti-inflammatory activities.
Collapse
Affiliation(s)
- Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Erika Tirotta
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cecilia Renzulli
- Reasearch & Development Department, Alfasigma SpA, Bologna, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elena Piccoli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Federica Fulceri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pablo Palazón-Riquelme
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom
| | - Gloria López-Castejón
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carmelo Scarpignato
- Clinical Pharmacology & Digestive Pathophysiology Unit, Department of Clinical & Experimental Medicine, University of Parma, Parma, Italy
| |
Collapse
|
10
|
Tedesco D, Thapa M, Chin CY, Ge Y, Gong M, Li J, Gumber S, Speck P, Elrod EJ, Burd EM, Kitchens WH, Magliocca JF, Adams AB, Weiss DS, Mohamadzadeh M, Grakoui A. Alterations in Intestinal Microbiota Lead to Production of Interleukin 17 by Intrahepatic γδ T-Cell Receptor-Positive Cells and Pathogenesis of Cholestatic Liver Disease. Gastroenterology 2018; 154:2178-2193. [PMID: 29454797 PMCID: PMC5985208 DOI: 10.1053/j.gastro.2018.02.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/16/2018] [Accepted: 02/07/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Variants at the ABCB4 or MDR2 locus, which encodes a biliary transport protein, are associated with a spectrum of cholestatic liver diseases. Exacerbation of liver disease has been linked to increased hepatic levels of interleukin (IL) 17, yet the mechanisms of this increase are not understood. We studied mice with disruption of Mdr2 to determine how defects in liver and alteration in the microbiota contribute to production of IL17 by intrahepatic γδ T cells. METHODS We performed studies with Mdr2-/- and littermate FVB/NJ (control) mice. IL17 was measured in serum samples by an enzyme-linked immunosorbent assay. Mice were injected with neutralizing antibodies against the γδ T-cell receptor (TCR; anti-γδ TCR) or mouse IL17A (anti-IL17A). Livers were collected and bacteria were identified in homogenates by culture procedures; TCRγδ+ cells were isolated by flow cytometry. Fecal samples were collected from mice and analyzed by 16S ribosomal DNA sequencing. Cells were stimulated with antibodies or bacteria, and cytokine production was measured. We obtained tissues from 10 patients undergoing liver transplantation for primary sclerosing cholangitis or chronic hepatitis C virus infection. Tissues were analyzed for cytokine production by γδ TCR+ cells. RESULTS Mdr2-/- mice had collagen deposition around hepatic bile ducts and periportal-bridging fibrosis with influx of inflammatory cells and increased serum levels of IL17 compared with control mice. Administration of anti-IL17A reduced hepatic fibrosis. Livers from Mdr2-/- mice had increased numbers of IL17A+ γδTCR+ cells-particularly of IL17A+ Vγ6Jγ1 γδ TCR+ cells. Fecal samples from Mdr2-/- mice were enriched in Lactobacillus, and liver tissues were enriched in Lactobacillus gasseri compared with control mice. Mdr2-/- mice also had increased intestinal permeability. The γδ TCR+ cells isolated from Mdr2-/- livers produced IL17 in response to heat-killed L gasseri. Intraperitoneal injection of control mice with L gasseri led to increased serum levels of IL17 and liver infiltration by inflammatory cells; injection of these mice with anti-γδ TCR reduced serum level of IL17. Intravenous injections of Mdr2-/- mice with anti-γδ TCR reduced fibrosis; liver levels of IL17, and inflammatory cells; and serum levels of IL17. γδTCR+ cells isolated from livers of patients with primary sclerosing cholangitis, but not hepatitis C virus infection, produced IL17. CONCLUSIONS In Mdr2-/- mice, we found development of liver fibrosis and inflammation to require hepatic activation of γδ TCR+ cells and production of IL17 mediated by exposure to L gasseri. This pathway appears to contribute to development of cholestatic liver disease in patients.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Adult
- Aged
- Animals
- Bile Ducts/cytology
- Bile Ducts/immunology
- Bile Ducts/microbiology
- Cells, Cultured
- Cholangitis, Sclerosing/microbiology
- Cholangitis, Sclerosing/pathology
- Cholangitis, Sclerosing/surgery
- Cholestasis/immunology
- Cholestasis/microbiology
- Cholestasis/pathology
- Cholestasis/surgery
- Disease Models, Animal
- End Stage Liver Disease/microbiology
- End Stage Liver Disease/pathology
- End Stage Liver Disease/surgery
- Female
- Gastrointestinal Microbiome
- Hepatitis C, Chronic/pathology
- Hepatitis C, Chronic/surgery
- Hepatitis C, Chronic/virology
- Humans
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/blood
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Intraepithelial Lymphocytes/metabolism
- Lactobacillus gasseri/immunology
- Liver/cytology
- Liver/immunology
- Liver/microbiology
- Liver/pathology
- Liver Cirrhosis/immunology
- Liver Cirrhosis/microbiology
- Liver Cirrhosis/pathology
- Liver Cirrhosis/surgery
- Liver Transplantation
- Male
- Mice
- Mice, Knockout
- Middle Aged
- Receptors, Antigen, T-Cell, gamma-delta/antagonists & inhibitors
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Young Adult
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Dana Tedesco
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia
| | - Manoj Thapa
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia
| | - Chui Yoke Chin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Emory Antibiotic Resistance Center, Atlanta, Georgia
| | - Yong Ge
- Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Minghao Gong
- Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Jing Li
- Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Sanjeev Gumber
- Division of Pathology and Laboratory Medicine, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia
| | - Patrick Speck
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia
| | - Elizabeth J Elrod
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia
| | - Eileen M Burd
