1
|
Yamada J, Fukui T, Yatani A, Mimura C, Fukuda K, Hazama D, Katsurada N, Nagano T, Yamamoto M, Tachihara M. Impact of concurrent medications on the outcome of immunotherapy in non-small cell lung carcinoma. Thorac Cancer 2024; 15:1228-1236. [PMID: 38622898 PMCID: PMC11128370 DOI: 10.1111/1759-7714.15294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND There have been reports on the impact of concurrent drugs on the outcome of immunotherapy for non-small cell lung carcinoma (NSCLC). However, the effect of some drugs, such as antibiotics and nonsteroidal anti-inflammatory drugs (NSAIDs), has not been clarified in patients with NSCLC. In the present study, we aimed to assess the association between concurrent drugs and the outcomes of immune checkpoint inhibitors (ICIs) alone or in combination with chemotherapy for patients with advanced NSCLC. METHODS We retrospectively assessed patients with advanced NSCLC who underwent ICI treatment between September 2017 and December 2021 at Kobe University Hospital. We evaluated the data regarding the use of antibiotics within 30 days before ICI initiation, as well as the use of proton pump inhibitors (PPIs) and NSAIDs during ICI initiation. RESULTS A total of 127 patients were assessed, among whom 28 (22.0%) patients received antibiotics, 39 (30.7%) PPIs, and 36 (28.3%) NSAIDs. No significant differences were observed between the patients with and without antibiotic use. However, patients using NSAIDs had significantly worse objective response rates (ORR) and progression-free survival (PFS) with ICI alone or in combination with chemotherapy compared to those who did not (ORR, 47.2% vs. 67.0%; p = 0.045. PFS, 6.3 months vs. 10.8 months; p = 0.02). Patients using PPIs demonstrated a worse ORR of ICI in combination with chemotherapy compared to those who did not (ORR, 45.2% vs. 72.6%; p = 0.013). CONCLUSIONS The unnecessary use of NSAIDs along with immunotherapy should be discouraged.
Collapse
Affiliation(s)
- Jun Yamada
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Takafumi Fukui
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Atsuhiko Yatani
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Chihiro Mimura
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Kiyoko Fukuda
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Naoko Katsurada
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
2
|
Whitfield-Cargile CM, Chung HC, Coleman MC, Cohen ND, Chamoun-Emanuelli AM, Ivanov I, Goldsby JS, Davidson LA, Gaynanova I, Ni Y, Chapkin RS. Integrated analysis of gut metabolome, microbiome, and exfoliome data in an equine model of intestinal injury. MICROBIOME 2024; 12:74. [PMID: 38622632 PMCID: PMC11017594 DOI: 10.1186/s40168-024-01785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND The equine gastrointestinal (GI) microbiome has been described in the context of various diseases. The observed changes, however, have not been linked to host function and therefore it remains unclear how specific changes in the microbiome alter cellular and molecular pathways within the GI tract. Further, non-invasive techniques to examine the host gene expression profile of the GI mucosa have been described in horses but not evaluated in response to interventions. Therefore, the objectives of our study were to (1) profile gene expression and metabolomic changes in an equine model of non-steroidal anti-inflammatory drug (NSAID)-induced intestinal inflammation and (2) apply computational data integration methods to examine host-microbiota interactions. METHODS Twenty horses were randomly assigned to 1 of 2 groups (n = 10): control (placebo paste) or NSAID (phenylbutazone 4.4 mg/kg orally once daily for 9 days). Fecal samples were collected on days 0 and 10 and analyzed with respect to microbiota (16S rDNA gene sequencing), metabolomic (untargeted metabolites), and host exfoliated cell transcriptomic (exfoliome) changes. Data were analyzed and integrated using a variety of computational techniques, and underlying regulatory mechanisms were inferred from features that were commonly identified by all computational approaches. RESULTS Phenylbutazone induced alterations in the microbiota, metabolome, and host transcriptome. Data integration identified correlation of specific bacterial genera with expression of several genes and metabolites that were linked to oxidative stress. Concomitant microbiota and metabolite changes resulted in the initiation of endoplasmic reticulum stress and unfolded protein response within the intestinal mucosa. CONCLUSIONS Results of integrative analysis identified an important role for oxidative stress, and subsequent cell signaling responses, in a large animal model of GI inflammation. The computational approaches for combining non-invasive platforms for unbiased assessment of host GI responses (e.g., exfoliomics) with metabolomic and microbiota changes have broad application for the field of gastroenterology. Video Abstract.
Collapse
Affiliation(s)
- C M Whitfield-Cargile
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| | - H C Chung
- Department of Statistics, College of Arts & Sciences, Texas A&M University, College Station, TX, USA
- Mathematics & Statistics Department, College of Science, University of North Carolina Charlotte, Charlotte, NC, USA
| | - M C Coleman
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - N D Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - A M Chamoun-Emanuelli
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - I Ivanov
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - J S Goldsby
- Program in Integrative Nutrition & Complex Diseases, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - L A Davidson
- Program in Integrative Nutrition & Complex Diseases, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - I Gaynanova
- Department of Statistics, College of Arts & Sciences, Texas A&M University, College Station, TX, USA
| | - Y Ni
- Department of Statistics, College of Arts & Sciences, Texas A&M University, College Station, TX, USA
| | - R S Chapkin
- Program in Integrative Nutrition & Complex Diseases, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
3
|
Vitória Minzoni de Souza Iacia M, Eduarda Ferraz Mendes M, Cristiny de Oliveira Vieira K, Cristine Marques Ruiz G, José Leopoldo Constantino C, da Silva Martin C, Eloizo Job A, Alborghetti Nai G, Kretli Winkelstroter Eller L. Evaluation of curcumin nanoemulsion effect to prevent intestinal damage. Int J Pharm 2024; 650:123683. [PMID: 38092264 DOI: 10.1016/j.ijpharm.2023.123683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
Curcumin has gained great prominence for the prevention and treatment of inflammatory bowel disease. However, studies have reported the low bioavailability of orally administered curcumin. This work aimed to evaluate the characteristics, stability and effects of a curcumin-carrying nanoemulsion in preventing intestinal damage induced by indomethacin. Nanoemulsions containing curcumin were prepared by spontaneous emulsification method and it was characterized by dynamic light scattering (DLS), zeta potential and the morphology was evaluated by scanning electron microscopy (SEM). Its stability was tested under different conditions of pH, temperature at 0, 7, 14, 21 and 28 days. In animal experimentation, 36 male mice of the Mus musculus lineage (C57BL/6) were used. The intestinal inflammation was evaluated based on macroscopic, histopathological and metagenomic analysis. It was found a stable nanoemulsion with a size of 409.8 nm, polydispersion index (PDI) of 0.132 and zeta potential of -18.8 mV. However, these lost charge in pH2, showing instability in acidic media (p < 0.05). In animal experiments, the nanoemulsion did not significantly improve intestinal inflammation. However, the group treated with curcumin nanoemulsion showed a higher relative abundance of the genus Lactobacillus (p < 0.05). In conclusion, the curcumin nanoemulsion was relevant in the modulation of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | | | - Gilia Cristine Marques Ruiz
- Department of Physics, Faculty of Science and Technology, Universidade Estadual Paulista, FCT/UNESP, Presidente Prudente, SP, Brazil
| | - Carlos José Leopoldo Constantino
- Department of Physics, Faculty of Science and Technology, Universidade Estadual Paulista, FCT/UNESP, Presidente Prudente, SP, Brazil
| | - Cibely da Silva Martin
- Department of Physics, Faculty of Science and Technology, Universidade Estadual Paulista, FCT/UNESP, Presidente Prudente, SP, Brazil
| | - Aldo Eloizo Job
- Department of Physics, Faculty of Science and Technology, Universidade Estadual Paulista, FCT/UNESP, Presidente Prudente, SP, Brazil
| | - Gisele Alborghetti Nai
- Master's in Health Sciences - Universidade do Oeste Paulista/UNOESTE, Presidente Prudente, SP, Brazil; Program of Animal Science - Universidade do Oeste Paulista/UNOESTE, Presidente Prudente, Brazil
| | - Lizziane Kretli Winkelstroter Eller
- Faculty of Health Sciences - Universidade do Oeste Paulista/UNOESTE, Presidente Prudente, SP, Brazil; Master's in Health Sciences - Universidade do Oeste Paulista/UNOESTE, Presidente Prudente, SP, Brazil; Program of Animal Science - Universidade do Oeste Paulista/UNOESTE, Presidente Prudente, Brazil.
| |
Collapse
|
4
|
Tian L, Huang C, Fu W, Gao L, Mi N, Bai M, Ma H, Zhang C, Lu Y, Zhao J, Zhang X, Jiang N, Lin Y, Yue P, Yuan J, Meng W. Proton pump inhibitors may enhance the risk of digestive diseases by regulating intestinal microbiota. Front Pharmacol 2023; 14:1217306. [PMID: 37529701 PMCID: PMC10387554 DOI: 10.3389/fphar.2023.1217306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Proton pump inhibitors (PPIs) are the most used acid-inhibitory drugs, with a wide range of applications in the treatment of various digestive diseases. However, recently, there has been a growing number of digestive complications linked to PPIs, and several studies have indicated that the intestinal flora play an important role in these complications. Therefore, developing a greater understanding of the role of the gut microbiota in PPI-related digestive diseases is essential. Here, we summarize the current research on the correlation between PPI-related digestive disorders and intestinal flora and establish the altered strains and possible pathogenic mechanisms of the different diseases. We aimed to provide a theoretical basis and reference for the future treatment and prevention of PPI-related digestive complications based on the regulation of the intestinal microbiota.
Collapse
Affiliation(s)
- Liang Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Chongfei Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wenkang Fu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Long Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ningning Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Mingzhen Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Haidong Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Chao Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yawen Lu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jinyu Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xianzhuo Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ningzu Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yanyan Lin
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ping Yue
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jinqiu Yuan
- Clinical Research Center, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wenbo Meng
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Ohkusa T, Nishikawa Y, Sato N. Gastrointestinal disorders and intestinal bacteria: Advances in research and applications in therapy. Front Med (Lausanne) 2023; 9:935676. [PMID: 36825261 PMCID: PMC9941163 DOI: 10.3389/fmed.2022.935676] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 12/27/2022] [Indexed: 02/09/2023] Open
Abstract
Intestinal bacteria coexist with humans and play a role in suppressing the invasion of pathogens, producing short-chain fatty acids, producing vitamins, and controlling the immune system. Studies have been carried out on culturable bacterial species using bacterial culture methods for many years. However, as metagenomic analysis of bacterial genes has been developed since the 1990s, it has recently revealed that many bacteria in the intestine cannot be cultured and that approximately 1,000 species and 40 trillion bacteria are present in the gut microbiota. Furthermore, the composition of the microbiota is different in each disease state compared with the healthy state, and dysbiosis has received much attention as a cause of various diseases. Regarding gastrointestinal diseases, dysbiosis has been reported to be involved in inflammatory bowel disease, irritable bowel syndrome, and non-alcoholic steatohepatitis. Recent findings have also suggested that dysbiosis is involved in colon cancer, liver cancer, pancreatic cancer, esophageal cancer, and so on. This review focuses on the relationship between the gut microbiota and gastrointestinal/hepatobiliary diseases and also discusses new therapies targeting the gut microbiota.
