Increased epithelial permeability syndrome in organic diseases of the upper gastrointestinal tract: systematization of literature data

Cover Page

Cite item

Full Text

Abstract

In 2021, the first Multidisciplinary national consensus was published in Russia. The document provides a comprehensive analysis of pathophysiological, clinical and pharmacotherapeutic aspects of the increased epithelial permeability syndrome (IEPS) as one of the main mechanisms of human diseases. A separate chapter of the consensus devoted to the role of IEPS in organic diseases of the upper gastrointestinal tract demonstrates the importance of this pathophysiological mechanism in the genesis of this group of diseases. This review aims to systematize the literature data on the role of IEPS in organic diseases of the upper gastrointestinal tract, including gastroesophageal reflux disease, chronic gastritis and gastric ulcer.

About the authors

Dmitrii N. Andreev

Yevdokimov Moscow State University of Medicine and Dentistry

Email: dna-mit8@mail.ru
канд. мед. наук, доц. каф. Moscow, Russia

Elena V. Ul'iankina

Yevdokimov Moscow State University of Medicine and Dentistry

канд. мед. наук, ассистент каф. Moscow, Russia

References

  1. Маев И.В., Андреев Д.Н., Дичева Д.Т. Кислотозависимые заболевания. М., 2020
  2. Sleisenger and Fordtran's Gastrointestinaland Liver Disease: Pathophysiology, Diagnosis, Management. Ed. M Feldman, LS Friedman, LJ Brandt. 11th ed. Elsevier, 2020.
  3. Ивашкин В.Т., Маев И.В., Каприн А.Д., и др. Раннее выявление онкологических заболеваний органов пищеварения (методическое руководство Российской гастроэнтерологической ассоциации и Ассоциации онкологов России для врачей первичного звена здравоохранения). Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2019;29(5):53-74
  4. Маев И.В., Самсонов А.А., Андреев Д.Н. Роль и место антацидов в современных алгоритмах терапии кислотозависимых заболеваний. Фарматека. 2013;2:65-72
  5. Spechler SJ. Refractory Gastroesophageal Reflux Disease and Functional Heartburn. Gastrointest Endosc Clin N Am. 2020;30(2):343-59. D0I:10.1016/j.giec.2019.12.003
  6. El-Serag H, Becher A, Jones R. Systematic review: persistent reflux symptoms on proton pump inhibitor therapy in primary care and community studies. Aliment Pharmacol Ther. 2010;32(6):720-37. D0I:10.1111/j.1365-2036.2010.04406.x
  7. Zullo A, Hassan C, De Francesco V, et al. Helicobacter pylori and functional dyspepsia: an unsolved issue? World J Gastroenterol. 2014;20(27):8957-63. doi: 10.3748/wjg.v20.i27.8957
  8. Bockerstett KA, DiPaolo RJ. Regulation of Gastric Carcinogenesis by Inflammatory Cytokines. Cell Mol Gastroenterol Hepatol. 2017;4(1):47-53.
  9. Андреев Д.Н., Заборовский А.В., Лобанова Е.Г. Гастроэзофагеальная рефлюксная болезнь: новые подходы к оптимизации фармакотерапии. Медицинский совет. 2021;5:30-7
  10. Wroblewski LE, Shen L, Ogden S, et al. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology. 2009;136(1):236-46.
  11. Saadat I, Higashi H, Obuse C, et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330-3.
  12. Симаненков В.И., Маев И.В., Ткачева О.Н., и др. Синдром повышенной эпителиальной проницаемости в клинической практике. Мультидисциплинарный национальный консенсус. Кардиоваскулярная терапия и профилактика. 2021;20(1):2758
