Yıl: 2022 Cilt: 54 Sayı: 1 Sayfa Aralığı: 120 - 126 Metin Dili: İngilizce DOI: 10.5152/eurasianjmed.2022.22312 İndeks Tarihi: 24-05-2023

Histopathological Overview of Experimental Ulcer Models

Öz:
Histopathology is the process of examining tissue that includes all the changes, when a diseased tissue shows compared to a healthy group with a result of a histological observation. Histopathology has become an essential process in medical experimental research and medical experimental models. Scientists have developed medical experimental animal models for these reasons and have pioneered new drug research for many years. One of these experimental researches is experimental ulcer models. This model, which was initially a single method, has led to the emergence of new models with the discovery of physiological processes on ulcers by scientists. Nowadays, researchers have performed many new peptic ulcer models on experimental animals over the years. The main point in the creation of the ulcer model is the increase in the stomach acid level and the removal or corruption of the gastric mucus. When the experimental models were examined histopathologically, it was seen that the most severe models were those induced by pyloric ligation, acetic acid application, and indomethacin. In these models, ulcer foci that progressed to the submucosa were common, while the superficial damage spreading to the entire surface was striking in the ethanol model. While epithelial losses are shown on the surface of the mucosa, foci of necrotic apoptotic cell clusters extending to the submucosa are shown according to the weight of the model. In addition, evidence of inflammation has been shared in almost all studies. All these results show that ulcer models can be created by many different mechanisms. However, similar findings were observed in almost all experiments. Whether the experimental model caused severe or mild ulceration changed the histological findings.
Anahtar Kelime:

Belge Türü: Makale Makale Türü: Derleme Erişim Türü: Erişime Açık
  • 1. Alturkistani HA, Tashkandi FM, Mohammedsaleh ZM. Histological stains: a literature review and case study. Glob J Health Sci. 2015;8(3):72-79. [CrossRef]
  • 2. Gurcan MN, Boucheron LE, Can A, Madabhushi A, Rajpoot NM, Yener B. Histopathological image analysis: a review. IEEE Rev Biomed Eng. 2009;2:147-171. [CrossRef]
  • 3. Mertz HR, Walsh JH. Peptic ulcer pathophysiology. Med Clin North Am. 1991;75(4):799-814. [CrossRef]
  • 4. Narayanan M, Reddy KM, Marsicano E. Peptic ulcer disease and Helicobacter pylori infection. Mo Med. 2018;115(3):219-224.
  • 5. Drini M. Peptic ulcer disease and non-steroidal anti-inflammatory drugs. Aust Prescr. 2017;40(3):91-93. [CrossRef]
  • 6. Kuna L, Jakab J, Smolic R, Raguz-Lucic N, Vcev A, Smolic M. Peptic ulcer disease: a brief review of conventional therapy and herbal treatment options. J Clin Med. 2019;8(2). [CrossRef]
  • 7. Beiranvand M. A review of the most common in vivo models of stomach ulcers and natural and synthetic anti-ulcer compounds: a comparative systematic study. Phytomed Plus. 2022;2(2):100264. [CrossRef]
  • 8. Lanas A, Chan FKL. Peptic ulcer disease. Lancet. 2017;390(10094):613-624. [CrossRef]
  • 9. Ramakrishnan K, Salinas RC. Peptic ulcer disease. Am Fam Physician. 2007;76(7):1005-1012.