Collapse
Affiliation(s)
| | - Yuriko Nishikawa
- Department of Microbiota Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuhiro Sato
- Department of Microbiota Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Zádori ZS, Király K, Al-Khrasani M, Gyires K. Interactions between NSAIDs, opioids and the gut microbiota - Future perspectives in the management of inflammation and pain. Pharmacol Ther 2023; 241:108327. [PMID: 36473615 DOI: 10.1016/j.pharmthera.2022.108327] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The composition of intestinal microbiota is influenced by a number of factors, including medications, which may have a substantial impact on host physiology. Nonsteroidal anti-inflammatory drugs (NSAIDs) and opioid analgesics are among those widely used medications that have been shown to alter microbiota composition in both animals and humans. Although much effort has been devoted to identify microbiota signatures associated with these medications, much less is known about the underlying mechanisms. Mucosal inflammation, changes in intestinal motility, luminal pH and bile acid metabolism, or direct drug-induced inhibitory effect on bacterial growth are all potential contributors to NSAID- and opioid-induced dysbiosis, however, only a few studies have addressed directly these issues. In addition, there is a notable overlap between the microbiota signatures of these drugs and certain diseases in which they are used, such as spondyloarthritis (SpA), rheumatoid arthritis (RA) and neuropathic pain associated with type 2 diabetes (T2D). The aims of the present review are threefold. First, we aim to provide a comprehensive up-to-date summary on the bacterial alterations caused by NSAIDs and opioids. Second, we critically review the available data on the possible underlying mechanisms of dysbiosis. Third, we review the current knowledge on gut dysbiosis associated with SpA, RA and neuropathic pain in T2D, and highlight the similarities between them and those caused by NSAIDs and opioids. We posit that drug-induced dysbiosis may contribute to the persistence of these diseases, and may potentially limit the therapeutic effect of these medications by long-term use. In this context, we will review the available literature data on the effect of probiotic supplementation and fecal microbiota transplantation on the therapeutic efficacy of NSAIDs and opioids in these diseases.
Collapse
Affiliation(s)
- Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, 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
| | - Klára Gyires
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
The Role of Probiotic Bacillus Spores and Amino Acids with Immunoglobulins on a Rat Enteropathy Model. Biomedicines 2022; 10:biomedicines10102508. [PMID: 36289770 PMCID: PMC9599762 DOI: 10.3390/biomedicines10102508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are some of the most widely used drugs due to their anti-inflammatory, analgesic and antipyretic pharmacological effects. Gastrointestinal side effects are some of the most severe and frequent side effects of NSAIDs. These depend on the balance of the gut microbiome, the abundance of Gram-negative bacteria, and the amount of lipopolysaccharide released. Therefore, restoring or improving gut bacteria balance with probiotic supplements could prove to be an adjuvant therapy against mild NSAID-induced enteropathy. Twenty-five Wistar albino male rats were divided into five groups. The negative control group was administered carboxymethylcellulose and the positive control group diclofenac (DIC), 8 mg/kg for 7 days, which represented the enteropathy model. Treatment groups consisted of a combination of pro-biotic spores (MSB), amino acids and immunoglobulins supplement (MM), which were also administered for 7 days. We analyzed hepatic injury markers (AST, ALT) and creatinine, and inflammatory markers, IL-6, TNF-α, PGE2, iNOS, as well as total antioxidant capacity. The results obtained in the present study suggest that the modulation of the intestinal microbiota by administration of probiotics (Bacillus spores), alone or in combination with immunoglobulins and amino acids, represents an attractive therapy for the prevention of NSAID-induced enteropathy.
Collapse
|
8
|
Effects of Canine-Obtained Lactic-Acid Bacteria on the Fecal Microbiota and Inflammatory Markers in Dogs Receiving Non-Steroidal Anti-Inflammatory Treatment. Animals (Basel) 2022; 12:ani12192519. [PMID: 36230259 PMCID: PMC9558503 DOI: 10.3390/ani12192519] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 11/17/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) may cause enteropathy in dogs and probiotics may be one option to prevent this. The objective of this study was to determine whether the administration of canine-obtained lactic acid bacteria (LAB) has an effect on the frequency of diarrhea, the composition of the fecal microbiota, and/or markers of gastrointestinal inflammation in dogs receiving NSAIDs when compared to dogs given NSAIDs and a placebo. A total of 22 dogs treated with NSAIDs for various clinical indications were enrolled in a seven-day randomized, double-blinded placebo-controlled interventional study. Dogs were randomized to receive either placebo or LAB, a product containing Limosilactobacillus fermentum, Lacticaseibacillus rhamnosus, and Lactiplantibacillus plantarum. Fecal samples were collected on days one and seven. The fecal microbiota was evaluated using the fecal dysbiosis index (DI) and individual bacterial taxa. Fecal calprotectin (CP) and S100A12/Calgranulin C concentrations were used as markers of gastrointestinal inflammation. There was a difference in frequency of diarrhea between groups, with it affecting 4/12 dogs (33%) in the placebo group and 1/10 dogs (10%) in the LAB group, but this difference did not reach statistical significance (p = 0.32). There was a correlation between S100A12 and CP (p < 0.001), and Clostridium perfringens correlated with S100A12 (p < 0.015). Neither treatment significantly affected S100A12 (p = 0.37), CP (p = 0.12), or fecal DI (p = 0.65). This study suggests that LAB is a safe supplement to use for short-term treatment in NSAID-treated dogs, but further studies are needed to determine its potential to prevent NSAID-induced enteropathy in dogs.
Collapse
|
9
|
Bodnarova T, Hall E, Duplan F. Prescribing habits for the use of omeprazole as a gastroprotectant in dogs in a veterinary teaching hospital. J Small Anim Pract 2022; 63:769-775. [PMID: 35840131 DOI: 10.1111/jsap.13531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/24/2022] [Accepted: 06/14/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To examine the prescribing habits for omeprazole in a veterinary teaching hospital and to evaluate the effect of a clinical audit on omeprazole prescription. MATERIAL AND METHODS Observational study with retrospective clinical audit followed by a prospective study. The evaluated data about omeprazole prescription included the dose, frequency and indication. These were assessed according to published guidelines. A seminar about the audit and current guidelines on the use of omeprazole was organised for all members of clinical staff. Prospective data collection was conducted after the seminar. A comparison of the collected data before and after the clinical audit was made. RESULTS A total of 301 dogs were prescribed omeprazole in the veterinary teaching hospital during the study period (including the retrospective and prospective parts). Complete data were acquired from 240 patients. The prescribed frequency of omeprazole was inappropriate in 23 (16.5%) of the prescriptions in the retrospective section but in only five (5.0%) in the prospective study. Inappropriate indications were reported in 12 (8.6%) patients in the retrospective section and in two patients (2.0%) in the prospective study. Overall inappropriate omeprazole prescription was identified in 34 (24.5%) patients in the retrospective part and in seven (6.9%) patients in the prospective part. There was a statistically significant difference between the two groups in frequency, indication and overall prescription of omeprazole. CLINICAL SIGNIFICANCE This study details the frequency of inappropriate prescription of omeprazole in a veterinary teaching hospital and provides some evidence that dissemination of guidelines based on clinical audit can improve prescribing habits.
Collapse
Affiliation(s)
- T Bodnarova
- Veterinary Clinic Podebrady, Podebrady, 29001, Czech Republic
| | - E Hall
- School of Veterinary Sciences, University of Bristol, Bristol, BS40 5DU, UK
| | - F Duplan
- School of Veterinary Sciences, University of Bristol, Bristol, BS40 5DU, UK
| |
Collapse
|
10
|
Zhang M, Xia F, Xia S, Zhou W, Zhang Y, Han X, Zhao K, Feng L, Dong R, Tian D, Yu Y, Liao J. NSAID-Associated Small Intestinal Injury: An Overview From Animal Model Development to Pathogenesis, Treatment, and Prevention. Front Pharmacol 2022; 13:818877. [PMID: 35222032 PMCID: PMC8864225 DOI: 10.3389/fphar.2022.818877] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
With the wide application of non-steroidal anti-inflammatory drugs (NSAIDs), their gastrointestinal side effects are an urgent health burden. There are currently sound preventive measures for upper gastrointestinal injury, however, there is a lack of effective defense against lower gastrointestinal damage. According to a large number of previous animal experiments, a variety of NSAIDs have been demonstrated to induce small intestinal mucosal injury in vivo. This article reviews the descriptive data on the administration dose, administration method, mucosal injury site, and morphological characteristics of inflammatory sites of various NSAIDs. The cells, cytokines, receptors and ligands, pathways, enzyme inhibition, bacteria, enterohepatic circulation, oxidative stress, and other potential pathogenic factors involved in NSAID-associated enteropathy are also reviewed. We point out the limitations of drug modeling at this stage and are also pleased to discover the application prospects of chemically modified NSAIDs, dietary therapy, and many natural products against intestinal mucosal injury.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Xia
- Department of Hepatic Surgery Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wangdong Zhou
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lina Feng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruonan Dong
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Induri SNR, Kansara P, Thomas SC, Xu F, Saxena D, Li X. The Gut Microbiome, Metformin, and Aging. Annu Rev Pharmacol Toxicol 2021; 62:85-108. [PMID: 34449247 DOI: 10.1146/annurev-pharmtox-051920-093829] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metformin has been extensively used for the treatment of type 2 diabetes, and it may also promote healthy aging. Despite its widespread use and versatility, metformin's mechanisms of action remain elusive. The gut typically harbors thousands of bacterial species, and as the concentration of metformin is much higher in the gut as compared to plasma, it is plausible that microbiome-drug-host interactions may influence the functions of metformin. Detrimental perturbations in the aging gut microbiome lead to the activation of the innate immune response concomitant with chronic low-grade inflammation. With the effectiveness of metformin in diabetes and antiaging varying among individuals, there is reason to believe that the gut microbiome plays a role in the efficacy of metformin. Metformin has been implicated in the promotion and maintenance of a healthy gut microbiome and reduces many age-related degenerative pathologies. Mechanistic understanding of metformin in the promotion of a healthy gut microbiome and aging will require a systems-level approach. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sri Nitya Reddy Induri
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Payalben Kansara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; .,Department of Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| |
Collapse
|
12
|
Tai FWD. How Capsule Endoscopy Has Illumened the Prevalence of NSAID-Related Distal Small Intestinal Ulceration. Dig Dis Sci 2021; 66:2481-2482. [PMID: 32949325 DOI: 10.1007/s10620-020-06604-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2020] [Indexed: 12/09/2022]
Affiliation(s)
- Foong Way David Tai
- Academic Department of Gastroenterology, Royal Hallamshire Hospital, Glossop Road, Sheffield, S10 2JF, UK.
| |
Collapse
|
13
|
The Role of H 2S in the Gastrointestinal Tract and Microbiota. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:67-98. [PMID: 34302689 DOI: 10.1007/978-981-16-0991-6_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathways and mechanisms of the production of H2S in the gastrointestinal tract are briefly described, including endogenous H2S produced by the organism and H2S from microorganisms in the gastrointestinal tract. In addition, the physiological regulatory functions of H2S on gastrointestinal motility, sensation, secretion and absorption, endocrine system, proliferation and differentiation of stem cells, and the possible mechanisms involved are introduced. In view of the complexity of biosynthesis, physiological roles, and the mechanism of H2S, this chapter focuses on the interactions and dynamic balance among H2S, gastrointestinal microorganisms, and the host. Finally, we focus on some clinical gastrointestinal diseases, such as inflammatory bowel disease, colorectal cancer, functional gastrointestinal disease, which might occur or develop when the above balance is broken. Pharmacological regulation of H2S or the intestinal microorganisms related to H2S might provide new therapeutic approaches for some gastrointestinal diseases.