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Emory Antibiotic Resistance Center, Atlanta, Georgia; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - William H Kitchens
- Department of Surgery, Transplant, Emory University School of Medicine, Atlanta, Georgia
| | - Joseph F Magliocca
- Department of Surgery, Transplant, Emory University School of Medicine, Atlanta, Georgia
| | - Andrew B Adams
- Department of Surgery, Transplant, Emory University School of Medicine, Atlanta, Georgia
| | - David S Weiss
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Emory Antibiotic Resistance Center, Atlanta, Georgia
| | - Mansour Mohamadzadeh
- Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida; Division of Hepatology, Gastroenterology, and Nutrition; University of Florida, Gainesville, Florida
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes Research Primate Center, Emory University School of Medicine, Atlanta, Georgia; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
11
|
Human and rat precision-cut intestinal slices as ex vivo models to study bile acid uptake by the apical sodium-dependent bile acid transporter. Eur J Pharm Sci 2018; 121:65-73. [PMID: 29751102 DOI: 10.1016/j.ejps.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 03/06/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
|
12
|
Abstract
PURPOSE OF REVIEW The review describes the effects of NSAID gastrointestinal toxicity on the small bowel, wherein injury is as prevalent as in the gastroduodenum. This is well documented by capsule endoscopy, which also provides an endoscopic endpoint for novel treatment strategies. RECENT FINDINGS Appreciation of the prevalence of NSAID enteropathy has grown with capsule endoscopy and the use of composite end points that include obscure haemoglobin decreases in clinical studies. Along with dual cyclo-oxygenase isoform inhibition, gram-negative antigen presentation plays a role in the pathogenesis of NSAID enteropathy. The concomitant use of proton pump inhibitor (PPI) agents with both selective and nonselective NSAIDs in gastroduodenal protection exacerbates endoscopic enteropathy, which is thought to be due to intestinal dysbiosis. Strategies for small bowel mucosal protection include prostaglandin repletion and augmenting the intestinal flora. SUMMARY NSAID- related enteropathy is common and often presents subclinically. The most common presentation is anaemia, which may be less common in those on cyclo-oxygenase (COX)-2 inhibitors than nonselective NSAIDs. PPI use worsens NSAID enteropathy with more occult bleeding and ulceration and has been linked to gram-negative intestinal dysbiosis. If NSAID cessation is not possible, COX-2 inhibition without PPI therapy should be considered in patients with upper gastrointestinal risk factors. Mucoprotective agents such as misoprostol and rebamipide show promise and probiotics may have a future role.
Collapse
|
13
|
Kurnijasanti R, Putri AA. THE EFFECTS OF BANANA STEM (Musa paradisiaca var. sapientum) EXTRACT ON HISTOPATHOLOGIC GASTRIC OF RATS INDUCED BY INDOMETACHIN. FOLIA MEDICA INDONESIANA 2017. [DOI: 10.20473/fmi.v52i4.5470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This research aims to find out the administration effect of Ambon banana stem extract (Musa paradisiaca var. sapientum) to prevent gastric damage and observe histopathology of rat’s gastric induced by indomethacin. This research used 30 male rats age 8-12 week having 150 gram weight. They were selected randomly and divided into five groups. Negative control (K-) was given 0.5 ml CMC Na 0.5 % for 9 days and 0.5 ml corn oil was given on 10th. Positive control (K+) was given 0.5 ml CMC Na 0.5% for 9 days and then given Indomethacin emultion 5 mg/0.5 ml/150 g BW once on 10th day, and the other groups were given Ambon banana stem extract for (P1) 20 mg/150 g BW, (P2) 40 mg/150 g BW and (P3) 80 mg/150 g BW for 9 days. Afterwards, they were given Indometachin emultion 5 mg/150 g BW once on 10th day. Each gastric specimen was processed and histopathological changes were observed. Scoring of mucosa epithelium erosion and hemorrhagic, as qualitative data, was analyzed by Kruskall-Wallis test and continued by using Z test. The result shows that Pisang Ambon’s stem extract reduce significantly in gastric mucosa epithelium erosion and hemorrhagic induced by Indomethacin (p<0.05).
Collapse
|
14
|
Lué A, Lanas A. Protons pump inhibitor treatment and lower gastrointestinal bleeding: Balancing risks and benefits. World J Gastroenterol 2016; 22:10477-10481. [PMID: 28082800 PMCID: PMC5192259 DOI: 10.3748/wjg.v22.i48.10477] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/28/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023] Open
Abstract
Proton pump inhibitors (PPIs) represent a milestone in the treatment of acid-related diseases, and are the mainstay in preventing upper gastrointestinal bleeding in high-risk patients treated with nonsteroidal anti-inflammatory drugs (NSAIDs) or low-dose aspirin. However, this beneficial effect does not extend to the lower gastrointestinal tract. PPIs do not prevent NSAID or aspirin-associated lower gastrointestinal bleeding (LGB). PPIs may increase both small bowel injury related to NSAIDs and low-dose aspirin treatment and the risk of LGB. Recent studies suggested that altering intestinal microbiota by PPIs may be involved in the pathogenesis of NSAID-enteropathy. An increase in LGB hospitalization rates may occur more frequently in older patients with more comorbidities and are associated with high hospital resource utilization, longer hospitalization, and increased mortality. Preventive strategies for NSAID and aspirin-associated gastrointestinal bleeding should be directed toward preventing both upper and lower gastrointestinal damage. Future research should be directed toward identifying patients at low-risk for gastrointestinal events associated with the use of NSAIDs or aspirin to avoid inappropriate PPI prescribing. Alternatively, the efficacy of new pharmacologic strategies should be evaluated in high-risk groups, with the aim of reducing the risk of both upper and lower gastrointestinal bleeding in these patients.