  • 10. Kılıçarslan H, Kalyon S, Yenice N, Etyopatogenezi PÜ. Okmeydanı Tıp Derg. 2011;27(2):65-69. [CrossRef]
  • 11. Malfertheiner P, Chan FK, McColl KE. Peptic ulcer disease. Lancet. 2009;374(9699):1449- 1461. [CrossRef]
  • 12. Sonnenberg A. Review article: historic changes of Helicobacter pylori-associated diseases. Aliment Pharmacol Ther. 2013;38(4):329-342. [CrossRef]
  • 13. Sonnenberg A. Time trends of ulcer mortality in Europe. Gastroenterology. 2007;132(7):2320- 2327. [CrossRef]
  • 14. Lanas A, García-Rodríguez LA, Polo-Tomás M, et al. The changing face of hospitalisation due to gastrointestinal bleeding and perforation. Aliment Pharmacol Ther. 2011;33(5):585-591. [CrossRef]
  • 15. Malmi H, Kautiainen H, Virta LJ, Färkkilä N, Koskenpato J, Färkkilä MA. Incidence and complications of peptic ulcer disease requiring hospitalisation have markedly decreased in Finland. Aliment Pharmacol Ther. 2014;39(5):496-506. [CrossRef]
  • 16. Albayrak A, Polat B, Cadirci E, et al. Gastric antiulcerative and anti-inflammatory activity of metyrosine in rats. Pharmacol Rep. 2010;62(1):113-119. [CrossRef]
  • 17. Kangwan N, Park JM, Kim EH, Hahm KB. Quality of healing of gastric ulcers: natural products beyond acid suppression. World J Gastrointest Pathophysiol. 2014;5(1):40-47. [CrossRef]
  • 18. Saxena B, Singh S. Investigations on gastroprotective effect of citalopram, an antidepressant drug against stress and pyloric ligation induced ulcers. Pharmacol Rep. 2011;63(6):1413-1426. [CrossRef]
  • 19. Chan FK, Leung WK. Peptic-ulcer disease. Lancet. 2002;360(9337):933-941. [CrossRef]
  • 20. Kurata JH, Nogawa AN. Meta-analysis of risk factors for peptic ulcer. Nonsteroidal antiinflammatory drugs, Helicobacter pylori, and smoking. J Clin Gastroenterol. 1997;24(1):2-17. [CrossRef]
  • 21. Salih BA. Helicobacter pylori infection in developing countries: the burden for how long? Saudi J Gastroenterol Off J Saudi Gastroenterol Assoc. 2009;15(3):201-207. [CrossRef]
  • 22. Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes VE. Effect of Helicobacter pylori on gastric epithelial cells. World J Gastroenterol. 2014;20(36):12767-12780. [CrossRef]
  • 23. Graham DY, Miftahussurur M. Helicobacter pylori urease for diagnosis of Helicobacter pylori infection: a mini review. J Adv Res. 2018;13:51-57. [CrossRef]
  • 24. Kao CY, Sheu BS, Wu JJ. Helicobacter pylori infection: an overview of bacterial virulence factors and pathogenesis. Biomed J. 2016;39(1):14-23. [CrossRef]
  • 25. Borekci B, Cadirci E, Albayrak A, Suleyman H, Halici Z, Kadanali S. Effects of progesterone on FSH-stimulated indomethacin ulcers in rats. Fertil Steril. 2008;90(5):1899-1903. [CrossRef]
  • 26. Whittle BJ. Gastrointestinal effects of nonsteroidal anti-inflammatory drugs. Fundam Clin Pharmacol. 2003;17(3):301-313. [CrossRef]
  • 27. Banoob DW, McCloskey WW, Webster W. Risk of gastric injury with enteric- versus nonentericcoated aspirin. Ann Pharmacother. 2002;36(1):163- 166. [CrossRef]
  • 28. Sokić-Milutinović A, Krstić M, Popović D, Mijalković N, Djuranović S, Culafić Dj. Role of Helicobacter pylori infection and use of NSAIDs in the etiopathogenesis of upper gastrointestinal bleeding. Acta Chir IugoSlavica. 2007;54(1):51-62. [CrossRef]