Collapse
|
14
|
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: 0.7] [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
|
15
|
Lázár B, László SB, Hutka B, Tóth AS, Mohammadzadeh A, Berekméri E, Ágg B, Balogh M, Sajtos V, Király K, Al-Khrasani M, Földes A, Varga G, Makra N, Ostorházi E, Szabó D, Ligeti B, Kemény Á, Helyes Z, Ferdinandy P, Gyires K, Zádori ZS. A comprehensive time course and correlation analysis of indomethacin-induced inflammation, bile acid alterations and dysbiosis in the rat small intestine. Biochem Pharmacol 2021; 190:114590. [PMID: 33940029 DOI: 10.1016/j.bcp.2021.114590] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
It has been proposed that changes in microbiota due to nonsteroidal anti-inflammatory drugs (NSAIDs) alter the composition of bile, and elevation of hydrophobic secondary bile acids contributes to small intestinal damage. However, little is known about the effect of NSAIDs on small intestinal bile acids, and whether bile alterations correlate with mucosal injury and dysbiosis. Here we determined the ileal bile acid metabolome and microbiota 24, 48 and 72 h after indomethacin treatment, and their correlation with each other and with tissue damage in rats. In parallel with the development of inflammation, indomethacin increased the ileal proportion of glycine and taurine conjugated bile acids, but not bile hydrophobicity. Firmicutes decreased with time, whereas Gammaproteobacteria increased first, but declined later and were partially replaced by Bilophila, Bacteroides and Fusobacterium. Mucosal injury correlated negatively with unconjugated bile acids and Gram-positive bacteria, and positively with taurine conjugates and some Gram-negative taxa. Strong positive correlation was found between Lactobacillaceae, Ruminococcaceae, Clostridiaceae and unconjugated bile acids. Indomethacin-induced dysbiosis was not likely due to direct antibacterial effects or alterations in luminal pH. Here we provide the first detailed characterization of indomethacin-induced time-dependent alterations in small intestinal bile acid composition, and their associations with mucosal injury and dysbiosis. Our results suggest that increased bile hydrophobicity is not likely to contribute to indomethacin-induced small intestinal damage.
Collapse
Affiliation(s)
- Bernadette Lázár
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Szilvia B László
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Barbara Hutka
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - András S Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Amir Mohammadzadeh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Department of Ecology, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Mihály Balogh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Viktor Sajtos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Kornél Király
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Anna Földes
- Department of Oral Biology, Semmelweis University, 1089 Budapest, Hungary
| | - Gábor Varga
- Department of Oral Biology, Semmelweis University, 1089 Budapest, Hungary
| | - Nóra Makra
- Department of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Eszter Ostorházi
- Department of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Dóra Szabó
- Department of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Balázs Ligeti
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, 1083 Budapest, Hungary
| | - Ágnes Kemény
- Department of Medical Biology, University of Pécs, 7624 Pécs, Hungary; Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Klára Gyires
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltán S Zádori
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary.
| |
Collapse
|
16
|
Mäkelä SM, Forssten SD, Kailajärvi M, Langén VL, Scheinin M, Tiihonen K, Ouwehand AC. Effects of Bifidobacterium animalis ssp. lactis 420 on gastrointestinal inflammation induced by a nonsteroidal anti-inflammatory drug: A randomized, placebo-controlled, double-blind clinical trial. Br J Clin Pharmacol 2021; 87:4625-4635. [PMID: 33908058 PMCID: PMC9291844 DOI: 10.1111/bcp.14880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/26/2021] [Accepted: 04/18/2021] [Indexed: 12/16/2022] Open
Abstract
Aims Use of nonsteroidal anti‐inflammatory drugs (NSAIDs) can cause damage to the gastric and duodenal mucosa. Some probiotics have proven useful in ameliorating the harmful side‐effects of NSAIDs. Our aim was to evaluate whether oral administration of Bifidobacterium animalis ssp. lactis 420 (B420) can attenuate the increase of calprotectin excretion into faeces induced by intake of diclofenac sustained‐release tablets. Methods A double‐blind, parallel‐group, placebo‐controlled and randomized clinical study was performed in 50 healthy male and female volunteers aged 20–40 years, in Finland. Study participation consisted of 4 phases: run‐in, intervention with B420 or placebo, B420 or placebo + NSAID treatment, and follow‐up. The primary outcome was the concentration of calprotectin in faeces. Secondary outcomes were haemoglobin and microbial DNA in faeces and blood haemoglobin levels. Results Intake of diclofenac increased the faecal excretion of calprotectin in both groups. The observed increases were 48.19 ± 61.55 μg/g faeces (mean ± standard deviation) in the B420 group and 31.30 ± 39.56 μg/g in the placebo group (difference estimate 16.90; 95% confidence interval: −14.00, 47.77; P = .276). There were no significant differences between the treatment groups in changes of faecal or blood haemoglobin. Faecal B. lactis DNA was much more abundant in the B420 group compared to the placebo group (ANOVA estimate for treatment difference 0.85 × 109/g faeces; 95% confidence interval: 0.50 × 109, 1.21 × 109; P < .0001). Conclusions Short‐term administration of the probiotic B420 did not protect the healthy adult study participants from diclofenac‐induced gastrointestinal inflammation as determined by analysis of faecal calprotectin levels.
Collapse
Affiliation(s)
- Sanna M Mäkelä
- Danisco Sweeteners Oy, IFF Health & Biosciences, ,Sokeritehtaantie 20, Kantvik, FI-02460, Finland
| | - Sofia D Forssten
- Danisco Sweeteners Oy, IFF Health & Biosciences, ,Sokeritehtaantie 20, Kantvik, FI-02460, Finland
| | - Marita Kailajärvi
- Clinical Research Services Turku (CRST) Oy and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ville L Langén
- Clinical Research Services Turku (CRST) Oy and Institute of Biomedicine, University of Turku, Turku, Finland.,Division of Medicine, Turku University Hospital, Turku, Finland
| | - Mika Scheinin
- Clinical Research Services Turku (CRST) Oy and Institute of Biomedicine, University of Turku, Turku, Finland.,Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Kirsti Tiihonen
- Danisco Sweeteners Oy, IFF Health & Biosciences, ,Sokeritehtaantie 20, Kantvik, FI-02460, Finland
| | - Arthur C Ouwehand
- Danisco Sweeteners Oy, IFF Health & Biosciences, ,Sokeritehtaantie 20, Kantvik, FI-02460, Finland
| |
Collapse
|
17
|
Whitfield-Cargile CM, Coleman MC, Cohen ND, Chamoun-Emanuelli AM, DeSolis CN, Tetrault T, Sowinski R, Bradbery A, Much M. Effects of phenylbutazone alone or in combination with a nutritional therapeutic on gastric ulcers, intestinal permeability, and fecal microbiota in horses. J Vet Intern Med 2021; 35:1121-1130. [PMID: 33656183 PMCID: PMC7995434 DOI: 10.1111/jvim.16093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022] Open
Abstract
Background Gastrointestinal (GI) injury and dysbiosis are adverse events associated with nonsteroidal anti‐inflammatory drug (NSAID) use in horses. Phenylbutazone has been shown to alter GI barrier function both in vitro and ex vivo, but its effects on barrier function have not been assessed in vivo. In addition, the ability of nutritional therapeutics to prevent these changes is not known. Objective Our objectives were to determine whether (a) phenylbutazone affected barrier function in vivo and (b) if phenylbutazone‐induced GI injury could be ameliorated by the use of a nutritional therapeutic. Animals Thirty healthy horses were randomly assigned to 3 groups (n = 10 per group): control, phenylbutazone, or phenylbutazone plus nutritional therapeutic. Methods This study was conducted as a blinded, randomized block design. All horses were managed identically throughout the study period. Samples were collected throughout the study period to monitor fecal microbiota changes and gastric ulcers before and after treatment. Quantification of the bacterial 16S rRNA gene in blood was used as a marker of intestinal permeability. Results Phenylbutazone increased amounts of bacterial 16S rDNA in circulation 3.02‐fold (95% confidence interval [CI], 0.1.89‐4.17), increased gastric ulceration score by a mean of 1.1 grade (P = .02), and induced specific changes in the microbiota, including loss of Pseudobutyrivibrio of family Lachnospiraceae. These changes were attenuated by nutritional treatment. Conclusions and Clinical Importance Collectively, these findings suggest that phenylbutazone induces GI injury, including impaired barrier function, and that nutritional treatment could attenuate these changes.
Collapse
Affiliation(s)
- Canaan M Whitfield-Cargile
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Michelle C Coleman
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Noah D Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ana M Chamoun-Emanuelli
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Cristobal Navas DeSolis
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Taylor Tetrault
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ryan Sowinski
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Amanda Bradbery
- Department of Animal Science, College of Agriculture & Life Sciences Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Mattea Much
- Department of Animal Science, College of Agriculture & Life Sciences Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
18
|
Guo CG, Leung WK. Potential Strategies in the Prevention of Nonsteroidal Anti-inflammatory Drugs-Associated Adverse Effects in the Lower Gastrointestinal Tract. Gut Liver 2021; 14:179-189. [PMID: 31547642 PMCID: PMC7096237 DOI: 10.5009/gnl19201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022] Open
Abstract
With the increasing use of nonsteroidal anti-inflammatory drugs (NSAIDs), the incidence of lower gastrointestinal (GI) complications is expected to increase. However, unlike upper GI complications, the burden, pathogenesis, prevention and treatment of NSAID-associated lower GI complications remain unclear. To date, no cost-effective and safe protective agent has been developed that can completely prevent or treat NSAID-related lower GI injuries. Selective COX-2 inhibitors, misoprostol, intestinal microbiota modulation, and some mucoprotective agents have been reported to show protective effects on NSAID-induced lower GI injuries. This review aims to provide an overview of the current evidence on the prevention of NSAID-related lower GI injuries.