  13. Ивашкин В.Т., Маев И.В., Трухманов А.С., и др. Рекомендации Российской гастроэнтерологической ассоциации по диагностике и лечению гастроэзофагеальной рефлюксной болезни. Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2020;30(4):70-97
  14. Маев И.В., Бусарова Г.А., Андреев Д.Н. Болезни пищевода. М.: ГЭОТАР-Медиа, 2019
  15. Nirwan JS, Hasan SS, Babar ZU, et al. Global Prevalence and Risk Factors of Gastro-oesophageal Reflux Disease (GORD): Systematic Review with Meta-analysis. Sci Rep. 2020;10(1):5814. doi: 10.1038/s41598-020-62795-1
  16. Cook MB, Corley DA, Murray LJ, et al. Gastroesophageal reflux in relation to adenocarcinomas of the esophagus: a pooled analysis from the Barrett's and Esophageal Adenocarcinoma Consortium (BEACON). PLoS One. 2014;9(7):e103508. doi: 10.1371/journal.pone.0103508
  17. Eusebi LH, Cirota GG, Zagari RM, Ford AC. Global prevalence of Barrett's oesophagus and oesophageal cancer in individuals with gastro-oesophageal reflux: a systematic review and metaanalysis. Gut. 2021;70(3):456-63. doi: 10.1136/gutjnl-2020-321365
  18. Rubenstein JH, Taylor JB. Meta-analysis: the association of oesophageal adenocarcinoma with symptoms of gastro-oesophageal reflux. Aliment Pharmacol Ther. 2010;32(10):1222-7. doi: 10.1111/j.1365-2036.2010.04471.x
  19. Orlando LA, Orlando RC. Dilated intercellular spaces as a marker of GERD. Curr Gastroenterol Rep. 2009;11(3):190-4. doi: 10.1007/s11894-009-0030-6
  20. Dellon ES, Shaheen NJ. Persistent reflux symptoms in the proton pump inhibitor era: the changing face of gastroesophageal reflux disease. Gastroenterology. 2010;139:7-13.e3
  21. Андреев Д.Н., Дичева Д.Т. Нарушение проницаемости слизистой оболочки кишечника как фактор этиопатогенеза функциональных заболеваний желудочно-кишечного тракта. Медицинский совет. 2020;5:87-95
  22. Bonfiglio F, Hysi PG, Ek W, et al. A meta-analysis of reflux genome-wide association studies in 6750 Northern Europeans from the general population. Neurogastroenterol Motil. 2017;29(2). doi: 10.1111/nmo.12923
  23. Маев И.В., Андреев Д.Н., Кучерявый Ю.А, Шабуров Р.И. Современные достижения в лечении гастроэзофагеальной рефлюксной болезни: фокус на эзофагопротек-цию. Терапевтический архив. 2019;91 (8):4-11
  24. Маев И.В., Гуленченко Ю.С., Андреев Д.Н., и др. Дуоденогастроэзофагеальный рефлюкс: клиническое значение и подходы к терапии. Consilium Medicum. 2014;16(8):5-8
  25. Tobey NA, Carson JL, Alkiek RA, Orlando RC. Dilated intercellular spaces: a morphological feature of acid reflux - damaged human esophageal epithelium. Gastroenterology. 1996;111(5):1200-5.
  26. Caviglia R, Ribolsi M, Gentile M, et al. Dilated intercellular spaces and acid reflux at the distal and proximal oesophagus in patients with non-erosive gastro-oesophageal reflux disease. Aliment Pharmacol Ther. 2007;25(5):629-36.
  27. Jovov B, Que J, Tobey NA, et al. Role of E-cadherin in the pathogenesis of gastroesophageal reflux disease. Am J Gastroenterol. 2011;106(6):1039-47. doi: 10.1038/ajg.2011.102
  28. Bjorkman EV, Edebo A, Oltean M, Casselbrant A. Esophageal barrier function and tight junction expression in healthy subjects and patients with gastroesophageal reflux disease: functionality of esophageal mucosa exposed to bile salt and trypsin in vitro. Scand J Gastroenterol. 2013;48(10):1118-26.