  • 29. Darling RL, Romero JJ, Dial EJ, Akunda JK, Langenbach R, Lichtenberger LM. The effects of aspirin on gastric mucosal integrity, surface hydrophobicity, and prostaglandin metabolism in cyclooxygenase knockout mice. Gastroenterology. 2004;127(1):94-104. [CrossRef]
  • 30. Levine RA, Nandi J, King RL. Nonsalicylate nonsteroidal antiinflammatory drugs augment prestimulated acid secretion in rabbit parietal cells. Investigation of the mechanisms of action. Gastroenterology. 1991;101(3):756-765. [CrossRef]
  • 31. Tolbert MK. Gastroprotective therapy. Vet Clin North Am Small Anim Pract. 2021;51(1):33-41. [CrossRef]
  • 32. Yoshikawa T, Ueda S, Naito Y, et al. Role of oxygen-derived free radicals in gastric mucosal injury induced by ischemia or ischemia-reperfusion in rats. Free Radic Res Commun. 1989;7(3- 6):285-291. [CrossRef]
  • 33. Ayala A, Muñoz MF, Argüelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy- 2-nonenal. Oxid Med Cell Longev. 2014;2014:360438. [CrossRef]
  • 34. Guzmán-Gómez O, García-Rodríguez RV, Quevedo-Corona L, et al. Amelioration of ethanol- induced gastric ulcers in rats pretreated with phycobiliproteins of Arthrospira (Spirulina) maxima. Nutrients. 2018;10(6). [CrossRef]
  • 35. Giordano OS, Guerreiro E, Pestchanker MJ, Guzman J, Pastor D, Guardia T. The gastric cytoprotective effect of several sesquiterpene lactones. J Nat Prod. 1990;53(4):803-809. [CrossRef]
  • 36. Shim YK, Kim N. The effect of H(2) receptor antagonist in acid inhibition and its clinical efficacy. Korean J Gastroenterol. 2017;70(1):4-12. [CrossRef]
  • 37. Howden CW. Use of proton-pump inhibitors in complicated ulcer disease and upper gastrointestinal tract bleeding. Am J Health Syst Pharm. 1999;56(23):S5-S11. [CrossRef]
  • 38. Walan A. Antacids and anticholinergics in the treatment of duodenal ulcer. Clin Gastroenterol. 1984;13(2):473-499. [CrossRef]
  • 39. Kivilaakso E. Pathogenetic mechanisms in experimental gastric stress ulceration. Scand J Gastroenterol Suppl. 1985;110:57-62. [CrossRef]
  • 40. Adinortey MB, Ansah C, Galyuon I, Nyarko A. In vivo models used for evaluation of potential antigastroduodenal ulcer agents. Ulcers. 2013;2013:796405. [CrossRef]
  • 41. Takagi K, Okabe S, Saziki R. A new method for the production of chronic gastric ulcer in rats and the effect of several drugs on its healing. Jpn J Pharmacol. 1969;19(3):418-426. [CrossRef]
  • 42. Okabe S, Pfeiffer CJ. Chronicity of acetic acid ulcer in the rat stomach. Am J Dig Dis. 1972;17(7):619-629. [CrossRef]
  • 43. Okabe S, Roth JL, Pfeiffer CJ. A method for experimental, penetrating gastric and duodenal ulcers in rats. Observations on normal healing. Am J Dig Dis. 1971;16(3):277-284. [CrossRef]
  • 44. Okabe S, Amagase K. An overview of acetic acid ulcer models--the history and state of the art of peptic ulcer research. Biol Pharm Bull. 2005;28(8):1321-1341. [CrossRef]
  • 45. Kolgazi M, Ozdemir-Kumral ZN, Cantali- Ozturk C, et al. Anti-inflammatory effects of nesfatin-1 on acetic acid-induced gastric ulcer in rats: involvement of cyclo-oxygenase pathway. J Physiol Pharmacol. 2017;68(5):765-777.
  • 46. Zhao X, Li J, Meng Y, Cao M, Wang J. Treatment effects of Jinlingzi powder and its extractive components on gastric ulcer induced by acetic acid in rats. Evid Based Complement Alternat Med. 2019;2019:7365841. [CrossRef]
  • 47. Wang L, Wang X, Zhang SL, et al. Gastroprotective effect of palmatine against acetic acidinduced gastric ulcers in rats. J Nat Med. 2017;71(1):257-264. [CrossRef]
  • 48. Lu YF, Zhang XX, Zhao G, Zhu QH. Gastroduodenal ulcer treated by pylorus and pyloric vaguspreserving gastrectomy. World J Gastroenterol. 1999;5(2):156-159. [CrossRef]
  • 49. Monteiro KM, Spindola HM, Possenti A, et al. Characterization of a refinement of the “pylorus ligation” model of rat gastric ulceration resulting in “no pain” and a more specific pharmacological response. J Pharmacol Toxicol Methods. 2013;67(2):121-128. [CrossRef]
  • 50. Wang XY, Yin JY, Zhao MM, Liu SY, Nie SP, Xie MY. Gastroprotective activity of polysaccharide from Hericium erinaceus against ethanolinduced gastric mucosal lesion and pylorus ligation-induced gastric ulcer, and its antioxidant activities. Carbohydr Polym. 2018;186:100-109. [CrossRef]
  • 51. Al-Gabri N, Elnagar GM, Saghir SAM, et al. Preliminary study of gastroprotective effect of Aloe perryi and date palm extracts on pyloric ligationinduced gastric ulcer in experimental rats. BioMed Res Int. 2022;2022:9246785. [CrossRef]
  • 52. Cadirci E, Suleyman H, Aksoy H, et al. Effects of Onosma armeniacum root extract on ethanolinduced oxidative stress in stomach tissue of rats. Chem Biol Interact. 2007;170(1):40-48. [CrossRef]
  • 53. Marotta F, Tajiri H, Safran P, Fesce E, Ideo G. Ethanol-related gastric mucosal damage: evidence of a free radical-mediated mechanism and beneficial effect of oral supplementation with bionormalizer, a novel natural antioxidant. Digestion. 1999;60(6):538-543. [CrossRef]
  • 54. Repetto MG, Llesuy SF. Antioxidant properties of natural compounds used in popular medicine for gastric ulcers. Braz J Med Biol Res. 2002;35(5):523-534. [CrossRef]
  • 55. Nordmann R. Alcohol and antioxidant systems. Alcohol Alcohol. 1994;29(5):513-522.
  • 56. Ibrahim IA, Abdulla MA, Hajrezaie M, et al. The gastroprotective effects of hydroalcoholic extract of Monolluma quadrangula against ethanol- induced gastric mucosal injuries in Sprague Dawley rats. Drug Des Dev Ther. 2016;10:93-105. [CrossRef]
  • 57. Sistani Karampour N, Arzi A, Rezaie A, Pashmforoosh M, Kordi F. Gastroprotective effect of zingerone on ethanol-induced gastric ulcers in rats. Medicina (Kaunas). 2019;55(3). [CrossRef]
  • 58. Mousa AM, El-Sammad NM, Hassan SK, et al. Antiulcerogenic effect of Cuphea ignea extract against ethanol-induced gastric ulcer in rats. BMC Complement Altern Med. 2019;19(1):345. [CrossRef]
  • 59. Guler MC, Tanyeli A, Eraslan E, et al. Persimmon (Diospyros kaki alleviates ethanol-induced gastric ulcer in rats/persimmon (Diospyros kaki L.) sicanlarda etanol ile induklenen Mide Ulserini Hafifletir. Southern Clinics of Istanbul Eurasia (SCIE). 2021;32(1):1-8.
  • 60. Tanyeli A, Eraslan E, Polat E, Bal T. Protective effect of salusin-alpha and salusin-beta against ethanolinduced gastric ulcer in rats. J Basic Clin Physiol Pharmacol. 2017;28(6):623-630. [CrossRef]
  • 61. Demirbilek S, Gürses I, Sezgin N, Karaman A, Gürbüz N. Protective effect of polyunsaturated phosphatidylcholine pretreatment on stress ulcer formation in rats. J Pediatr Surg. 2004;39(1):57-62. [CrossRef]