Collapse
Affiliation(s)
- Chuan-Guo Guo
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Wai K Leung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
19
|
Wang X, Tang Q, Hou H, Zhang W, Li M, Chen D, Gu Y, Wang B, Hou J, Liu Y, Cao H. Gut Microbiota in NSAID Enteropathy: New Insights From Inside. Front Cell Infect Microbiol 2021; 11:679396. [PMID: 34295835 PMCID: PMC8290187 DOI: 10.3389/fcimb.2021.679396] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022] Open
Abstract
As a class of the commonly used drugs in clinical practice, non-steroidal anti-inflammatory drugs (NSAIDs) can cause a series of adverse events including gastrointestinal injuries. Besides upper gastrointestinal injuries, NSAID enteropathy also attracts attention with the introduction of capsule endoscopy and double balloon enteroscopy. However, the pathogenesis of NSAID enteropathy remains to be entirely clarified. Growing evidence from basic and clinical studies presents that gut microbiota is a critical factor in NSAID enteropathy progress. We have reviewed the recent data about the interplay between gut microbiota dysbiosis and NSAID enteropathy. The chronic medication of NSAIDs could change the composition of the intestinal bacteria and aggravate bile acids cytotoxicity. Meanwhile, NSAIDs impair the intestinal barrier by inhibiting cyclooxygenase and destroying mitochondria. Subsequently, intestinal bacteria translocate into the mucosa, and then lipopolysaccharide released from gut microbiota combines to Toll-like receptor 4 and induce excessive production of nitric oxide and pro-inflammatory cytokines. Intestinal injuries present in the condition of intestinal inflammation and oxidative stress. In this paper, we also have reviewed the possible strategies of regulating gut microbiota for the management of NSAID enteropathy, including antibiotics, probiotics, prebiotics, mucosal protective agents, and fecal microbiota transplant, and we emphasized the adverse effects of proton pump inhibitors on NSAID enteropathy. Therefore, this review will provide new insights into a better understanding of gut microbiota in NSAID enteropathy.
Collapse
Affiliation(s)
- Xianglu Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Qiang Tang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Huiqin Hou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Wanru Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Danfeng Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
- *Correspondence: Hailong Cao, ; Jingli Hou, ; Yangping Liu,
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
- *Correspondence: Hailong Cao, ; Jingli Hou, ; Yangping Liu,
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- *Correspondence: Hailong Cao, ; Jingli Hou, ; Yangping Liu,
| |
Collapse
|
20
|
Jones SM, Gaier A, Enomoto H, Ishii P, Pilla R, Price J, Suchodolski J, Steiner JM, Papich MG, Messenger K, Tolbert MK. The effect of combined carprofen and omeprazole administration on gastrointestinal permeability and inflammation in dogs. J Vet Intern Med 2020; 34:1886-1893. [PMID: 32893926 PMCID: PMC7517840 DOI: 10.1111/jvim.15897] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background Proton pump inhibitors (eg, omeprazole) commonly are administered concurrently with nonsteroidal anti‐inflammatory drugs (NSAIDs; eg, carprofen) as prophylaxis to decrease the risk of gastrointestinal (GI) injury. However, evidence to support this practice is weak, and it might exacerbate dysbiosis and inflammation. Hypothesis/Objectives To evaluate the effect of carprofen alone or combined with omeprazole in dogs. We hypothesized that coadministration of omeprazole and carprofen would significantly increase GI permeability and dysbiosis index (DI) compared to no treatment or carprofen alone. Animals Six healthy adult colony beagle dogs. Methods Gastrointestinal permeability and inflammation were assessed by serum lipopolysaccharide (LPS) concentration, plasma iohexol concentration, fecal DI, and fecal calprotectin concentration in a prospective, 3‐period design. In the first 7‐day period, dogs received no intervention (baseline). During the 2nd period, dogs received 4 mg/kg of carprofen q24h PO for 7 days. In the 3rd period, dogs received 4 mg/kg of carprofen q24h and 1 mg/kg of omeprazole q12h PO for 7 days. Gastrointestinal permeability testing was performed at the end of each period. Data were analyzed using repeated measures mixed model analysis of variance with Tukey‐Kramer post hoc tests (P < .05). Results Serum LPS and plasma iohexol concentrations did not differ between treatments. Fecal calprotectin concentrations differed between treatments (P = .03). The DI varied over time based on the treatment received (P = .03). Coadministration of omeprazole and carprofen significantly increased fecal calprotectin concentration and DI compared to baseline and carprofen alone. Conclusions and Clinical Importance Omeprazole prophylaxis induces fecal dysbiosis and increases intestinal inflammatory markers when coadministered with carprofen to otherwise healthy dogs with no other risk factors for GI bleeding.
Collapse
Affiliation(s)
- Susan M Jones
- College of Veterinary Medicine, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Ann Gaier
- College of Veterinary Medicine, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Hiroko Enomoto
- College of Veterinary Medicine, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Patricia Ishii
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Rachel Pilla
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Josh Price
- Office of Information and Technology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jan Suchodolski
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Joerg M Steiner
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Mark G Papich
- College of Veterinary Medicine, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Kristen Messenger
- College of Veterinary Medicine, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - M Katherine Tolbert
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
21
|
Flowers SA, Bhat S, Lee JC. Potential Implications of Gut Microbiota in Drug Pharmacokinetics and Bioavailability. Pharmacotherapy 2020; 40:704-712. [DOI: 10.1002/phar.2428] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Stephanie A. Flowers
- Department of Pharmacy Practice University of Illinois at Chicago Chicago Illinois USA
| | - Shubha Bhat
- Department of Pharmacy Practice Boston Medical Center Boston Massachusetts USA
| | - James C. Lee
- Department of Pharmacy Practice University of Illinois at Chicago Chicago Illinois USA
| |
Collapse
|
22
|
Cervantes-García D, Bahena-Delgado AI, Jiménez M, Córdova-Dávalos LE, Ruiz-Esparza Palacios V, Sánchez-Alemán E, Martínez-Saldaña MC, Salinas E. Glycomacropeptide Ameliorates Indomethacin-Induced Enteropathy in Rats by Modifying Intestinal Inflammation and Oxidative Stress. Molecules 2020; 25:molecules25102351. [PMID: 32443501 PMCID: PMC7287897 DOI: 10.3390/molecules25102351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
Nonsteroidal anti-inflammatory drug (NSAID)-induced enteropathy is considered a serious and increasing clinical problem without available treatment. Glycomacropeptide (GMP) is a 64-amino acid peptide derived from milk κ-casein with numerous biological activities. The aim of this study was to investigate the protective effect of GMP on NSAID enteropathy in rats. Enteropathy was induced by seven days oral indomethacin administration. Rats were orally GMP treated from seven days previous and during the establishment of the enteropathy model. Changes in metabolism, hematological and biochemical blood alterations, intestinal inflammation and oxidative damage were analyzed. Integrity barrier markers, macroscopic intestinal damage and survival rate were also evaluated. GMP treatment prevented anorexia and weight loss in animals. Furthermore, prophylaxis with GMP ameliorated the decline in hemoglobin, hematocrit, albumin and total protein levels. The treatment had no therapeutic efficacy on the decrease of occludin and mucin (MUC)-2 expression in intestinal tissue. However, GMP markedly decreased neutrophil infiltration, and CXCL1, interleukin-1β and inducible nitric oxide synthase expression. Nitric oxide production and lipid hydroperoxide level in the small intestine were also diminished. These beneficial effects were mirrored by preventing ulcer development and increasing animal survival. These results suggest that GMP may protect against NSAID enteropathy through anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Daniel Cervantes-García
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
- National Council of Science and Technology, Mexico City 03940, Mexico
| | - Armida I. Bahena-Delgado
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Mariela Jiménez
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Laura E. Córdova-Dávalos
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Vanessa Ruiz-Esparza Palacios
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
| | - Esperanza Sánchez-Alemán
- Department of Morphology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (E.S.-A.); (M.C.M.-S.)
- Unit of Familiar Medicine #8, Mexican Institute of Social Security, Aguascalientes 20180, Mexico
| | - María C. Martínez-Saldaña
- Department of Morphology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (E.S.-A.); (M.C.M.-S.)
| | - Eva Salinas
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes 20131, Mexico; (D.C.-G.); (A.I.B.-D.); (M.J.); (L.E.C.-D.); (V.R.-E.P.)
- Correspondence: ; Tel.: +52-449-910-8424
| |
Collapse
|
23
|
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
|
24
|
Maseda D, Ricciotti E. NSAID-Gut Microbiota Interactions. Front Pharmacol 2020; 11:1153. [PMID: 32848762 PMCID: PMC7426480 DOI: 10.3389/fphar.2020.01153] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAID)s relieve pain, inflammation, and fever by inhibiting the activity of cyclooxygenase isozymes (COX-1 and COX-2). Despite their clinical efficacy, NSAIDs can cause gastrointestinal (GI) and cardiovascular (CV) complications. Moreover, NSAID use is characterized by a remarkable individual variability in the extent of COX isozyme inhibition, therapeutic efficacy, and incidence of adverse effects. The interaction between the gut microbiota and host has emerged as a key player in modulating host physiology, gut microbiota-related disorders, and metabolism of xenobiotics. Indeed, host-gut microbiota dynamic interactions influence NSAID disposition, therapeutic efficacy, and toxicity. The gut microbiota can directly cause chemical modifications of the NSAID or can indirectly influence its absorption or metabolism by regulating host metabolic enzymes or processes, which may have consequences for drug pharmacokinetic and pharmacodynamic properties. NSAID itself can directly impact the composition and function of the gut microbiota or indirectly alter the physiological properties or functions of the host which may, in turn, precipitate in dysbiosis. Thus, the complex interconnectedness between host-gut microbiota and drug may contribute to the variability in NSAID response and ultimately influence the outcome of NSAID therapy. Herein, we review the interplay between host-gut microbiota and NSAID and its consequences for both drug efficacy and toxicity, mainly in the GI tract. In addition, we highlight progress towards microbiota-based intervention to reduce NSAID-induced enteropathy.
Collapse
Affiliation(s)
- Damian Maseda
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, and Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Emanuela Ricciotti,
| |
Collapse
|
25
|
Chao G, Ye F, Shen W, Gong W, Zhang S. Study on the characteristic of intestinal flora in patients with dual antiplatelet therapy. J Drug Target 2019; 28:500-507. [PMID: 31613141 DOI: 10.1080/1061186x.2019.1681433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective: The objective of the study was to explore the intestinal flora in patients with dual antiplatelet therapy.Method: We collected fresh stool specimens from 10 patients receiving dual antiplatelet therapy and 10 healthy people as control group. Then, we extracted faecal DNA, amplified 16 s rNDA V3-V4 area, and applied Illumina Miseq to analyse the structure of intestinal flora.Result: At class level, DAPT group show higher abundance of class Bacilli and lower abundance of class Erysipelotrichia. At order level, the abundance of order Lactobacillales in DAPT group is higher (p < .05), while the abundance of order Erysipelotrichales is lower in DAPT group (p < .05). At family level, the abundance of family Streptococcaceae and family Lactobacillaceae in DAPT group is higher (p < .05), while the abundance of family Acidaminococcaceae and family Erysipelotrichaceae is lower in DAPT group (p < .05). At genus level, the abundance of genus Streptococcus and genus Klebsiella in DAPT group is higher (p < .05), while the abundance of genus Blautia, genus Phascolarctobacterium and genus Megamonas is lower in DAPT group (p < .05).Conclusion: Taking aspirin and clopidogrel will not cause a change in the biodiversity of intestinal flora. There are significant differences in the intestinal flora of DAPT group compared with the control group at class, order, family and genus level.