  29. Weijenborg PW, Smout AJ, Verseijden C, et al. Hypersensitivity to acid is associated with impaired esophageal mucosal integrity in patients with gastroesophageal reflux disease with and without esophagitis. Am J Physiol Gastrointest Liver Physiol. 2014;307(3):G323-9. doi: 10.1152/ajpgi.00345.2013
  30. Маев И.В., Андреев Д.Н., Самсонов А.А., и др. Эволюция представлений о дефиниции, классификации, диагностике и лечении гастрита, ассоциированного с инфекцией Helicobacter pylori. Фарматека. 2016;6:24-33
  31. Hooi JKY, Lai WY, Ng WK, et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology. 2017;153(2):420-9.
  32. Zamani M, Ebrahimtabar F, Zamani V, et al. Systematic review with meta-analysis: the worldwide prevalence of Helicobacter pylori infection. Aliment Pharmacol Ther. 2018;47(7):868-76.
  33. Lanas A, Chan FK. Peptic ulcer disease. Lancet. 2017;390(10094):613-24.
  34. Ивашкин В.Т., Маев И.В., Царьков П.В., и др. Диагностика и лечение язвенной болезни у взрослых (Клинические рекомендации Российской гастроэнтерологической ассоциации, Российского общества колоректальных хирургов и Российского эндоскопического общества). Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2020;30(1):49-70
  35. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Schistosomes, Liver Flukes and Helicobacter pylori. Lyon (FR): International Agency for Research on Cancer; 1994 (IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, No. 61). Available at: https:// www.ncbi.nlm.nih.gov/books/NBK487770/Accessed: 15.05.2021.
  36. Matos JI, de Sousa HA, Marcos-Pinto R, Dinis-Ribeiro M. Helicobacter pylori CagA and VacA genotypes and gastric phenotype: a meta-analysis. Eur J Gastroenterol Hepatol. 2013;25(12):1431-41. doi: 10.1097/MEG.0b013e328364b53e
  37. Kpoghomou MA, Wang J, Wang T, Jin G. Association of Helicobacter pylori babA2 gene and gastric cancer risk: a meta-analysis. BMC Cancer. 2020;20(1):465. doi: 10.1186/s12885-020-06962-7
  38. Song H, Ekheden IG, Zheng Z, et al. Incidence of gastric cancer among patients with gastric precancerous lesions: observational cohort study in a low risk Western population. BMJ. 2015;351:h3867. doi: 10.1136/bmj.h3867
  39. Hansson LE, Nyren O, Hsing AW, et al. The risk of stomach cancer in patients with gastric or duodenal ulcer disease. N Engl J Med. 1996;335(4):242-9. doi: 10.1056/NEJM199607253350404
  40. Sogaard KK, Farkas DK, Pedersen L, et al. Long-term risk of gastrointestinal cancers in persons with gastric or duodenal ulcers. Cancer Med. 2016;5(6):1341-51. doi: 10.1002/cam4.680
  41. Wroblewski LE, Peek RM Jr. Targeted disruption of the epithelial-barrier by Helicobacter pylori. Cell Commun Signal. 2011;9(1):29.
  42. Suzuki H, Moayyedi P. Helicobacter pylori infection in functional dyspepsia. Nat Rev Gastroenterol Hepatol. 2013;10(3):168-74. doi: 10.1038/nrgastro.2013.9
  43. Pristautz H. Evaluation of the epithelial gastric mucosal barrier in gastric diseases using supravital staining of mucosal biopsies. Wien Med Wochenschr Suppl. 1984;86:1-15.
  44. Sutherland LR, Verhoef M, Wallace JL, et al. A simple, non-invasive marker of gastric damage: sucrose permeability. Lancet. 1994;343(8904):998-1000.
  45. Vera JF, Gotteland M, Chavez E, et al. Sucrose permeability in children with gastric damage and Helicobacter pylori infection. J Pediatr Gastroenterol Nutr. 1997;24(5):506-11.