  • 62. Brodie DA, Hanson HM. A study of the factors involved in the production of gastric ulcers by the restraint technique. Gastroenterology. 1960;38(3):353-360. [CrossRef]
  • 63. Guo S, Gao Q, Jiao Q, Hao W, Gao X, Cao JM. Gastric mucosal damage in water immersion stress: mechanism and prevention with GHRP-6. World J Gastroenterol. 2012;18(24):3145-3155. [CrossRef]
  • 64. Konturek PC, Brzozowski T, Kania J, et al. Pioglitazone, a specific ligand of peroxisome proliferator- activated receptor-gamma, accelerates gastric ulcer healing in rat. Eur J Pharmacol. 2003;472(3):213-220. [CrossRef]
  • 65. Kitagawa H, Fujiwara M, Osumi Y. Effects of water-immersion stress on gastric secretion and mucosal blood flow in rats. Gastroenterology. 1979;77(2):298-302. [CrossRef]
  • 66. Guth PH. Gastric blood flow in restraint stress. Am J Dig Dis. 1972;17(9):807-813. [CrossRef] 67. Liu X, Chen Z, Mao N, Xie Y. The protective of hydrogen on stress-induced gastric ulceration. Int Immunopharmacol. 2012;13(2):197- 203. [CrossRef]
  • 68. Morsy MA, Heeba GH, Abdelwahab SA, Rofaeil RR. Protective effects of nebivolol against cold restraint stress-induced gastric ulcer in rats: role of NO, HO-1, and COX-1,2. Nitric Oxide. 2012;27(2):117-122. [CrossRef]
  • 69. Dengiz GO, Odabasoglu F, Halici Z, Suleyman H, Cadirci E, Bayir Y. Gastroprotective and antioxidant effects of amiodarone on indomethacininduced gastric ulcers in rats. Arch Pharm Res. 2007;30(11):1426-1434. [CrossRef]
  • 70. Halici Z, Polat B, Cadirci E, et al. Inhibiting renin angiotensin system in rate limiting step by aliskiren as a new approach for preventing indomethacin induced gastric ulcers. Chem Biol Interact. 2016;258:266-275. [CrossRef]
  • 71. Atalay F, Odabasoglu F, Halici M, et al. Gastroprotective and antioxidant effects of Lobaria pulmonaria and its metabolite rhizonyl alcohol on indomethacin-induced gastric ulcer. Chem Biodivers. 2015;12(11):1756-1767. [CrossRef]
  • 72. Rainsford KD. The effects of 5-lipoxygenase inhibitors and leukotriene antagonists on the development of gastric lesions induced by nonsteroidal antiinflammatory drugs in mice. Agents Actions. 1987;21(3-4):316-319. [CrossRef]
  • 73. Hayllar J, Bjarnason I. NSAIDs, Cox-2 inhibitors, and the gut. Lancet. 1995;346(8974):521-522. [CrossRef]
  • 74. Ugan RA, Un H. The protective roles of butein on indomethacin induced gastric ulcer in mice. Eurasian J Med. 2020;52(3):265-270. [CrossRef]
  • 75. Suleyman H, Albayrak A, Bilici M, Cadirci E, Halici Z. Different mechanisms in formation and prevention of indomethacin-induced gastric ulcers. Inflammation. 2010;33(4):224-234. [CrossRef]
  • 76. Borekci B, Kumtepe Y, Karaca M, et al. Role of alpha-2 adrenergic receptors in anti-ulcer effect mechanism of estrogen and luteinising hormone on rats. Gynecol Endocrinol. 2009;25(4):264-268. [CrossRef]
  • 77. Ugan RA, Un H, Kose D, et al. Can aprepitant used for nausea and vomiting be good gastrointestinal complaints? Naunyn-Schmiedebergs Arch Pharmacol. 2020;393(12):2463-2472. [CrossRef]
  • 78. Suleyman H, Cadirci E, Albayrak A, et al. Comparative study on the gastroprotective potential of some antidepressants in indomethacininduced ulcer in rats. Chem Biol Interact. 2009;180(2):318-324. [CrossRef]
  • 79. Karaoglan ES, Bayir Y, Albayrak A, et al. Isolation of major compounds and gastroprotective activity of Alchemilla caucasica on indomethacin induced gastric ulcers in rats. Eurasian J Med. 2020;52(3):249-253. [CrossRef]