Collapse
Affiliation(s)
- Guanqun Chao
- Department of Family Medicine, Sir Run Run Shaw Hospital, Zhejiang University, China
| | - Fangxu Ye
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, China
| | - Wei Shen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, China
| | - Wenqian Gong
- Department of Integrated Traditional Chinese and Western Medicine on Oncology, Ningbo Yinzhou People Hospital, Zhejiang, China
| | - Shuo Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang Chinese Medical University, China
| |
Collapse
|
26
|
Sugimura N, Otani K, Watanabe T, Nakatsu G, Shimada S, Fujimoto K, Nadatani Y, Hosomi S, Tanaka F, Kamata N, Taira K, Nagami Y, Tanigawa T, Uematsu S, Fujiwara Y. High-fat diet-mediated dysbiosis exacerbates NSAID-induced small intestinal damage through the induction of interleukin-17A. Sci Rep 2019; 9:16796. [PMID: 31727909 PMCID: PMC6856170 DOI: 10.1038/s41598-019-52980-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/25/2019] [Indexed: 01/03/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) cause damage in the small intestine in a bacteria-dependent manner. As high-fat diet (HFD) is a potent inducer of gut dysbiosis, we investigated the effects of HFD on bacterial flora in the small intestine and NSAID-induced enteropathy. 16S rRNA gene analysis revealed that the population of Bifidobacterium spp. significantly decreased by fold change of individual operational taxonomic units in the small intestine of mice fed HFD for 8 weeks. HFD increased intestinal permeability, as indicated by fluorescein isothiocyanate-dextran absorption and serum lipopolysaccharide levels, accompanied by a decrease in the protein expressions of ZO-1 and occludin and elevated mRNA expression of interleukin (IL)-17A in the small intestine. HFD-fed mice exhibited increased susceptibility to indomethacin-induced damage in the small intestine; this phenotype was observed in normal diet-fed mice that received small intestinal microbiota from HFD-fed mice. Administration of neutralizing antibodies against IL-17A to HFD-fed mice reduced intestinal permeability and prevented exacerbation of indomethacin-induced damage. Thus, HFD-induced microbial dysbiosis in small intestine caused microinflammation through the induction of IL-17A and increase in intestinal permeability, resulting in the aggravation of NSAID-induced small intestinal damage.
Collapse
Affiliation(s)
- Naoki Sugimura
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koji Otani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Toshio Watanabe
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Geicho Nakatsu
- Department of Immunology and Infectious Diseases/Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Room 904, Building 1, 665 Huntington Avenue, Boston, Massachusetts, 02115, United States
| | - Sunao Shimada
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Noriko Kamata
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koichi Taira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasuaki Nagami
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
27
|
Mortensen B, Murphy C, O'Grady J, Lucey M, Elsafi G, Barry L, Westphal V, Wellejus A, Lukjancenko O, Eklund AC, Nielsen HB, Baker A, Damholt A, van Hylckama Vlieg JET, Shanahan F, Buckley M. Bifidobacteriumbreve Bif195 Protects Against Small-Intestinal Damage Caused by Acetylsalicylic Acid in Healthy Volunteers. Gastroenterology 2019; 157:637-646.e4. [PMID: 31095949 DOI: 10.1053/j.gastro.2019.05.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/07/2019] [Accepted: 05/07/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Enteropathy and small-intestinal ulcers are common adverse effects of nonsteroidal anti-inflammatory drugs such as acetylsalicylic acid (ASA). Safe, cytoprotective strategies are needed to reduce this risk. Specific bifidobacteria might have cytoprotective activities, but little is known about these effects in humans. We used serial video capsule endoscopy (VCE) to assess the efficacy of a specific Bifidobacterium strain in healthy volunteers exposed to ASA. METHODS We performed a single-site, double-blind, parallel-group, proof-of-concept analysis of 75 heathy volunteers given ASA (300 mg) daily for 6 weeks, from July 31 through October 24, 2017. The participants were randomly assigned (1:1) to groups given oral capsules of Bifidobacterium breve (Bif195) (≥5 × 1010 colony-forming units) or placebo daily for 8 weeks. Small-intestinal damage was analyzed by serial VCE at 6 visits. The area under the curve (AUC) for intestinal damage (Lewis score) and the AUC value for ulcers were the primary and first-ranked secondary end points of the trial, respectively. RESULTS Efficacy data were obtained from 35 participants given Bif195 and 31 given placebo. The AUC for Lewis score was significantly lower in the Bif195 group (3040 ± 1340 arbitrary units) than the placebo group (4351 ± 3195) (P = .0376). The AUC for ulcer number was significantly lower in the Bif195 group (50.4 ± 53.1 arbitrary units) than in the placebo group (75.2 ± 85.3 arbitrary units) (P = .0258). Twelve adverse events were reported from the Bif195 group and 20 from the placebo group. None of the events was determined to be related to Bif195 intake. CONCLUSIONS In a randomized, double-blind trial of healthy volunteers, we found oral Bif195 to safely reduce the risk of small-intestinal enteropathy caused by ASA. ClinicalTrials.gov no: NCT03228589.
Collapse
Affiliation(s)
| | - Clodagh Murphy
- APC Microbiome Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - John O'Grady
- APC Microbiome Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Mary Lucey
- Centre for Gastroenterology, Mercy University Hospital, Cork, Ireland
| | - Gafer Elsafi
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Lillian Barry
- Centre for Gastroenterology, Mercy University Hospital, Cork, Ireland
| | - Vibeke Westphal
- Chr. Hansen A/S, Human Health Innovation, Hoersholm, Denmark
| | - Anja Wellejus
- Chr. Hansen A/S, Human Health Innovation, Hoersholm, Denmark
| | | | | | | | - Adam Baker
- Chr. Hansen A/S, Human Health Innovation, Hoersholm, Denmark
| | - Anders Damholt
- Chr. Hansen A/S, Human Health Innovation, Hoersholm, Denmark
| | | | - Fergus Shanahan
- APC Microbiome Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland.
| | - Martin Buckley
- APC Microbiome Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland; Centre for Gastroenterology, Mercy University Hospital, Cork, Ireland
| |
Collapse
|
28
|
Bruno G, Zaccari P, Rocco G, Scalese G, Panetta C, Porowska B, Pontone S, Severi C. Proton pump inhibitors and dysbiosis: Current knowledge and aspects to be clarified. World J Gastroenterol 2019; 25:2706-2719. [PMID: 31235994 PMCID: PMC6580352 DOI: 10.3748/wjg.v25.i22.2706] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/02/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Proton pump inhibitors (PPIs) are common medications within the practice of gastroenterology. These drugs, which act through the irreversible inhibition of the hydrogen/potassium pump (H+/K+-ATPase pump) in the gastric parietal cells, are used in the treatment of several acid-related disorders. PPIs are generally well tolerated but, through the long-term reduction of gastric acid secretion, can increase the risk of an imbalance in gut microbiota composition (i.e., dysbiosis). The gut microbiota is a complex ecosystem in which microbes coexist and interact with the human host. Indeed, the resident gut bacteria are needed for multiple vital functions, such as nutrient and drug metabolism, the production of energy, defense against pathogens, the modulation of the immune system and support of the integrity of the gut mucosal barrier. The bacteria are collected in communities that vary in density and composition within each segment of the gastrointestinal (GI) tract. Therefore, every change in the gut ecosystem has been connected to an increased susceptibility or exacerbation of various GI disorders. The aim of this review is to summarize the recently available data on PPI-related microbiota alterations in each segment of the GI tract and to analyze the possible involvement of PPIs in the pathogenesis of several specific GI diseases.
Collapse
Affiliation(s)
- Giovanni Bruno
- Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Rome 00161, Italy
| | - Piera Zaccari
- Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Rome 00161, Italy
| | - Giulia Rocco
- Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Rome 00161, Italy
| | - Giulia Scalese
- Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Rome 00161, Italy
| | - Cristina Panetta
- Department of Surgical Sciences, Sapienza University of Rome, Rome 00161, Italy
| | - Barbara Porowska
- Department of Cardio-Thoracic, Vascular Surgery and Transplants, Sapienza University of Rome, Rome 00161, Italy
| | - Stefano Pontone
- Department of Surgical Sciences, Sapienza University of Rome, Rome 00161, Italy
| | - Carola Severi
- Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Rome 00161, Italy
| |
Collapse
|
29
|
Lee MY, Choi SC, Kim YS. The Role of Gut Microbiota and Use of Probiotics in the Treatment of Upper Gastrointestinal Diseases. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2019. [DOI: 10.7704/kjhugr.2019.19.2.99] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
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:medicina55030059. [PMID: 30818850 PMCID: PMC6473446 DOI: 10.3390/medicina55030059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [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
|
31
|
Morhardt TL, Hayashi A, Ochi T, Quirós M, Kitamoto S, Nagao-Kitamoto H, Kuffa P, Atarashi K, Honda K, Kao JY, Nusrat A, Kamada N. IL-10 produced by macrophages regulates epithelial integrity in the small intestine. Sci Rep 2019; 9:1223. [PMID: 30718924 PMCID: PMC6362270 DOI: 10.1038/s41598-018-38125-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Macrophages (Mϕs) are known to be major producers of the anti-inflammatory cytokine interleukin-10 (IL-10) in the intestine, thus playing an important role in maintaining gastrointestinal homeostasis. Mϕs that reside in the small intestine (SI) have been previously shown to be regulated by dietary antigens, while colonic Mϕs are regulated by the microbiota. However, the role which resident Mϕs play in SI homeostasis has not yet been fully elucidated. Here, we show that SI Mϕs regulate the integrity of the epithelial barrier via secretion of IL-10. We used an animal model of non-steroidal anti-inflammatory drug (NSAID)-induced SI epithelial injury to show that IL-10 is mainly produced by MHCII+ CD64+ Ly6Clow Mϕs early in injury and that it is involved in the restoration of the epithelial barrier. We found that a lack of IL-10, particularly its secretion by Mϕs, compromised the recovery of SI epithelial barrier. IL-10 production by MHCII+ CD64+ Ly6Clow Mϕs in the SI is not regulated by the gut microbiota, hence depletion of the microbiota did not influence epithelial regeneration in the SI. Collectively, these results highlight the critical role IL-10-producing Mϕs play in recovery from intestinal epithelial injury induced by NSAID.
Collapse
Affiliation(s)
- Tina L Morhardt
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Division of Pediatric Gastroenterology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Atsushi Hayashi
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Research Laboratory, Miyarisan Pharmaceutical Co., Ltd, Tokyo, 114-0016, Japan
| | - Takanori Ochi
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Miguel Quirós
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sho Kitamoto
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Hiroko Nagao-Kitamoto
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Peter Kuffa
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - John Y Kao
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Whitfield-Cargile CM, Chamoun-Emanuelli AM, Cohen ND, Richardson LM, Ajami NJ, Dockery HJ. Differential effects of selective and non-selective cyclooxygenase inhibitors on fecal microbiota in adult horses. PLoS One 2018; 13:e0202527. [PMID: 30138339 PMCID: PMC6107168 DOI: 10.1371/journal.pone.0202527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/03/2018] [Indexed: 01/15/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are routinely used in both veterinary and human medicine. Gastrointestinal injury is a frequent adverse event associated with NSAID use and evidence suggests that NSAIDs induce gastrointestinal microbial imbalance (i.e., dysbiosis) in both animals and people. It is unknown, however, whether cyclooxygenase (COX)-2-selective NSAIDs induce dysbiosis, or if this phenomenon occurs in horses administered any class of NSAIDs. Therefore, our objectives were to determine whether the composition and diversity of the fecal microbiota of adult horses were altered by NSAID use, and whether these effects differed between non-selective and COX-2-selective NSAIDs. Twenty-five adult horses were randomly assigned to 1 of 3 groups: control (n = 5); phenylbutazone (n = 10); or, firocoxib (n = 10). Treatments were administered for 10 days. Fecal samples were collected every 5 days for 25 days. DNA was extracted from feces and the 16S rRNA gene amplified and sequenced to determine the composition of the microbiota and the inferred metagenome. While the fecal microbiota profile of the control group remained stable over time, the phenylbutazone and firocoxib groups had decreased diversity, and alteration of their microbiota profiles was most pronounced at day 10. Similarly, there were clear alterations of the inferred metagenome at day 10 compared to all other days, indicating that use of both non-selective and selective COX inhibitors resulted in temporary alterations of the fecal microbiota and inferred metagenome. Dysbiosis associated with NSAID administration is clinically relevant because dysbiosis has been associated with several important diseases of horses including abdominal pain (colic), colitis, enteric infections, and laminitis.
Collapse
Affiliation(s)
- Canaan M. Whitfield-Cargile
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| | - Ana M. Chamoun-Emanuelli
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Noah D. Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Lauren M. Richardson
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Nadim J. Ajami
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hannah J. Dockery
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
33
|
Jia L, Chopp M, Wang L, Lu X, Szalad A, Zhang ZG. Exosomes derived from high-glucose-stimulated Schwann cells promote development of diabetic peripheral neuropathy. FASEB J 2018; 32:fj201800597R. [PMID: 29932869 PMCID: PMC6219828 DOI: 10.1096/fj.201800597r] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Schwann cells actively interact with axons of dorsal root ganglia (DRG) neurons. Exosomes mediate intercellular communication by transferring their biomaterials, including microRNAs (miRs) into recipient cells. We hypothesized that exosomes derived from Schwann cells stimulated by high glucose (HG) exosomes accelerate development of diabetic peripheral neuropathy and that exosomal cargo miRs contribute to this process. We found that HG exosomes contained high levels of miR-28, -31a, and -130a compared to exosomes derived from non-HG-stimulated Schwann cells. In vitro, treatment of distal axons with HG exosomes resulted in reduction of axonal growth, which was associated with elevation of miR-28, -31a, and -130a and reduction of their target proteins of DNA methyltransferase-3α, NUMB (an endocytic adaptor protein), synaptosome associated protein 25, and growth-associated protein-43 in axons. In vivo, administration of HG exosomes to sciatic nerves of diabetic db/db mice at 7 wk of age promoted occurrence of peripheral neuropathy characterized by impairment of nerve conduction velocity and induction of mechanic and thermal hypoesthesia, which was associated with substantial decreases in intraepidermal nerve fibers. Our findings demonstrate a functional role of exosomes derived from HG-stimulated Schwann cells in mediating development of diabetic peripheral neuropathy.-Jia, L., Chopp, M., Wang, L., Lu, X., Szalad, A., Zhang, Z. G. Exosomes derived from high-glucose-stimulated Schwann cells promote development of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Longfei Jia
- Inovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Lei Wang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
| | - Xuerong Lu
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA; and
| |
Collapse
|
34
|
Edogawa S, Peters SA, Jenkins GD, Gurunathan SV, Sundt WJ, Johnson S, Lennon RJ, Dyer RB, Camilleri M, Kashyap PC, Farrugia G, Chen J, Singh RJ, Grover M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota. FASEB J 2018; 32:fj201800560R. [PMID: 29897814 PMCID: PMC6219825 DOI: 10.1096/fj.201800560r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Intestinal barrier function and microbiota are integrally related and play critical roles in maintenance of host physiology. Sex is a key biologic variable for several disorders. Our aim was to determine sex-based differences in response to perturbation and subsequent recovery of intestinal barrier function and microbiota in healthy humans. Twenty-three volunteers underwent duodenal biopsies, mucosal impedance, and in vivo permeability measurement. Permeability testing was repeated after administration of indomethacin, then 4 to 6 wk after its discontinuation. Duodenal and fecal microbiota composition was determined using 16S rRNA amplicon sequencing. Healthy women had lower intestinal permeability and higher duodenal and fecal microbial diversity than healthy men. Intestinal permeability increases after indomethacin administration in both sexes. However, only women demonstrated decreased fecal microbial diversity, including an increase in Prevotella abundance, after indomethacin administration. Duodenal microbiota composition did not show sex-specific changes. The increase in permeability and microbiota changes normalized after discontinuation of indomethacin. In summary, women have lower intestinal permeability and higher microbial diversity. Intestinal permeability is sensitive to perturbation but recovers to baseline. Gut microbiota in women is sensitive to perturbation but appears to be more stable in men. Sex-based differences in intestinal barrier function and microbiome should be considered in future studies.-Edogawa, S., Peters, S. A., Jenkins, G. D., Gurunathan, S. V., Sundt, W. J., Johnson, S., Lennon, R. J., Dyer, R. B., Camilleri, M., Kashyap, P. C., Farrugia, G., Chen, J., Singh, R. J., Grover, M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota.
Collapse
Affiliation(s)
- Shoko Edogawa
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephanie A. Peters
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory D. Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Wendy J. Sundt
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen Johnson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan J. Lennon
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Roy B. Dyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jun Chen
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ravinder J. Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
35
|
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
|
36
|
Wallace JL, Motta JP, Buret AG. Hydrogen sulfide: an agent of stability at the microbiome-mucosa interface. Am J Physiol Gastrointest Liver Physiol 2018; 314:G143-G149. [PMID: 29025733 PMCID: PMC5866422 DOI: 10.1152/ajpgi.00249.2017] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/19/2017] [Accepted: 10/04/2017] [Indexed: 01/31/2023]
Abstract
A diverse range of effects of the intestinal microbiota on mucosal defense and injury has become increasingly clear over the past decade. Hydrogen sulfide (H2S) has emerged as an important mediator of many physiological functions, including gastrointestinal mucosal defense and repair. Hydrogen sulfide is produced by gastrointestinal tract tissues and by bacteria residing within the gut and can influence the function of a wide range of cells. The microbiota also appears to be an important target of hydrogen sulfide. H2S donors can modify the gut microbiota, and the gastrointestinal epithelium is a major site of oxidation of microbial-derived H2S. When administered together with nonsteroidal anti-inflammatory drugs, H2S can prevent some of the dysbiosis those drugs induce, possibly contributing to the observed prevention of gastrointestinal damage. Exogenous H2S can also markedly reduce the severity of experimental colitis and plays important roles in modulating epithelial cell-mucus-bacterial interactions in the intestine, contributing to its ability to promote resolution of inflammation and repair of tissue injury. In this paper we review recent studies examining the roles of H2S in mucosal defense, the possibility that H2S can damage the gastrointestinal epithelium, and effects of H2S on the gut microbiota and on mucus and biofilm interactions in the context of intestinal inflammation.
Collapse
Affiliation(s)
- John L Wallace
- Department of Physiology and Pharmacology, University of Calgary , Calgary, Alberta , Canada
- Faculty of Medicine, Universidade Camilo Castelo Branco, Fernandópolis, Brazil
| | - Jean-Paul Motta
- Institut de Recherche en Santé Digestive, Université de Toulouse , Toulouse , France
- Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier , Toulouse , France
- Department of Biological Sciences, University of Calgary , Calgary, Alberta , Canada
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
37
|
The non-invasive exfoliated transcriptome (exfoliome) reflects the tissue-level transcriptome in a mouse model of NSAID enteropathy. Sci Rep 2017; 7:14687. [PMID: 29089621 PMCID: PMC5665873 DOI: 10.1038/s41598-017-13999-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most frequently used classes of medications in the world, yet they induce an enteropathy that is associated with high morbidity and mortality. A major limitation to better understanding the pathophysiology and diagnosis of this enteropathy is the difficulty of obtaining information about the primary site of injury, namely the distal small intestine. We investigated the utility of using mRNA from exfoliated cells in stool as a means to surveil the distal small intestine in a murine model of NSAID enteropathy. Specifically, we performed RNA-Seq on exfoliated cells found in feces and compared these data to RNA-Seq from both the small intestinal mucosa and colonic mucosa of healthy control mice or those exhibiting NSAID-induced enteropathy. Global gene expression analysis, data intersection, pathway analysis, and computational approaches including linear discriminant analysis (LDA) and sparse canonical correlation analysis (CCA) were used to assess the inter-relatedness of tissue (invasive) and stool (noninvasive) datasets. These analyses revealed that the exfoliated cell transcriptome closely mirrored the transcriptome of the small intestinal mucosa. Thus, the exfoliome may serve as a non-invasive means of detecting and monitoring NSAID enteropathy (and possibly other gastrointestinal mucosal inflammatory diseases).
Collapse
|
38
|
Shin SJ, Noh CK, Lim SG, Lee KM, Lee KJ. Non-steroidal anti-inflammatory drug-induced enteropathy. Intest Res 2017; 15:446-455. [PMID: 29142512 PMCID: PMC5683975 DOI: 10.5217/ir.2017.15.4.446] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 12/18/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are well known to be associated with serious upper gastrointestinal complications, such as peptic ulcer, bleeding, perforation, and obstruction. Recently, attention has been mainly focused on the small bowel injuries caused by NSAIDs, and new endoscopic techniques such as capsule endoscopy and double balloon endoscopy can help in detecting such injuries. This article reviewed the epidemiology, pathogenesis, clinical manifestations, diagnosis, and treatment of small bowel injuries caused by NSAIDs. Small bowel injures by NSAIDs might occur with a similar frequency and extent as those observed in the upper gastrointestinal tract. The pathogenesis of NSAID-induced enteropathy is complex and not clearly understood. The various lesions observed in the small bowel, including petechiae, reddened folds, loss of villi, erosions, and ulcers can be detected by capsule endoscopy. A drug that could prevent or treat NSAID-induced enteropathy has not yet been developed. Therefore, further investigations should be performed to elucidate the pathogenesis of such enteropathy and develop suitable preventive and treatment strategies.