  46. Zsigmond CS, Hannestad U, Franzen L, et al. Atrophic gastritis is associated with increased sucrose permeability related to chronic inflammation. Digestion. 2005;72(4):201-6.
  47. Kawabata H, Meddings JB, Uchida Y, et al. Sucrose permeability as a means of detecting diseases of the upper digestive tract. J Gastroenterol Hepatol. 1998;13(10):1002-6.
  48. Андреев Д.Н., Кулиева А.К. Механизмы действия ребамипида: систематизация литературных данных. Consilium Medicum. 2020;22(8):41-5
  49. Naito Y, Yoshikawa T. Rebamipide: a gastrointestinal protective drug with pleiotropic activities. Expert Rev Gastroenterol Hepatol. 2010;4(3):261-70.
  50. Gweon TG, Park JH, Kim BW, et al.; Incheon and Western Kyonggi Gastrointestinal Study. Additive Effects of Rebamipide Plus Proton Pump Inhibitors on the Expression of Tight Junction Proteins in a Rat Model of Gastro-Esophageal Reflux Disease. Gut Liver. 2018;12(1):46-50. doi: 10.5009/gnl17078
  51. Suzuki T, Yoshida N, Nakabe N, et al. Prophylactic effect of rebamipide on aspirin-induced gastric lesions and disruption of tight junctional protein zonula occludens-1 distribution. J Pharmacol Sci. 2008;106(3):469-77. doi: 10.1254/jphs.fp0071422
  52. Shim S, Jang HS, Myung HW, et al. Rebamipide ameliorates radiation-induced intestinal injury in a mouse model. Toxicol Appl Pharmacol. 2017;329:40-7. doi: 10.1016/j.taap.2017.05.012
  53. Jang H, Park S, Lee J, et al. Rebamipide alleviates radiation-induced colitis through improvement of goblet cell differentiation in mice. J Gastroenterol Hepatol. 2018;33(4):878-86. doi: 10.1111/jgh.14021
  54. Андреев Д.Н., Маев И.В. Ребамипид: доказательная база применения в гастроэнтерологии. Терапевтический архив. 2020;92(12):97-104
  55. Ивашкин В.Т., Трухманов А.С., Гоник М.И. Применение ребамипида в лечении гастроэзофагеальной рефлюксной болезни. Терапевтический архив. 2020;92(4):98-104
  56. Yoshida N, Kamada K, Tomatsuri N, et al. Management of recurrence of symptoms of gastroesophageal reflux disease: synergistic effect of rebamipide with 15 mg lansoprazole. Dig Dis Sci. 2010;55(12):3393-8. doi: 10.1007/s10620-010-1166-9
  57. Hong SJ, Park SH, Moon JS, et al. The Benefits of Combination Therapy with Esomeprazole and Rebamipide in Symptom Improvement in Reflux Esophagitis: An International Multicenter Study. Gut Liver. 2016;10(6):910-6. doi: 10.5009/gnl15537
  58. Li M, Yin T, Lin B. Rebamipide for chronic gastritis: a meta-analysis. Chinese J Gastroenterol Hepatol. 2015;24:667-73.
  59. Haruma K, Ito M, Kido S, et al. Long-term rebamipide therapy improves Helicobacter pylori-associated chronic gastritis. Dig Dis Sci. 2002;47(4):862-7.
  60. Kamada T, Sato M, Tokutomi T, et al. Rebamipide improves chronic inflammation in the lesser curvature of the corpus after Helicobacter pylori eradication: a multicenter study. Biomed Res Int. 2015;2015:865146.
  61. Andreev DN, Maev IV, Dicheva DT. Efficiency of the Inclusion of Rebamipide in the Eradication Therapy for Helicobacter pylori Infection: Meta-Analysis of Randomized Controlled Studies. J Clin Med. 2019;8(9):1498. doi: 10.3390/jcm8091498
  62. Ивашкин В.Т., Маев И.В., Шептулин А.А., и др. Клинические рекомендации Российской гастроэнтерологической ассоциации и Российского эндоскопического общества по диагностике и лечению гастрита и дуоденита. М., 2021

Copyright (c) 2021 Consilium Medicum

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies