Lafutidine Ameliorates Indomethacin-Induced Small Intestinal Damage in Rats by Modifying the Intestinal Mucosal Barrier, Inflammation, and Microbiota

Zhu L. · Guo J. · Liu Q. · Luo Y. · Zhao J. · Zhong W. · Sun S. · Xu X. · Liang H. · Lou C. · Song C. · Chen J. · Zhao J. · Wang B. · Chen X.

Author affiliations

Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China

Log in to MyKarger to check if you already have access to this content.

Buy FullText & PDF Unlimited re-access via MyKarger Unrestricted printing, no saving restrictions for personal use
read more

CHF 38.00 *
EUR 35.00 *
USD 39.00 *

Select

KAB

Buy a Karger Article Bundle (KAB) and profit from a discount!

If you would like to redeem your KAB credit, please log in.

Save over 20% compared to the individual article price.

Learn more

Rent via DeepDyve Unlimited fulltext viewing of this article Organize, annotate and mark up articles Printing and downloading restrictions apply

Start free trial

Subscribe Access to all articles of the subscribed year(s) guaranteed for 5 years Unlimited re-access via Subscriber Login or MyKarger Unrestricted printing, no saving restrictions for personal use read more

Subcription rates

Select

* The final prices may differ from the prices shown due to specifics of VAT rules.

Article / Publication Details

Received: January 23, 2022
Accepted: February 20, 2023
Published online: April 06, 2023

Number of Print Pages: 15
Number of Figures: 6
Number of Tables: 0

ISSN: 0031-7012 (Print)
eISSN: 1423-0313 (Online)

For additional information: https://www.karger.com/PHA

Abstract

Introduction: Nonsteroidal anti-inflammatory drug (NSAID)-induced small intestinal damage is a serious and escalating clinical problem without effective treatment. Lafutidine (LAF) is a novel histamine H2 receptor antagonist with a mucosal protective action. This study aimed to investigate the protective effect of LAF on indomethacin (IND)-induced enteropathy in rats. Methods: Rats were treated with LAF for 10 days with concomitant IND treatment on the final 5 days. Changes in metabolism and hematological and biochemical parameters were measured, and intestinal damage was blindly scored. Intestinal mucosal tissue and luminal contents were collected for transcriptome and microbiota sequencing. Intestinal inflammation and barrier function were also evaluated. Results: LAF treatment prevented anorexia and weight loss in rats and ameliorated reductions in hemoglobin, hematocrit, total protein, and albumin levels. LAF reduced the severity of IND-induced intestinal damage including macroscopic and histopathological damage score. Transcriptome sequencing results indicated that LAF might have positive effects on intestinal inflammation and the intestinal mucosal barrier. Further research revealed that LAF decreased neutrophil infiltration, and IL-1β and TNF-α expression in intestinal tissue. Besides, the treatment increased mucus secretion, MUC2, Occludin, and ZO-1 expression, and decreased serum D-lactate levels. LAF treatment also ameliorates microbial dysbiosis in small intestine induced by IND and increased the abundance of Lactobacillus acidophilus. Conclusion: LAF may protect against NSAID enteropathy via enhancing the intestinal mucosal barrier, inhibiting inflammation, and regulating microbiota.

© 2023 S. Karger AG, Basel

References Bjarnason I, Scarpignato C, Holmgren E, Olszewski M, Rainsford KD, Lanas A. Mechanisms of damage to the gastrointestinal tract from nonsteroidal anti-inflammatory drugs. Gastroenterology. 2018 Feb;154(3):500–14. Fletcher RH. Review: aspirin reduces colorectal cancer incidence and mortality in patients at average risk. Ann Intern Med. 2016 Aug 16;165(4):JC16. Watanabe T, Fujiwara Y, Chan FKL. Current knowledge on non-steroidal anti-inflammatory drug-induced small-bowel damage: a comprehensive review. J Gastroenterol. 2020 May;55(5):481–95. Tachecí I, Bradna P, Douda T, Baštecká D, Kopáčová M, Rejchrt S, et al. NSAID-induced enteropathy in rheumatoid arthritis patients with chronic occult gastrointestinal bleeding: a prospective capsule endoscopy study. Gastroenterol Res Pract. 2013;2013:268382. Ullah S, Ajab S, Rao R, Raghunathan G, DaCosta P. Diaphragm disease of the small intestine: an interesting case report. Int J Surg Pathol. 2015 Jun;23(4):322–4. Bjarnason I, Hayllar J, MacPherson AJ, Russell AS. Side effects of nonsteroidal anti-inflammatory drugs on the small and large intestine in humans. Gastroenterology. 1993 Jun;104(6):1832–47. Monteros MJM, Galdeano CM, Balcells MF, Weill R, De Paula JA, Perdigón G, et al. Probiotic lactobacilli as a promising strategy to ameliorate disorders associated with intestinal inflammation induced by a non-steroidal anti-inflammatory drug. Sci Rep. 2021 Jan 12;11(1):571. Scheiman JM, Yeomans ND, Talley NJ, Vakil N, Chan FKL, Tulassay Z, et al. Prevention of ulcers by esomeprazole in at-risk patients using non-selective NSAIDs and COX-2 inhibitors. Am J Gastroenterol. 2006 Apr;101(4):701–10. Otani K, Tanigawa T, Watanabe T, Shimada S, Nadatani Y, Nagami Y, et al. Microbiota plays a key role in non-steroidal anti-inflammatory drug-induced small intestinal damage. Digestion. 2017;95(1):22–8. Goldstein JL, Eisen GM, Lewis B, Gralnek IM, Zlotnick S, Fort JG, et al. Video capsule endoscopy to prospectively assess small bowel injury with celecoxib, naproxen plus omeprazole, and placebo. Clin Gastroenterol Hepatol. 2005 Feb;3(2):133–41. Endo H, Sakai E, Taniguchi L, Kessoku T, Komiya Y, Ezuka A, et al. Risk factors for small-bowel mucosal breaks in chronic low-dose aspirin users: data from a prospective multicenter capsule endoscopy registry. Gastrointest Endosc. 2014 Nov;80(5):826–34. Washio E, Esaki M, Maehata Y, Miyazaki M, Kobayashi H, Ishikawa H, et al. Proton pump inhibitors increase incidence of nonsteroidal anti-inflammatory drug-induced small bowel injury: a randomized, placebo-controlled trial. Clin Gastroenterol Hepatol. 2016 Jun;14(6):809–15.e1. Nuki Y, Umeno J, Washio E, Maehata Y, Hirano A, Miyazaki M, et al. The influence of CYP2C19 polymorphisms on exacerbating effect of rabeprazole in celecoxib-induced small bowel injury. Aliment Pharmacol Ther. 2017 Aug;46(3):331–6. Yoshihara T, Oikawa Y, Kato T, Kessoku T, Kobayashi T, Kato S, et al. The protective effect of Bifidobacterium bifidum G9-1 against mucus degradation by Akkermansia muciniphila following small intestine injury caused by a proton pump inhibitor and aspirin. Gut Microbes. 2020 Sep 2;11(5):1385–404. Wallace JL, Syer S, Denou E, de Palma G, Vong L, McKnight W, et al. Proton pump inhibitors exacerbate NSAID-induced small intestinal injury by inducing dysbiosis. Gastroenterology. 2011 Oct;141(4):1314–22, 1322.e1-5. Sugimura N, Otani K, Watanabe T, Nakatsu G, Shimada S, Fujimoto K, et al. High-fat diet-mediated dysbiosis exacerbates NSAID-induced small intestinal damage through the induction of interleukin-17A. Sci Rep. 2019 Nov 14;9(1):16796. Nadatani Y, Watanabe T, Suda W, Nakata A, Matsumoto Y, Kosaka S, et al. Gastric acid inhibitor aggravates indomethacin-induced small intestinal injury via reducing Lactobacillus johnsonii. Sci Rep. 2019 Nov 25;9(1):17490. Shibata M, Yamaura T, Inaba N, Onodera S, Chida Y, Ohnishi H. Gastric antisecretory effect of FRG-8813, a new histamine H2 receptor antagonist, in rats and dogs. Eur J Pharmacol. 1993 Apr 28;235(2–3):245–53. Kato S, Tanaka A, Kunikata T, Umeda M, Takeuchi K. Protective effect of lafutidine against indomethacin-induced intestinal ulceration in rats: relation to capsaicin-sensitive sensory neurons. Digestion. 2000;61(1):39–46. Sano T, Utsumi D, Amagase K, Matsumoto K, Tominaga M, Higuchi K, et al. Lafutidine, a histamine H2 receptor antagonist with mucosal protective properties, attenuates 5-fluorouracil-induced intestinal mucositis in mice through activation of extrinsic primary afferent neurons. J Physiol Pharmacol. 2017 Feb;68(1):79–90. Amagase K, Ochi A, Sugihara T, Kato S, Takeuchi K. Protective effect of lafutidine, a histamine H2 receptor antagonist, against loxoprofen-induced small intestinal lesions in rats. J Gastroenterol Hepatol. 2010 May;25(Suppl 1):S111–8. Singh DP, Borse SP, Nivsarkar M. A novel model for NSAID induced gastroenteropathy in rats. J Pharmacol Toxicol Methods. 2016 Mar-Apr;78:66–75. Saegusa Y, Ichikawa T, Iwai T, Goso Y, Ikezawa T, Nakano M, et al. Effects of acid antisecretory drugs on mucus barrier of the rat against 5-fluorouracil-induced gastrointestinal mucositis. Scand J Gastroenterol. 2008;43(5):531–7. Chiu CJ, McArdle AH, Brown R, Scott HJ, Gurd FN. Intestinal mucosal lesion in low-flow states. I. A morphological, hemodynamic, and metabolic reappraisal. Arch Surg. 1970 Oct;101(4):478–83. Cervantes-García D, Bahena-Delgado AI, Jiménez M, Córdova-Dávalos LE, Ruiz-Esparza Palacios V, Sánchez-Alemán E, et al. Glycomacropeptide ameliorates indomethacin-induced enteropathy in rats by modifying intestinal inflammation and oxidative stress. Molecules. 2020 May 18;25(10):2351. Bradley PP, Priebat DA, Christensen RD, Rothstein G. Measurement of cutaneous inflammation: estimation of neutrophil content with an enzyme marker. J Invest Dermatol. 1982 Mar;78(3):206–9. Aadland E, Fagerhol MK. Faecal calprotectin: a marker of inflammation throughout the intestinal tract. Eur J Gastroenterol Hepatol. 2002 Aug;14(8):823–5. Abrahamian GA, Polhamus CD, Muskat P, Karulf RE. Diaphragm-like strictures of the ileum associated with NSAID use: a rare complication. South Med J. 1998 Apr;91(4):395–7. Bjarnason I, Zanelli G, Prouse P, Smethurst P, Smith T, Levi S, et al. Blood and protein loss via small-intestinal inflammation induced by non-steroidal anti-inflammatory drugs. Lancet. 1987 Sep 26;2(8561):711–4. Morris AJ, Wasson LA, MacKenzie JF. Small bowel enteroscopy in undiagnosed gastrointestinal blood loss. Gut. 1992 Jul;33(7):887–9. Bjarnason I, Macpherson AJ. Intestinal toxicity of non-steroidal anti-inflammatory drugs. Pharmacol Ther. 1994 Apr–May;62(1–2):145–57. Hayllar J, Smith T, Macpherson A, Price AB, Gumpel M, Bjarnason I. Nonsteroidal antiinflammatory drug-induced small intestinal inflammation and blood loss. Effects of sulfasalazine and other disease-modifying antirheumatic drugs. Arthritis Rheum. 