Collapse
Affiliation(s)
- Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Sun Gyo Lim
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Kee Myung Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Kwang Jae Lee
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
39
|
Srinivasan A, De Cruz P. Review article: a practical approach to the clinical management of NSAID enteropathy. Scand J Gastroenterol 2017; 52:941-947. [PMID: 28587496 DOI: 10.1080/00365521.2017.1335769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Co-prescription of acid suppressive therapy, together with advances in small bowel imaging techniques, have shifted the burden of NSAID-related toxicity from gastro-duodenal to more distal small bowel injury. Due to predominantly subclinical disease, NSAID enteropathy remains under-recognised, with an incidence of 53-80% amongst healthy short-term users, and a prevalence of 50-71% following long-term (>3 months) use. Despite their distinct pathogenesis, those at risk of NSAID-related gastro-duodenal and small bowel complications share several risk factors. Clinical complications of NSAID enteropathy such as protein-losing enteropathy, small bowel strictures and diaphragm disease, confer significant morbidity, and are often irreversible. Small bowel prophylaxis has proven of modest efficacy after short-term, high-dose NSAID use in asymptomatic patients. While selective COX-2 inhibitors are associated with fewer gastro-duodenal complications relative to non-selective NSAIDs, their comparative benefit in protecting against small bowel enteropathy remains unclear. Prophylaxis should be considered in those at high risk of small bowel complications, as treatment options for established disease remain limited; however, the optimal agent remains unclear. We propose a clinical algorithm that may help prevent, monitor, investigate, and manage the sequelae of NSAID-induced small bowel toxicity.
Collapse
Affiliation(s)
- Ashish Srinivasan
- a Department of Gastroenterology , Austin Health , Melbourne , Australia
| | - Peter De Cruz
- a Department of Gastroenterology , Austin Health , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia
| |
Collapse
|
40
|
Otani K, Tanigawa T, Watanabe T, Shimada S, Nadatani Y, Nagami Y, Tanaka F, Kamata N, Yamagami H, Shiba M, Tominaga K, Fujiwara Y, Arakawa T. Microbiota Plays a Key Role in Non-Steroidal Anti-Inflammatory Drug-Induced Small Intestinal Damage. Digestion 2017; 95:22-28. [PMID: 28052268 DOI: 10.1159/000452356] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) damage the small intestine by causing multiple erosions and ulcers. However, to date, no established therapies and prophylactic agents are available to treat such damages. We reviewed the role of intestinal microbiota in NSAID-induced intestinal damage and identified potential therapeutic candidates. SUMMARY The composition of the intestinal microbiota is an important factor in the pathophysiology of NSAID-induced small intestinal damage. Once mucosal barrier function is disrupted due to NSAID-induced prostaglandin deficiency and mitochondrial malfunction, lipopolysaccharide from luminal gram-negative bacteria and high mobility group box 1 from the injured epithelial cells activate toll-like receptor 4-signaling pathway and nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 inflammasome; this leads to the release of proinflammatory cytokines such as tumor necrosis factor-α and interleukin-1β. Proton pump inhibitors (PPIs) are often used for the prevention of NSAID-induced injuries to the upper gastrointestinal tract. However, several studies indicate that PPIs may induce dysbiosis, which may exacerbate the NSAID-induced small intestinal damage. Our recent research suggests that probiotics and rebamipide could be used to prevent NSAID-induced small intestinal damage by regulating the intestinal microbiota. Key Messages: Intestinal microbiota plays a key role in NSAID-induced small intestinal damage, and modulating the composition of the intestinal microbiota could be a new therapeutic strategy for treating this damage.
Collapse
Affiliation(s)
- Koji Otani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Xiao X, Nakatsu G, Jin Y, Wong S, Yu J, Lau JYW. Gut Microbiota Mediates Protection Against Enteropathy Induced by Indomethacin. Sci Rep 2017; 7:40317. [PMID: 28067296 PMCID: PMC5220306 DOI: 10.1038/srep40317] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/05/2016] [Indexed: 02/05/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) can cause significant small bowel injuries. The role of gut microbiota in this NSAID-induced enteropathy is poorly understood. We studied the dynamic changes in gut microbiota following indomethacin administration in mice, and investigated the effects of these adaptive changes on subsequent NSAID-induced enteropathy. The changes in gut microbiota were studied using 16S rRNA sequencing, and the effects of such changes were investigated using antibiotics and a faecal transplantation model. After indomethacin treatment, significant adaptive changes in gut microbiota were observed, including increased abundance of Firmicutes and decreased abundance in that of Bacteroidetes. Depletion of gut microbiota with antibiotics led to a higher mortality (P = 0.0021) in mice compared to controls. Mice pre-transplanted with adaptively changed microbiota showed less small bowel injury and lower levels of pro-inflammatory cytokines when exposed to indomethacin. In summary, this study identifies adaptive changes in the gut microbiota upon indomethacin administration, which can in turn ameliorate further NSAID-induced injury. The heightened mortality with antibiotic depletion of the adaptively changed microbiota suggests its important role in protecting against such injury. This study provides insight for future efforts to target the microbiota as a therapeutic strategy.
Collapse
Affiliation(s)
- Xue Xiao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.,Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.,Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Geicho Nakatsu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Ye Jin
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sunny Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - James Y W Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.,Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
42
|
Byun S, Lim T, Lim Y, Seo J, Chung M. In vivo effects of s-pantoprazole, polaprenzinc, and probiotic blend on chronic small intestinal injury induced by indomethacin. Benef Microbes 2016; 7:731-737. [DOI: 10.3920/bm2016.0029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment and prevention methods for non-steroidal anti-inflammatory drug-induced enteropathy have not yet been established. We tested the preventive effects of s-pantoprazole sodium trihydrate (PAN), polaprezinc (PZ), and probiotics on an indomethacin (Indo)-induced small intestinal injury in a rat model. Rats were randomised into 6 groups to receive: normal saline (control), Indo (6 mg/kg), PZ plus Indo, PAN plus Indo, or probiotics plus Indo (at 108 and 109 cfu/head) for 2 weeks. We measured body weight, food intake, severity of small intestinal damage, haemoglobin (Hb) levels in the small intestinal fluid, intestinal inflammatory cytokines, and a few groups of faecal bacteria. The experimental groups were found to have the following survival rates: 0% for the Indo, PZ, and PAN groups; 50% for both probiotic groups; and 100% for control. Treatment with probiotics of different concentrations reduced small intestinal lesion scores and intestinal fluid Hb as compared with the Indo group, while these parameters did not reduce in the PZ and PAN groups. The anti-inflammatory marker interleukin 10 increased in both probiotic groups. Analysis of a few groups of faecal bacteria revealed that Indo-induced a significant increase in Gram-negative bacteria and decreases in Bifidobacterium and Lactobacillus. Similar changes were also observed in the PZ and PAN groups. However, opposite effects were found in both probiotic groups. The use of probiotics appeared to be beneficial in preventing Indo-induced chronic small intestinal injury.
Collapse
Affiliation(s)
- S.J. Byun
- Department of Internal Medicine, Ilsan Hospital, College of Medicine, Dongguk University, Goyang 410-773, Republic of Korea
| | - T.J. Lim
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| | - Y.J. Lim
- Department of Internal Medicine, Ilsan Hospital, College of Medicine, Dongguk University, Goyang 410-773, Republic of Korea
| | - J.G. Seo
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| | - M.J. Chung
- Research and Development Center, Cell Biotech Co. Ltd., Gimpo, Gyeonggi 157-030, Republic of Korea
| |
Collapse
|
43
|
Montrose DC, Zhou XK, McNally EM, Sue E, Yantiss RK, Gross SS, Leve ND, Karoly ED, Suen CS, Ling L, Benezra R, Pamer EG, Dannenberg AJ. Celecoxib Alters the Intestinal Microbiota and Metabolome in Association with Reducing Polyp Burden. Cancer Prev Res (Phila) 2016; 9:721-31. [PMID: 27432344 PMCID: PMC5010963 DOI: 10.1158/1940-6207.capr-16-0095] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/12/2016] [Indexed: 12/14/2022]
Abstract
Treatment with celecoxib, a selective COX-2 inhibitor, reduces formation of premalignant adenomatous polyps in the gastrointestinal tracts of humans and mice. In addition to its chemopreventive activity, celecoxib can exhibit antimicrobial activity. Differing bacterial profiles have been found in feces from colon cancer patients compared with those of normal subjects. Moreover, preclinical studies suggest that bacteria can modulate intestinal tumorigenesis by secreting specific metabolites. In the current study, we determined whether celecoxib treatment altered the luminal microbiota and metabolome in association with reducing intestinal polyp burden in mice. Administration of celecoxib for 10 weeks markedly reduced intestinal polyp burden in APC(Min/+) mice. Treatment with celecoxib also altered select luminal bacterial populations in both APC(Min/+) and wild-type mice, including decreased Lactobacillaceae and Bifidobacteriaceae as well as increased Coriobacteriaceae Metabolomic analysis demonstrated that celecoxib caused a strong reduction in many fecal metabolites linked to carcinogenesis, including glucose, amino acids, nucleotides, and lipids. Ingenuity Pathway Analysis suggested that these changes in metabolites may contribute to reduced cell proliferation. To this end, we showed that celecoxib reduced cell proliferation in the base of normal appearing ileal and colonic crypts of APC(Min/+) mice. Consistent with this finding, lineage tracing indicated that celecoxib treatment reduced the rate at which Lgr5-positive stem cells gave rise to differentiated cell types in the crypts. Taken together, these results demonstrate that celecoxib alters the luminal microbiota and metabolome along with reducing epithelial cell proliferation in mice. We hypothesize that these actions contribute to its chemopreventive activity. Cancer Prev Res; 9(9); 721-31. ©2016 AACR.
Collapse
Affiliation(s)
- David C Montrose
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Erin M McNally
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Erika Sue
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rhonda K Yantiss
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell Medical College, New York, New York
| | - Nitai D Leve
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | | | - Chen S Suen
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Lilan Ling
- Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric G Pamer
- Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, New York. Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York. Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
44
|
Whitfield-Cargile CM, Cohen ND, Chapkin RS, Weeks BR, Davidson LA, Goldsby JS, Hunt CL, Steinmeyer SH, Menon R, Suchodolski JS, Jayaraman A, Alaniz RC. The microbiota-derived metabolite indole decreases mucosal inflammation and injury in a murine model of NSAID enteropathy. Gut Microbes 2016; 7:246-61. [PMID: 27007819 PMCID: PMC4939928 DOI: 10.1080/19490976.2016.1156827] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently used classes of medications in the world. Unfortunately, NSAIDs induce an enteropathy associated with high morbidity and mortality. Although the pathophysiology of this condition involves the interaction of the gut epithelium, microbiota, and NSAIDs, the precise mechanisms by which microbiota influence NSAID enteropathy are unclear. One possible mechanism is that the microbiota may attenuate the severity of disease by specific metabolite-mediated regulation of host inflammation and injury. The microbiota-derived tryptophan-metabolite indole is abundant in the healthy mammalian gut and positively influences intestinal health. We thus examined the effects of indole administration on NSAID enteropathy. Mice (n = 5 per group) were treated once daily for 7 days with an NSAID (indomethacin; 5 mg/kg), indole (20 mg/kg), indomethacin plus indole, or vehicle only (control). Outcomes compared among groups included: microscopic pathology; fecal calprotectin concentration; proportion of neutrophils in the spleen and mesenteric lymph nodes; fecal microbiota composition and diversity; small intestinal mucosal transcriptome; and, fecal tryptophan metabolites. Co-administration of indole with indomethacin: significantly reduced mucosal pathology scores, fecal calprotectin concentrations, and neutrophilic infiltration of the spleen and mesenteric lymph nodes induced by indomethacin; modulated NSAID-induced perturbation of the microbiota, fecal metabolites, and inferred metagenome; and, abrogated a pro-inflammatory gene expression profile in the small intestinal mucosa induced by indomethacin. The microbiota-derived metabolite indole attenuated multiple deleterious effects of NSAID enteropathy, including modulating inflammation mediated by innate immune responses and altering indomethacin-induced shift of the microbiota.