Collapse
|
15
|
Davison G, Marchbank T, March DS, Thatcher R, Playford RJ. Zinc carnosine works with bovine colostrum in truncating heavy exercise-induced increase in gut permeability in healthy volunteers. Am J Clin Nutr 2016; 104:526-36. [PMID: 27357095 DOI: 10.3945/ajcn.116.134403] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Heavy exercise causes gut symptoms and, in extreme cases, heat stroke that is due to the increased intestinal permeability of luminal toxins. OBJECTIVE We examined whether zinc carnosine (ZnC), a health-food product taken alone or in combination with bovine colostrum (a natural source of growth factors), would moderate such effects. DESIGN Eight volunteers completed a 4-arm, double-blind, placebo-controlled crossover protocol (14 d of placebo, ZnC, colostrum, or ZnC plus colostrum) before undertaking standardized exercise 2 and 14 d after the start of treatment. Changes in epithelial resistance, apoptosis signaling molecules, and tight junction (TJ) protein phosphorylation in response to a 2°C rise in body temperature were determined with the use of Caco-2 and HT29 intestinal cells. RESULTS Body temperature increased 2°C, and gut permeability (5-h urinary lactulose:rhamnose ratios) increased 3-fold after exercise (from 0.32 ± 0.016 baseline to 1.0 ± 0.017 at 14 d; P < 0.01). ZnC or colostrum truncated the rise by 70% after 14 d of treatment. The combination treatment gave an additional benefit, and truncated exercise induced increase at 2 d (30% reduction; P < 0.01). A 2°C temperature rise in in vitro studies caused the doubling of apoptosis and reduced epithelial resistance 3-4-fold. ZnC or colostrum truncated these effects (35-50%) with the greatest response seen with the combination treatment (all P < 0.01). Mechanisms of action included increasing heat shock protein 70 and truncating temperature-induced changes in B cell leukemia/lymphoma-2 associated X protein α and B cell lymphoma 2. ZnC also increased total occludin and reduced phosphorylated tyrosine claudin, phosphorylated tyrosine occludin, and phosphorylated serine occludin, thereby enhancing the TJ formation and stabilization. CONCLUSION ZnC, taken alone or with colostrum, increased epithelial resistance and the TJ structure and may have value for athletes and in the prevention of heat stroke in military personnel. This trial was registered at www.isrctn.com as ISRCTN51159138.
Collapse
Affiliation(s)
- Glen Davison
- Endurance Research Group, School of Sport and Exercise Sciences, University of Kent, Kent, United Kingdom
| | - Tania Marchbank
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, United Kingdom; Centre for Immunobiology, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine, Queen Mary, University of London, London, United Kingdom
| | - Daniel S March
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom; and
| | - Rhys Thatcher
- Institute of Biological Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - Raymond J Playford
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, United Kingdom;
| |
Collapse
|
16
|
Verhaegh BPM, de Vries F, Masclee AAM, Keshavarzian A, de Boer A, Souverein PC, Pierik MJ, Jonkers DMAE. High risk of drug-induced microscopic colitis with concomitant use of NSAIDs and proton pump inhibitors. Aliment Pharmacol Ther 2016; 43:1004-13. [PMID: 26956016 DOI: 10.1111/apt.13583] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/02/2015] [Accepted: 02/17/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Microscopic colitis (MC) is a chronic bowel disorder characterised by watery diarrhoea. Nonsteroidal anti-inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs), selective serotonin reuptake inhibitors (SSRIs) and statins have been associated with MC. However, underlying mechanisms remain unclear. AIM To study the association between exposure to these drugs and MC, with attention to time of exposure, duration, dosage and combined exposure, and to test hypotheses on underlying pharmacological mechanisms. METHODS A case-control study was conducted using the British Clinical Practice Research Datalink. MC cases (1992-2013) were matched to MC-naive controls on age, sex and GP practice. Drug exposure was stratified according to time of exposure, duration of exposure or dosage. Conditional logistic regression analysis was applied to calculate adjusted odds ratios (AORs). RESULTS In total, 1211 cases with MC were matched to 6041 controls. Mean age was 63.4 years, with 73.2% being female. Current use of NSAIDs (AOR 1.86, 95% CI 1.39-2.49), PPIs (AOR 3.37, 95% CI 2.77-4.09) or SSRIs (AOR 2.03, 95% CI 1.58-2.61) was associated with MC compared to never or past use. Continuous use for 4-12 months further increased the risk of MC. Strongest associations (fivefold increased risk) were observed for concomitant use of PPIs and NSAIDs. Statins were not associated with MC. CONCLUSIONS Current exposure to NSAIDs, PPIs or SSRIs and prolonged use for 4-12 months increased the risk of MC. Concomitant use of NSAIDs and PPIs showed the highest risk of MC. Acid suppression related dysbiosis may contribute to the PPI effect, which may be exacerbated by NSAID-related side-effects.
Collapse
Affiliation(s)
- B P M Verhaegh
- Division of Gastroenterology - Hepatology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - F de Vries
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht, The Netherlands
- Clinical Pharmacology & Toxicology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - A A M Masclee
- Division of Gastroenterology - Hepatology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - A Keshavarzian
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht, The Netherlands
- Division of Digestive Diseases and Nutrition, Rush University, Chicago, IL, USA
| | - A de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht, The Netherlands
| | - P C Souverein
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht, The Netherlands
| | - M J Pierik
- Division of Gastroenterology - Hepatology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - D M A E Jonkers
- Division of Gastroenterology - Hepatology, Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
17
|
Vemu B, Selvasubramanian S, Pandiyan V. Emu oil offers protection in Crohn's disease model in rats. Altern Ther Health Med 2016; 16:55. [PMID: 26852336 PMCID: PMC4744428 DOI: 10.1186/s12906-016-1035-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 02/01/2016] [Indexed: 01/09/2023]
Abstract
Background Emu oil is a product of animal origin used for the treatment of inflammation, burns etc. as a part of aboriginal medicine in Australia. Crohn’s disease is a common inflammatory manifestation in humans and other animal species relating to the ulceration and digestive disturbances in upper gastro-intestinal tract. Aloe vera is commonly used substance from plant sources for inflammation, wound healing and various other properties. Given the difference in the source of the substances all the while playing a similar therapeutic role in different parts of the world, the present investigation was undertaken to evaluate the protective effect of aloe vera and emu oil alone and in combination; in comparison to sulfasalazine (Allopathic drug) as an alternative for the treatment of Crohn’s disease. Methods Wistar albino rats were divided into six groups with two sub-groups of six animals each. After pre-treating the animals with sulfasalazine, aloe vera, emu oil and their combination for five consecutive days, the animals were sub-cutaneously administered indomethacin on 4th and 5th day and each sub-group was sacrificed on day 6 and 9. After sacrifice, serum and intestine of these animals was collected. Intestine length from duodenum till caecum was measured for estimating relative organ weight and disease activity index. Part of intestine was preserved in formalin for histopathology while the rest was used for analysis of oxidative parameters and myeloperoxidase. Serum collected was used for measuring alkaline phosphatase and cholesterol. Results Assessment of the parameters in treatment groups indicated that the combination of aloe vera and emu oil resulted in better protection by suppressing the oxidative (P < 0.05) and histomorphological changes indicating a enhanced effect of these two agents which was found to be better than sulfasalazine. Conclusion The combination of emu oil and aloe vera exhibited enhanced effect resulting in significant protection from indomethacin induced ulceration. This might be due to the different mechanism of anti-inflammatory effects (Salicylic acid in aloe vera and n3, n6 fatty acids acting as pseudosubstrates to cyclooxygenase enzyme) of components of the animal and plant products tested.
Collapse
|
18
|
Blackler RW, De Palma G, Manko A, Da Silva GJ, Flannigan KL, Bercik P, Surette MG, Buret AG, Wallace JL. Deciphering the pathogenesis of NSAID enteropathy using proton pump inhibitors and a hydrogen sulfide-releasing NSAID. Am J Physiol Gastrointest Liver Physiol 2015; 308:G994-1003. [PMID: 25882612 DOI: 10.1152/ajpgi.00066.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The small intestine is a significant site of ulceration and bleeding induced by nonsteroidal anti-inflammatory drugs (NSAIDs). The pathogenesis is poorly understood. The present study explored the roles of bile, bacteria, and enterohepatic circulation to NSAID enteropathy, using both a conventional NSAID (naproxen) and a gastrointestinal-safe naproxen derivative (ATB-346), as well as proton pump inhibitors (PPIs). Rats were treated orally with naproxen or equimolar doses of ATB-346 over a 5-day period, with or without PPI administration, and intestinal damage was quantified. The cytotoxicity of bile from the rats was evaluated in vitro. Biliary excretion of naproxen and ATB-346 was determined. The impact of the NSAIDs and of PPIs on the composition of the intestinal microbiota was examined by deep sequencing of 16s rRNA. Naproxen caused significant intestinal damage and inflammation, whereas ATB-346 did not. Naproxen, but not ATB-346, dose dependently increased the cytotoxicity of bile, and it was further increased by PPI coadministration. Whereas biliary excretion of naproxen was significant in naproxen-treated rats, it was greatly reduced in rats treated with ATB-346. The enteric microbiota of naproxen-treated rats was distinct from that in vehicle- or ATB-346-treated rats, and PPI administration caused significant intestinal dysbiosis. The increase in cytotoxicity of bile induced by naproxen and PPIs may contribute significantly to intestinal ulceration and bleeding. Some of these effects may occur secondary to significant changes in the jejunal microbiota induced by both naproxen and PPIs.
Collapse
Affiliation(s)
- Rory W. Blackler
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Giada De Palma
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anna Manko
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Gabriela J. Da Silva
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Faculty of Pharmacy and Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Kyle L. Flannigan
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Premysl Bercik
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, Alberta; and
| | - John L. Wallace
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Faculty of Medicine, Universidade Camilo Castelo Branco, Sao Paulo, SP, Brazil
| |
Collapse
|
19
|
Dial EJ, Dawson PA, Lichtenberger LM. In vitro evidence that phosphatidylcholine protects against indomethacin/bile acid-induced injury to cells. Am J Physiol Gastrointest Liver Physiol 2015; 308:G217-22. [PMID: 25477376 PMCID: PMC4312955 DOI: 10.1152/ajpgi.00322.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Indomethacin is a powerful analgesic nonsteroidal anti-inflammatory drug (NSAID), but is limited in use by its primary side effect to cause gastrointestinal bleeding and serious injury. One factor important for exacerbating NSAID injury is the presence of bile acids, which may interact with indomethacin to form toxic mixed micelles in the gut. The development of a safer gastrointestinal formulation of indomethacin that is chemically complexed with phosphatidylcholine (PC-indomethacin) may offer an improved therapeutic agent, particularly in the presence of bile acid, but its potential protective mechanism is incompletely understood. Intestinal epithelial cells (IEC-6) were tested for injury with indomethacin (alone and plus various bile acids) compared with PC-indomethacin (alone and plus bile acids). To explore a role for bile acid uptake into cells as a requirement for NSAID injury, studies were performed using Madin-Darby canine kidney cells transfected with the apical sodium-dependent bile acid transporter (ASBT). Indomethacin, but not PC-indomethacin, was directly and dose-dependently injurious to IEC-6 cells. Similarly, the combination of any bile acid plus indomethacin, but not PC-indomethacin, induced cell injury. The expression of ASBT had a modest effect on the acute cytotoxicity of indomethacin in the presence of some conjugated bile acids. Complexing PC with indomethacin protected against the acute intestinal epithelial injury caused by indomethacin regardless of the presence of bile acids. The presence of luminal bile acid, but not its carrier-mediated uptake into the enterocyte, is required for acute indomethacin-induced cell injury. It is likely that initial cell damage induced by indomethacin occurs at or near the cell membrane, an effect exacerbated by bile acids and attenuated by PC.