  • 80. Wallace JL, McKnight W, Reuter BK, Vergnolle N. NSAID-induced gastric damage in rats: requirement for inhibition of both cyclooxygenase 1 and 2. Gastroenterology. 2000;119(3):706-714. [CrossRef]
  • 81. Lamarque D. Pathogenesis of gastroduodenal lesions induced by non-steroidal anti-inflammatory drugs. Gastroenterol Clin Biol. 2004;28(3):C18- C26. [CrossRef]
  • 82. Dengiz GO, Odabasoglu F, Halici Z, Cadirci E, Suleyman H. Gastroprotective and antioxidant effects of montelukast on indomethacin-induced gastric ulcer in rats. J Pharmacol Sci. 2007;105(1):94-102. [CrossRef]
  • 83. Suleyman H, Halici Z, Cadirci E, Hacimuftuoglu A, Keles S, Gocer F. Indirect role of alpha2- adrenoreceptors in anti-ulcer effect mechanism of nimesulide in rats. Naunyn-Schmiedebergs Arch Pharmacol. 2007;375(3):189-198. [CrossRef]
  • 84. Eraslan E, Tanyeli A, Güler MC, Kurt N, Yetim Z. Agomelatine prevents indomethacin-induced gastric ulcer in rats. Pharmacol Rep. 2020;72(4):984-991. [CrossRef]
  • 85. Toktay E, Yayla M, Sahin L, et al. The effects of dragon fruit (Hylocereus polyrhizus). J Food Biochem. 2022;46(9):e14274. [CrossRef]
  • 86. Koc K, Cerig S, Ucar S, et al. Gastroprotective effects of oleuropein and thymol on indomethacin- induced gastric ulcer in Sprague-Dawley rats. Drug Chem Toxicol. 2020;43(5):441-453. [CrossRef]
APA TOKTAY E, JALE S (2022). Histopathological Overview of Experimental Ulcer Models. , 120 - 126. 10.5152/eurasianjmed.2022.22312
Chicago TOKTAY Erdem,JALE SELLİ Histopathological Overview of Experimental Ulcer Models. (2022): 120 - 126. 10.5152/eurasianjmed.2022.22312
MLA TOKTAY Erdem,JALE SELLİ Histopathological Overview of Experimental Ulcer Models. , 2022, ss.120 - 126. 10.5152/eurasianjmed.2022.22312
AMA TOKTAY E,JALE S Histopathological Overview of Experimental Ulcer Models. . 2022; 120 - 126. 10.5152/eurasianjmed.2022.22312
Vancouver TOKTAY E,JALE S Histopathological Overview of Experimental Ulcer Models. . 2022; 120 - 126. 10.5152/eurasianjmed.2022.22312
IEEE TOKTAY E,JALE S "Histopathological Overview of Experimental Ulcer Models." , ss.120 - 126, 2022. 10.5152/eurasianjmed.2022.22312
ISNAD TOKTAY, Erdem - JALE, SELLİ. "Histopathological Overview of Experimental Ulcer Models". (2022), 120-126. https://doi.org/10.5152/eurasianjmed.2022.22312
APA TOKTAY E, JALE S (2022). Histopathological Overview of Experimental Ulcer Models. Eurasian Journal of Medicine, 54(1), 120 - 126. 10.5152/eurasianjmed.2022.22312
Chicago TOKTAY Erdem,JALE SELLİ Histopathological Overview of Experimental Ulcer Models. Eurasian Journal of Medicine 54, no.1 (2022): 120 - 126. 10.5152/eurasianjmed.2022.22312
MLA TOKTAY Erdem,JALE SELLİ Histopathological Overview of Experimental Ulcer Models. Eurasian Journal of Medicine, vol.54, no.1, 2022, ss.120 - 126. 10.5152/eurasianjmed.2022.22312
AMA TOKTAY E,JALE S Histopathological Overview of Experimental Ulcer Models. Eurasian Journal of Medicine. 2022; 54(1): 120 - 126. 10.5152/eurasianjmed.2022.22312
Vancouver TOKTAY E,JALE S Histopathological Overview of Experimental Ulcer Models. Eurasian Journal of Medicine. 2022; 54(1): 120 - 126. 10.5152/eurasianjmed.2022.22312
IEEE TOKTAY E,JALE S "Histopathological Overview of Experimental Ulcer Models." Eurasian Journal of Medicine, 54, ss.120 - 126, 2022. 10.5152/eurasianjmed.2022.22312
ISNAD TOKTAY, Erdem - JALE, SELLİ. "Histopathological Overview of Experimental Ulcer Models". Eurasian Journal of Medicine 54/1 (2022), 120-126. https://doi.org/10.5152/eurasianjmed.2022.22312