Collapse
Affiliation(s)
- Canaan M. Whitfield-Cargile
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Noah D. Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Robert S. Chapkin
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA,Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas, USA
| | - Brad R. Weeks
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Laurie A. Davidson
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Jennifer S. Goldsby
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Carrie L. Hunt
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas, USA
| | - Shelby H. Steinmeyer
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas, USA
| | - Rani Menon
- Department of Chemical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
| | - Jan S. Suchodolski
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
45
|
Rogers MAM, Aronoff DM. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin Microbiol Infect 2016; 22:178.e1-178.e9. [PMID: 26482265 PMCID: PMC4754147 DOI: 10.1016/j.cmi.2015.10.003] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/14/2015] [Accepted: 10/05/2015] [Indexed: 01/11/2023]
Abstract
The composition of the gut microbiome with the use of non-steroidal anti-inflammatory drugs (NSAIDs) has not been fully characterized. Drug use within the past 30 days was ascertained in 155 adults, and stool specimens were submitted for analysis. Area under the receiver operating characteristic curve (AUC) was calculated in logit models to distinguish the relative abundance of operational taxonomic units (OTUs) by medication class. The type of medication had a greater influence on the gut microbiome than the number of medications. NSAIDs were particularly associated with distinct microbial populations. Four OTUs (Prevotella species, Bacteroides species, family Ruminococcaceae, and Barnesiella species) discriminated aspirin users from those using no medication (AUC = 0.96; 95% CI 0.84-1.00). The microbiome profile of celecoxib users was similar to that of ibuprofen users, with both showing enrichment of Acidaminococcaceae and Enterobacteriaceae. Bacteria from families Propionibacteriaceae, Pseudomonadaceae, Puniceicoccaceae and Rikenellaceae were more abundant in ibuprofen users than in controls or naproxen users. Bacteroides species and Erysipelotrichaceae species discriminated individuals using NSAIDs plus proton-pump inhibitors from those using NSAIDs alone (AUC = 0.96; 95% CI 0.87-1.00). Bacteroides species and a bacterium of family Ruminococcaceae discriminated individuals using NSAIDs in combination with antidepressants and laxatives from those using NSAIDs alone (AUC = 0.98; 95% CI 0.93-1.00). In conclusion, bacteria in the gastrointestinal tract reflect the combinations of medications that people ingest. The bacterial composition of the gut varied with the type of NSAID ingested.
Collapse
Affiliation(s)
- M A M Rogers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - D M Aronoff
- Department of Medicine and Department of Pathology, Microbiology, & Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
46
|
Pavlidis P, Powell N, Vincent RP, Ehrlich D, Bjarnason I, Hayee B. Systematic review: bile acids and intestinal inflammation-luminal aggressors or regulators of mucosal defence? Aliment Pharmacol Ther 2015. [PMID: 26223936 DOI: 10.1111/apt.13333] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD), comprising Crohn's disease and ulcerative colitis (UC), are chronic conditions attributed to an aberrant immune response to luminal triggers. Recently, published work suggests a pathogenic role for bile acids in this context. AIM To perform a systematic review of studies investigating the role of bile acids in intestinal inflammation and present potentially relevant clinical implications. METHODS Pubmed search for English language articles published up to May 2015. Terms used were: 'bile', 'bile acid', 'barrier', 'small bowel injury', 'Crohn's' and 'colitis'. RESULTS Experimental studies support a variable role for bile acids in intestinal barrier homoeostasis. This may be attributed to different physicochemical properties, variable effects on epithelia and immune cells via bile acids-specific receptors, or through a cross-talk with the gut microbiome. A reduction in the bile acids pool, with lower concentrations of secondary forms, has been recognised for some time in Crohn's disease and associated to ileal dysfunction and bile acids malabsorption. Recent work suggests that these changes, including an increase in sulphated forms, are related to inflammatory activity in both Crohn's disease and UC. The detrimental effects of 'western diet' elements such as emulsifiers and fat, which have been implicated in the development of the current IBD and obesity epidemics, may also be bile acid-mediated. CONCLUSIONS Although there are only a few observational clinical studies to support an interaction, in vivo human and animal studies support an association between bile acids metabolism, the gut microbiome and intestinal inflammation. This may well prove to have significant diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- P Pavlidis
- Department of Gastroenterology, King's College Hospital, London, UK
| | - N Powell
- Division of Transplantation and Mucosal Biology, King's College London, London, UK
| | - R P Vincent
- Department of Biochemistry, King's College Hospital, London, UK
| | - D Ehrlich
- Centre of Host-Microbiome Interactions, King's College London, London, UK
| | - I Bjarnason
- Department of Gastroenterology, King's College Hospital, London, UK
| | - B Hayee
- Department of Gastroenterology, King's College Hospital, London, UK
| |
Collapse
|
47
|
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: 4.2] [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
|
48
|
NSAID enteropathy and bacteria: a complicated relationship. J Gastroenterol 2015; 50:387-93. [PMID: 25572030 DOI: 10.1007/s00535-014-1032-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 02/04/2023]
Abstract
The clinical significance of small intestinal damage caused by nonsteroidal anti-inflammatory drugs (NSAIDs) remains under-appreciated. It occurs with greater frequency than the damage caused by these drugs in the upper gastrointestinal tract, but is much more difficult to diagnose and treat. Although the pathogenesis of NSAID enteropathy remains incompletely understood, it is clear that bacteria, bile, and the enterohepatic circulation of NSAIDs are all important factors. However, they are also interrelated with one another. Bacterial enzymes can affect the cytotoxicity of bile and are essential for enterohepatic circulation of NSAIDs. Gram-negative bacteria appear to be particularly important in the pathogenesis of NSAID enteropathy, possibly through release of endotoxin. Inhibitors of gastric acid secretion significantly aggravate NSAID enteropathy, and this effect is due to significant changes in the intestinal microbiome. Treatment with antibiotics can, in some circumstances, reduce the severity of NSAID enteropathy, but published results are inconsistent. Specific antibiotic-induced changes in the microbiota have not been causally linked to prevention of intestinal damage. Treatment with probiotics, particularly Bifidobacterium, Lactobacillus, and Faecalibacteriaum prausnitzii, has shown promising effects in animal models. Our studies suggest that these beneficial effects are due to colonization by the bacteria, rather than to products released by the bacteria.
Collapse
|
49
|
Marlicz W, Loniewski I, Grimes DS, Quigley EM. Nonsteroidal anti-inflammatory drugs, proton pump inhibitors, and gastrointestinal injury: contrasting interactions in the stomach and small intestine. Mayo Clin Proc 2014; 89:1699-709. [PMID: 25440891 DOI: 10.1016/j.mayocp.2014.07.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/20/2014] [Accepted: 07/28/2014] [Indexed: 02/06/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) and proton pump inhibitors (PPIs) are among the most frequently prescribed groups of drugs worldwide. The use of NSAIDs is associated with a high number of significant adverse effects. Recently, the safety of PPIs has also been challenged. Capsule endoscopy studies reveal that even low-dose NSAIDs are responsible for gut mucosal injury and numerous clinical adverse effects, for example, bleeding and anemia, that might be difficult to diagnose. The frequent use of PPIs can exacerbate NSAID-induced small intestinal injury by altering intestinal microbiota. Thus, the use of PPI is considered to be an independent risk factor associated with NSAID-associated enteropathy. In this review, we discuss this important clinical problem and review relevant aspects of epidemiology, pathophysiology, and management. We also present the hypothesis that even minor and subclinical injury to the intestinal mucosa can result in significant, though delayed, metabolic consequences, which may seriously affect the health of an individual. PubMed was searched using the following key words (each key word alone and in combination): gut microbiota, microbiome, non-steroidal anti inflammatory drugs, proton pump inhibitors, enteropathy, probiotic, antibiotic, mucosal injury, enteroscopy, and capsule endoscopy. Google engine search was also carried out to identify additional relevant articles. Both original and review articles published in English were reviewed.
Collapse
Affiliation(s)
- Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland.
| | - Igor Loniewski
- International Pharmaceutical Consulting, Szczecin, Poland
| | | | - Eamonn M Quigley
- Division of Gastroenterology and Hepatology, Houston Methodist Hospital, and Weill Cornell Medical College, Houston, TX
| |
Collapse
|
50
|
Blackler RW, Motta JP, Manko A, Workentine M, Bercik P, Surette MG, Wallace JL. Hydrogen sulphide protects against NSAID-enteropathy through modulation of bile and the microbiota. Br J Pharmacol 2014; 172:992-1004. [PMID: 25297699 DOI: 10.1111/bph.12961] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/16/2014] [Accepted: 09/24/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulphide is an important mediator of gastrointestinal mucosal defence. The use of non-steroidal anti-inflammatory drugs (NSAIDs) is significantly limited by their toxicity in the gastrointestinal tract. Particularly concerning is the lack of effective preventative or curative treatments for NSAID-induced intestinal damage and bleeding. We evaluated the ability of a hydrogen sulphide donor to protect against NSAID-induced enteropathy. EXPERIMENTAL APPROACH Intestinal ulceration and bleeding were induced in Wistar rats by oral administration of naproxen. The effects of suppression of endogenous hydrogen sulphide synthesis or administration of a hydrogen sulphide donor (diallyl disulphide) on naproxen-induced enteropathy was examined. Effects of diallyl disulphide on small intestinal inflammation and intestinal microbiota were also assessed. Bile collected after in vivo naproxen and diallyl disulphide administration was evaluated for cytotoxicity in vitro using cultured intestinal epithelial cells. KEY RESULTS Suppression of endogenous hydrogen sulphide synthesis by β-cyano-L-alanine exacerbated naproxen-induced enteropathy. Diallyl disulphide co-administration dose-dependently reduced the severity of naproxen-induced small intestinal damage, inflammation and bleeding. Diallyl disulphide administration attenuated naproxen-induced increases in the cytotoxicity of bile on cultured enterocytes, and prevented or reversed naproxen-induced changes in the intestinal microbiota. CONCLUSIONS AND IMPLICATIONS Hydrogen sulphide protects against NSAID-enteropathy in rats, in part reducing the cytotoxicity of bile and preventing NSAID-induced dysbiosis.
Collapse
Affiliation(s)
- Rory W Blackler
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | | | |
Collapse
|