Collapse
Affiliation(s)
- Elizabeth J. Dial
- 1Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Paul A. Dawson
- 2Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Lenard M. Lichtenberger
- 1Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| |
Collapse
|
20
|
How to mechanistically explain the CONDOR study data. Med Hypotheses 2014; 84:14-9. [PMID: 25433956 DOI: 10.1016/j.mehy.2014.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 11/04/2014] [Accepted: 11/09/2014] [Indexed: 02/02/2023]
Abstract
Results of the CONDOR study suggest that in osteoarthritis and rheumatoid arthritis patients at elevated risk of gastrointestinal (GI) events, treatment with celecoxib, a cyclooxygenase (COX)-2 selective non-steroidal anti-inflammatory drug (NSAID), demonstrated significantly lower toxicity in the upper and lower (GI) tract when compared to the non-selective NSAID diclofenac plus a proton-pump-inhibitor (PPI), omeprazole. According to current knowledge, traditional NSAIDs (tNSAIDs) as non-selective COX-inhibitors exert their damaging effects on the upper GI tract, largely by reduction of the COX-1 related synthesis of gastro-protective prostaglandins. Thus, the question arises, how NSAIDs do exert their damaging effects especially in the lower GI tract and how to explain the reduced risk of a COX-2 selective inhibitor, celecoxib. Here we hypothesize, that the toxicity of celecoxib on enteral mucosa cells is lower than observed with other NSAIDs, and can be explained COX-independently by typical physicochemical properties of the NSAID substances (e.g., acidic, lipophilic, amphiphilic, surfactant properties). As a consequence these features account for differences in (1) uncoupling effects on mitochondria, (2) effects on cell membrane integrity, and/or (3) formation of "toxic micelles" with bile salts. The evidence for these differences is mainly based on experimental findings. However, several phenomena show differences in extent (e.g., uncoupling effects). The reduced toxicity appears to be rather a substance-specific characteristic. This is an unconditional reason to carry on investigating these phenomena in experimental and large-scale clinical trials.
Collapse
|
21
|
Prakash P, Gorfe AA. Phosphatidylcholine Attenuates Aggregation of Nonsteroidal Anti-Inflammatory Drugs with Bile Acid. Biochemistry 2013; 52:7461-9. [DOI: 10.1021/bi400723r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Priyanka Prakash
- Department of Integrative
Biology and Pharmacology, University of Texas at Houston, 6431
Fannin Street, Houston, Texas 77030, United States
| | - Alemayehu A. Gorfe
- Department of Integrative
Biology and Pharmacology, University of Texas at Houston, 6431
Fannin Street, Houston, Texas 77030, United States
| |
Collapse
|
22
|
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used drugs in the world; nevertheless, about 50-70% of patients on long-term NSAIDs develop small intestine injury, namely NSAID enteropathy, sometimes with serious outcomes. No medications with proven efficacy are yet available to prevent NSAID enteropathy. A series of therapeutic strategies targeting the different mechanisms involved in small bowel injury have been investigated, but without definitive results. Intestinal bacteria and their degradation products are essential for the development of NSAID-induced small bowel lesions, because "germ-free" animals were found to be resistant to indomethacin injuries. Therefore, it has been suggested that modulating the intestinal flora, for example by using probiotics, could protect against NSAID enteropathy. In this work, we reviewed the main therapeutic strategies for NSAID enteropathy, in particular analyzing the available studies relating to the eventual protective role of probiotics. We found that results are not all concordant; nevertheless, the more recent studies provide better understanding about pathogenetic mechanisms involved in small intestinal injury and the role of probiotics, and show encouraging results. Larger and well-designed studies should be performed to evaluate the actual role of probiotics in NSAID enteropathy, the eventual differences among probiotic strains, dose-responses, and optimal duration of therapy.
Collapse
|
23
|
Prakash P, Sayyed-Ahmad A, Zhou Y, Volk DE, Gorenstein DG, Dial E, Lichtenberger LM, Gorfe AA. Aggregation behavior of ibuprofen, cholic acid and dodecylphosphocholine micelles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:3040-7. [PMID: 22885171 DOI: 10.1016/j.bbamem.2012.07.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/20/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently used to treat chronic pain and inflammation. However, prolonged use of NSAIDs has been known to result in Gastrointestinal (GI) ulceration/bleeding, with a bile-mediated mechanism underlying their toxicity to the lower gut. Bile acids (BAs) and phosphatidylcholines (PCs), the major components of bile, form mixed micelles to reduce the membrane disruptive actions of monomeric BAs and simple BA micelles. NSAIDs are suspected to alter the BA/PC balance in the bile, but the molecular interactions of NSAID-BA or NSAID-BA-PC remain undetermined. In this work, we used a series of all-atom molecular dynamics simulations of cholic acid (CA), ibuprofen (IBU) and dodecylphosphocholine (DPC) mixtures to study the spontaneous aggregation of CA and IBU as well as their adsorption on a DPC micelle. We found that the size of CA-IBU mixed micelles varies with their molar ratio in a non-linear manner, and that micelles of different sizes adopt similar shapes but differ in composition and internal interactions. These observations are supported by NMR chemical shift changes, NMR ROESY crosspeaks between IBU and CA, and dynamic light scattering experiments. Smaller CA-IBU aggregates were formed in the presence of a DPC micelle due to the segregation of CA and IBU away from each other by the DPC micelle. While the larger CA-IBU aggregates arising from higher IBU concentrations might be responsible for NSAID-induced intestinal toxicity, the absence of larger CA-IBU aggregates in the presence of DPC micelles may explain the observed attenuation of NSAID toxicity by PCs.
Collapse
Affiliation(s)
- Priyanka Prakash
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Cattle bile aggravates diclofenac sodium-induced small intestinal injury in mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:315858. [PMID: 21584236 PMCID: PMC3092561 DOI: 10.1155/2011/315858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 02/17/2011] [Accepted: 02/18/2011] [Indexed: 11/18/2022]
Abstract
Cattle bile (CB) has long been used in Japan as an ingredient of digestive medicines. Bile acids are major chemical constituents of CB, and CB ingestion is assumed to affect small intestinal injury induced by nonsteroidal anti-inflammatory drugs (NSAIDs). Mice were fed a diet supplemented with or without CB for 7 days and treated with diclofenac sodium (DIF) to induce small intestinal injury. Lesion formation was enhanced, and PGE2 content and COX expression levels were elevated in the small intestine of DIF-treated mice fed the CB diet compared with those fed the control diet. The administration of a reconstituted mixture of bile acids found in CB enhanced lesion formation in DIF-treated mice. CB administration elevated the contents of CB-derived bile acids in the small intestine, some of which exhibited a high cytotoxicity to cultured intestinal epithelial cells. These results suggest that the elevated levels of CB-derived cytotoxic bile acids in the small intestine contribute to the aggravation of DIF-induced small intestinal injury. The use of CB may be limited during the therapy of inflammatory diseases with NSAIDs.
Collapse
|
25
|
Marchbank T, Davison G, Oakes JR, Ghatei MA, Patterson M, Moyer MP, Playford RJ. The nutriceutical bovine colostrum truncates the increase in gut permeability caused by heavy exercise in athletes. Am J Physiol Gastrointest Liver Physiol 2011; 300:G477-84. [PMID: 21148400 DOI: 10.1152/ajpgi.00281.2010] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Heavy exercise causes gut symptoms and, in extreme cases, "heat stroke" partially due to increased intestinal permeability of luminal toxins. We examined bovine colostrum, a natural source of growth factors, as a potential moderator of such effects. Twelve volunteers completed a double-blind, placebo-controlled, crossover protocol (14 days colostrum/placebo) prior to standardized exercise. Gut permeability utilized 5 h urinary lactulose-to-rhamnose ratios. In vitro studies (T84, HT29, NCM460 human colon cell lines) examined colostrum effects on temperature-induced apoptosis (active caspase-3 and 9, Baxα, Bcl-2), heat shock protein 70 (HSP70) expression and epithelial electrical resistance. In both study arms, exercise increased blood lactate, heart rate, core temperature (mean 1.4°C rise) by similar amounts. Gut hormone profiles were similar in both arms although GLP-1 levels rose following exercise in the placebo but not the colostrum arm (P = 0.026). Intestinal permeability in the placebo arm increased 2.5-fold following exercise (0.38 ± 0.012 baseline, to 0.92 ± 0.014, P < 0.01), whereas colostrum truncated rise by 80% (0.38 ± 0.012 baseline to 0.49 ± 0.017) following exercise. In vitro apoptosis increased by 47-65% in response to increasing temperature by 2°C. This effect was truncated by 60% if colostrum was present (all P < 0.01). Similar results were obtained examining epithelial resistance (colostrum truncated temperature-induced fall in resistance by 64%, P < 0.01). Colostrum increased HSP70 expression at both 37 and 39°C (P < 0.001) and was truncated by addition of an EGF receptor-neutralizing antibody. Temperature-induced increase in Baxα and reduction in Bcl-2 was partially reversed by presence of colostrum. Colostrum may have value in enhancing athletic performance and preventing heat stroke.
Collapse
Affiliation(s)
- Tania Marchbank
- Digestive Diseases, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine, Queen Mary University of London, UK
| | | | | | | | | | | | | |
Collapse
|
26
|
Goldstein JL, Jungwirthová A, David J, Spindel E, Bouchner L, Pešek F, Searle S, Skopek J, Grim J, Ulč I, Sewell KL. Clinical trial: endoscopic evaluation of naproxen etemesil, a naproxen prodrug, vs. naproxen - a proof-of-concept, randomized, double-blind, active-comparator study. Aliment Pharmacol Ther 2010; 32:1091-101. [PMID: 20804454 DOI: 10.1111/j.1365-2036.2010.04442.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs are associated with upper gastrointestinal mucosal injury. Naproxen etemesil is a lipophilic, non-acidic, inactive prodrug of naproxen that is hydrolysed to pharmacologically active naproxen once absorbed. We hypothesized that with lesser topical exposure to naproxen from the prodrug, there would be reduced gastroduodenal mucosal injury compared with naproxen. AIM To compare the degree of endoscopic mucosal damage of naproxen etemesil vs. naproxen. METHODS This multicentre, randomized, double-blind, double-dummy trial compared oral naproxen etemesil 1200 mg twice daily (n = 61) with naproxen 500 mg twice daily (n = 59) for 7.5 days in 120 healthy subjects (45-70 years; mean 51 years; 58% female) with baseline total modified gastroduodenal Lanza score ≤ 2 (no erosions/ulcers) on endoscopy. The primary endpoint was mean total modified gastroduodenal Lanza score on day 7. A secondary endpoint was incidence of gastric ulcers. RESULTS The day 7 mean total modified gastroduodenal Lanza score was 2.8 ± 1.7 for naproxen etemesil vs. 3.5 ± 2.0 for naproxen (P = 0.03), and significantly fewer naproxen etemesil-treated subjects (3.3%) developed gastric ulcers compared with naproxen-treated subjects (15.8%) (P = 0.02). CONCLUSION In this first proof-of-concept study, naproxen etemesil was associated with significantly lower gastroduodenal mucosal injury compared with naproxen after 7 days of exposure ( CLINICAL TRIAL NUMBER NCT00750243).
Collapse
Affiliation(s)
- J L Goldstein
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhou Y, Dial EJ, Doyen R, Lichtenberger LM. Effect of indomethacin on bile acid-phospholipid interactions: implication for small intestinal injury induced by nonsteroidal anti-inflammatory drugs. Am J Physiol Gastrointest Liver Physiol 2010; 298:G722-31. [PMID: 20203063 PMCID: PMC2867422 DOI: 10.1152/ajpgi.00387.2009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 02/24/2010] [Indexed: 01/31/2023]
Abstract
The injurious effect of nonsteroidal anti-inflammatory drugs (NSAIDs) in the small intestine was not appreciated until the widespread use of capsule endoscopy. Animal studies found that NSAID-induced small intestinal injury depends on the ability of these drugs to be secreted into the bile. Because the individual toxicity of amphiphilic bile acids and NSAIDs directly correlates with their interactions with phospholipid membranes, we propose that the presence of both NSAIDs and bile acids alters their individual physicochemical properties and enhances the disruptive effect on cell membranes and overall cytotoxicity. We utilized in vitro gastric AGS and intestinal IEC-6 cells and found that combinations of bile acid, deoxycholic acid (DC), taurodeoxycholic acid, glycodeoxycholic acid, and the NSAID indomethacin (Indo) significantly increased cell plasma membrane permeability and became more cytotoxic than these agents alone. We confirmed this finding by measuring liposome permeability and intramembrane packing in synthetic model membranes exposed to DC, Indo, or combinations of both agents. By measuring physicochemical parameters, such as fluorescence resonance energy transfer and membrane surface charge, we found that Indo associated with phosphatidylcholine and promoted the molecular aggregation of DC and potential formation of larger and isolated bile acid complexes within either biomembranes or bile acid-lipid mixed micelles, which leads to membrane disruption. In this study, we demonstrated increased cytotoxicity of combinations of bile acid and NSAID and provided a molecular mechanism for the observed toxicity. This mechanism potentially contributes to the NSAID-induced injury in the small bowel.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Pediatrics-Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | | | | | | |
Collapse
|
28
|
Ibuprofen and chronic pyloric stricture. J Pediatr Gastroenterol Nutr 2010; 50:222-4. [PMID: 19668008 PMCID: PMC3816498 DOI: 10.1097/mpg.0b013e3181a1c45e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
29
|
Marchbank T, Weaver G, Nilsen-Hamilton M, Playford RJ. Pancreatic secretory trypsin inhibitor is a major motogenic and protective factor in human breast milk. Am J Physiol Gastrointest Liver Physiol 2009; 296:G697-703. [PMID: 19147803 DOI: 10.1152/ajpgi.90565.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Colostrum is the first milk produced after birth and is rich in immunoglobulins and bioactive molecules. We examined whether human colostrum and milk contained pancreatic secretory trypsin inhibitor (PSTI), a peptide of potential relevance for mucosal defense and, using in vitro and in vivo models, determined whether its presence influenced gut integrity and repair. Human milk was collected from individuals over various times from parturition and PSTI concentrations determined with the use of immunoassay. Human milk samples were analyzed for proliferation and promigratory activity (wounded monolayers) and antiapoptotic activity (caspase-3 activity) with the use of intestinal HT29 cells with or without neutralizing antibodies to PSTI and epidermal growth factor (EGF). Rats were restrained and given indomethacin to induce gastric injury. Effect of gavage with human breast milk with or without neutralizing antibodies on amount of injury were compared with animals receiving a commercial formula feed. PSTI is secreted into human milk, with colostrum containing a much higher concentration of PSTI than human milk obtained later. Human milk stimulated migration and proliferation about threefold and reduced indomethacin-induced apoptosis by about 70-80%. Sixty-five percent of the migratory effect of human milk could be removed by immunoneutralization of PSTI. PSTI worked synergistically with EGF in mediating these effects. Gastric damage in rats was reduced by about 75% in the presence of human milk and was more efficacious than the formula feed (P<0.001). Protective effects of the milk were reduced by about 60% by PSTI immunoneutralization. We concluded that PSTI is secreted into human milk at concentrations that have probable pathophysiological relevance.
Collapse
Affiliation(s)
- Tania Marchbank
- Centre for Gastroenterology, Institute of Cell and Molecular Science, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, and Queen Charlotte's Hospital, Turner St., London E1 2AD, UK
| | | | | | | |
Collapse
|
30
|
Marchbank T, Elia G, Playford RJ. Intestinal protective effect of a commercial fish protein hydrolysate preparation. ACTA ACUST UNITED AC 2009; 155:105-9. [PMID: 19323982 DOI: 10.1016/j.regpep.2009.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/08/2009] [Accepted: 02/03/2009] [Indexed: 12/15/2022]
Abstract
OBJECTIVES A partially hydrolysed, dried, product of pacific whiting fish is marketed as a health food supplement supporting 'intestinal health'. Scientific data supporting these claims are severely limited. We, therefore, examined if it influenced intestinal injury caused by the NSAID, indomethacin. METHODS Effects of fish hydrolysate on proliferation ([3H]-thymidine) and indomethacin-induced apoptosis (active caspase-3-immunostaining) utilised HT29 cells. In vivo studies used mice (n=8/group). 4/6 groups had fish hydrolysate (25 or 50 mg/ml) supplemented to their drinking water for 7 days. All mice received indomethacin (85 mg/kg subcutaneously) or placebo, 12 h before killing. Small intestinal injury was assessed using morphometry and morphology, proliferation (crypt BrdU labelling ) and apoptosis (active caspase-3 immunostaining). RESULTS Fish hydrolysate stimulated proliferation of HT29 cells. Apoptosis increased 3-fold following incubation with indomethacin but co-presence of fish hydrolysate truncated this effect by 40% (p<0.01). In mice, fish hydrolysate reduced the villus damaging effects of indomethacin by 60% (p<0.05). Indomethacin increased intestinal proliferation by 65%, irrespective of presence of hydrolysate. In contrast, intestinal caspase-3 activity increased by 83% in animals given indomethacin but this rise was truncated by 70% by co-presence of hydrolysate (p<0.01). CONCLUSION This natural bioactive product reduced apoptosis and the gut damaging effects of indomethacin.
Collapse
Affiliation(s)
- Tania Marchbank
- Centre for Gastroenterology, Institute of Cell and Molecular Science, Barts and The London School of Medicine, Queen Mary University of London, UK
| | | | | |
Collapse
|
31
|
Blandizzi C, Tuccori M, Colucci R, Fornai M, Antonioli L, Ghisu N, Del Tacca M. Role of coxibs in the strategies for gastrointestinal protection in patients requiring chronic non-steroidal anti-inflammatory therapy. Pharmacol Res 2008; 59:90-100. [PMID: 19073262 DOI: 10.1016/j.phrs.2008.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 12/16/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly prescribed drugs due to their high efficacy in the treatment of pain, fever, inflammation and rheumatic disorders. However, their use is associated with the occurrence of adverse effects at the level of digestive tract, ranging from dyspeptic symptoms, gastrointestinal erosions and peptic ulcers to more serious complications, such as overt bleeding or perforation. To overcome problems related to NSAID-induced digestive toxicity, different therapeutic strategies can presently be considered, including the co-administration of drugs endowed with protective activity on the upper gastrointestinal tract, such as the proton pump inhibitors, or the prescription of coxibs, which have been clinically developed as anti-inflammatory/analgesic drugs characterized by reduced damaging activity on gastrointestinal mucosa. The availability of different treatment options, to reduce the risk of NSAID-induced adverse digestive effects, has fostered intensive preclinical and clinical research aimed at addressing a number of unresolved issues and to establish rational criteria for an appropriate use of coxibs in the medical practice. Particular attention is being paid to the management of patients with high degrees of digestive risk, resulting by concomitant treatment with low-dose aspirin for anti-thrombotic prophylaxis or ongoing symptomatic gastroduodenal ulcers. The present review discusses the most relevant lines of evidence concerning the position of coxibs in the therapeutic strategies for gastrointestinal protection in patients who require NSAID therapy and hold different levels of risk of developing adverse effects at the level of digestive tract.
Collapse
Affiliation(s)
- Corrado Blandizzi
- Division of Pharmacology and Chemotherapy, Department of Internal Medicine, University of Pisa, Via Roma 55, Pisa 56126, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
Marchbank T, Limdi JK, Mahmood A, Elia G, Playford RJ. Clinical trial: protective effect of a commercial fish protein hydrolysate against indomethacin (NSAID)-induced small intestinal injury. Aliment Pharmacol Ther 2008; 28:799-804. [PMID: 19145735 DOI: 10.1111/j.1365-2036.2008.03783.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND A partially hydrolysed and dried product of pacific whiting fish is marketed as a health food supplement supporting 'intestinal health'. AIM To examine whether the partially hydrolysed and dried product of pacific whiting fish influenced the small intestinal damaging side effects of the nonsteroidal anti-inflammatory drug, indomethacin. METHODS Eight human volunteers completed a double-blind, placebo-controlled, crossover protocol of clinically relevant dose of indomethacin (50 mg t.d.s. p.o. for 5 days) with 7 days of fish hydrolysate or placebo starting 2 days prior to indomethacin. Changes in gut permeability were assessed using 5 h urinary lactulose:rhamnose (L/R) ratios. RESULTS Fish hydrolysate given alone did not affect permeability. In the main study (n = 8), baseline values were similar for both arms (0.28 +/- 0.05 and 0.35 +/- 0.07). Administration of indomethacin (+placebo) caused a fivefold rise in L/R ratios (increasing to 1.54 +/- 0.35), whereas L/R ratios in the same subjects ingesting indomethacin + fish hydrolysate was only 0.59 +/- 0.14 (P < 0.01 vs. indomethacin alone). Dyspeptic symptoms occurred in four of eight subjects taking indomethacin alone, but zero of eight when hydrolysate was co-administered. CONCLUSION Natural bioactive products (nutriceuticals), such as fish hydrolysates, may provide a novel approach to the prevention and treatment of NSAID-induced and other gastrointestinal injurious conditions.
Collapse
Affiliation(s)
- T Marchbank
- Centre for Gastroenterology, Institute of Cell and Molecular Science, Barts & The London School of Medicine, Queen Mary University of London, London, UK
| | | | | | | | | |
Collapse
|
33
|
Dial EJ, Darling RL, Lichtenberger LM. Importance of biliary excretion of indomethacin in gastrointestinal and hepatic injury. J Gastroenterol Hepatol 2008; 23:e384-9. [PMID: 18086111 DOI: 10.1111/j.1440-1746.2007.05266.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS A mechanism for protection of gastrointestinal (GI) and hepatic cells from damaging detergent actions of bile acids appears to involve the bile component, phosphatidylcholine (PC). Non-steroidal anti-inflammatory drugs (NSAIDs) induce intestinal injury in direct proportion to their ability to be excreted into bile, and are known to chemically associate with PC. We investigated the role of bile acids and PC in the mechanism of indomethacin-induced epithelial injury. METHODS Rats were injected orally or intravenously with radiolabeled indomethacin and their bile was collected over time for determination of NSAID secretion. Bile from rats treated with or without indomethacin was used in studies of red blood cell (RBC) hemolysis as a measure of membrane cytotoxicity. The bile salt, sodium deoxycholate (SDC), and indomethacin were tested alone and in combination with PC on RBC and on hepatic HepG2 cells. RESULTS Intravenously or orally given indomethacin was quantitatively excreted (approximately 50%) into bile over a 2-h study period. Bile from a rat treated with indomethacin or bile with exogenous indomethacin was cytotoxic to RBC, and the injury was prevented by the addition of PC. Hepatocytes exposed to SDC showed injury that could be dose-dependently prevented by PC, and reversed by indomethacin. CONCLUSIONS Biliary PC plays an important physiological role in protecting GI and hepatic epithelia from the cytotoxic actions of bile salts. The ability of NSAIDs excreted into the bile to associate with mixed bile salt micelles and reduce the protective action of the PC may be a critical component in the drugs' pathogenic mechanism.
Collapse
Affiliation(s)
- Elizabeth J Dial
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
34
|
An assessment of the interactions between diclofenac sodium and ammonio methacrylate copolymer using thermal analysis and Raman spectroscopy. J Pharm Biomed Anal 2008; 46:288-94. [DOI: 10.1016/j.jpba.2007.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 10/04/2007] [Accepted: 10/06/2007] [Indexed: 11/18/2022]
|
35
|
Abstract
PURPOSE OF REVIEW The gastroduodenum resists mucosal injury despite continuous exposure to concentrated gastric acid. The mucosal barrier consists of a preepithelial mucus HCO3- layer, intercellular tight junctions connecting the epithelial cells, and submucosal acid sensors, prostaglandins, cytokines, enteric nerves and blood flow. In the past year, study of these defensive mechanisms has revealed new insight into the observed sex differences in ulcer prevalence, the protective role of transforming growth factor, the role of serotonin in regulating HCO3- secretion, the role of mechanisms in ulcer healing, the interaction of trefoil factors with the mucus gel, the interaction of glucocorticoids with cyclooxygenase and the characterization of novel, mucosal sparing antiinflammatory agents. RECENT FINDINGS Transforming growth factor, melatonin, serotonin, trefoil factors and H2S all enhance mucosal barrier function or accelerate ulcer healing. Newer coxibs may have safety and advantages over existing compounds. Existing nonsteroidal antiinflammatory drugs may be safer than originally thought. SUMMARY The continued elucidation of basic defense mechanisms has led to the development of several new compounds designed to enhance barrier function and repair mechanisms.
Collapse
Affiliation(s)
- Maggie Ham
- The David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | |
Collapse
|