1994 Aug;37(8):1146–50. Lanas A, Goldstein JL, Chan FKL, Wilcox CM, Peura DA, Li C, et al. Risk factors associated with a decrease ≥2 g/dL in haemoglobin and/or ≥10% haematocrit in osteoarthritis patients taking celecoxib or a nonselective NSAID plus a PPI in a large randomised controlled trial (CONDOR). Aliment Pharmacol Ther. 2012 Sep;36(5):485–92. Filaretova LP, Bagaeva TR, Morozova OY, Zelena D. The healing of NSAID-induced gastric lesion may be followed by small intestinal and cardiovascular side effects. J Physiol Pharmacol. 2011 Dec;62(6):619–25. Tibble JA, Sigthorsson G, Foster R, Scott D, Fagerhol MK, Roseth A, et al. High prevalence of NSAID enteropathy as shown by a simple faecal test. Gut. 1999 Sep;45(3):362–6. Utzeri E, Usai P. Role of non-steroidal anti-inflammatory drugs on intestinal permeability and nonalcoholic fatty liver disease. World J Gastroenterol. 2017 Jun 14;23(22):3954–63. Bjarnason I, Rainsford KD. NSAID-enteropathy and intestinal microbes. Inflammopharmacology. 2021 Feb;29(1):1–4. Shi S, Wang H, Gao H, Li Z, Chen FX, Zuo XL, et al. Increased gap density predicts weakness of the epithelial barrier in vivo by confocal laser endomicroscopy in indomethacin-induced enteropathy. Dig Dis Sci. 2014 Jul;59(7):1398–405. Robert A, Asano T. Resistance of germfree rats to indomethacin-induced intestinal lesions. Prostaglandins. 1977 Aug;14(2):333–41. Hagiwara M, Kataoka K, Arimochi H, Kuwahara T, Ohnishi Y. Role of unbalanced growth of gram-negative bacteria in ileal ulcer formation in rats treated with a nonsteroidal anti-inflammatory drug. J Med Invest. 2004 Feb;51(1–2):43–51. Watanabe T, Higuchi K, Kobata A, Nishio H, Tanigawa T, Shiba M, et al. Non-steroidal anti-inflammatory drug-induced small intestinal damage is Toll-like receptor 4 dependent. Gut. 2008 Feb;57(2):181–7. Terán-Ventura E, Aguilera M, Vergara P, Martínez V. Specific changes of gut commensal microbiota and TLRs during indomethacin-induced acute intestinal inflammation in rats. J Crohns Colitis. 2014 Sep;8(9):1043–54. Mayo SA, Song YK, Cruz MR, Phan TM, Singh KV, Garsin DA, et al. Indomethacin injury to the rat small intestine is dependent upon biliary secretion and is associated with overgrowth of enterococci. Physiol Rep. 2016 Mar;4(6):e12725. Kinouchi T, Kataoka K, Bing SR, Nakayama H, Uejima M, Shimono K, et al. Culture supernatants of Lactobacillus acidophilus and Bifidobacterium adolescentis repress ileal ulcer formation in rats treated with a nonsteroidal antiinflammatory drug by suppressing unbalanced growth of aerobic bacteria and lipid peroxidation. Microbiol Immunol. 1998;42(5):347–55. Endo H, Higurashi T, Hosono K, Sakai E, Sekino Y, Iida H, et al. Efficacy of Lactobacillus casei treatment on small bowel injury in chronic low-dose aspirin users: a pilot randomized controlled study. J Gastroenterol. 2011 Jul;46(7):894–905. Mortensen B, Murphy C, O’Grady J, Lucey M, Elsafi G, Barry L, et al. Bifidobacteriumbreve Bif195 protects against small-intestinal damage caused by acetylsalicylic acid in healthy volunteers. Gastroenterology. 2019 Sep;157(3):637–46.e4. Article / Publication Details

Received: January 23, 2022
Accepted: February 20, 2023
Published online: April 06, 2023

Number of Print Pages: 15
Number of Figures: 6
Number of Tables: 0

ISSN: 0031-7012 (Print)
eISSN: 1423-0313 (Online)

For additional information: https://www.karger.com/PHA

Copyright / Drug Dosage / Disclaimer Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.

留言 (0)

沒有登入
gif