Submission of the manuscript is online via e-mail
ecgarticle@gmail.com or
cholerez@mail.ru

Tel: +7 903 250 5288

Editorial Correspondence e-mail
gastrossr@gmail.com


Publishing, Subscriptions, Sales and Advertising, Correspondence e-mail
journal@cniig.ru

Tel: +7 917 561 9505

SCImago Journal & Country Rank

    1. Russian Medical Academy of Continuous Professional Education (Moscow, Russian Federation)
    2. State Scientific Centre of Coloproctology (Moscow, Russian Federation)
    3. NRC Institute of Immunology FMBA of Russia (Moscow, Russian Federation)

    Keywords:ulcerative colitis, Crohn’s disease, microbiome

    Abstract:The review deals modern ideas about state of microbiome in healthy people and its change in inflammatory bowel disease.

      1. Atlas. Vospalitel’nye zabolevaniya kishechnika: diagnostika i lechenie / pod red. prof. Halifa I. L., chlen-korr. RAN Shelygina YU.A. SPb: Gippokrat. - 2017. - 116 S.
      2. Loranskaya I.D., Boldyreva M. N., Lavrent’eva O. A. Sostav mukoznoj mikroflory zheludochno-kishechnogo trakta pri sindrome razdrazhennogo kishechnika. Eksperimental’naya i klinicheskaya gastroehnterologiya. - 2013. - № 3. - S. 15-22.
      3. Pristenochnaya mikroflora kishechnika // I. D. Loranskaya i dr. - M.: Prima Print, 2015. - 100 S.
      4. Chaplin A.V., Rebrikov D. V., Boldyreva M. N. Mikrobiom cheloveka // VESTNIK RGMU. - 2017. - № 2. - S. 5-13.
      5. Al-Mofarreh M.A., Al-Mofleh I. A. Emerging inflammatory bowel disease in Saudi outpatients: a report of 693 cases // Saudi J Gastroenterol. - 2013. - № 19. - Р.16-22.
      6. Ananthakrishnan A. N. Epidemiology and risk factors for IBD // Nat Rev Gastroenterol Hepatol. - 2015. - № 12. - Р. 205-17.
      7. Biasco G., Zannoni U., Paganelli G. M. et al. Folic acid supplementation and cell kinetics of rectal mucosa in patients with ulcerative colitis // Cancer Epidemiol. Biomarkers Prev. - 1997. - № 6. - Р.469-471.
      8. Cho J.H., Brant S. R. Recent insights into the genetics of inflammatory bowel disease // Gastroenterology. - 2011. - № 140. - Р.1704-12.
      9. Clemente J.C., Ursell L. K., Parfrey L. W. et al. The impact of the gut microbiota on human health: an integrative view // Cell. - 2012. - № 148. - Р. 1258-70.
      10. Darfeuille-Michaud A., Boudeau J., Bulois P. et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease // Gastroenterology. - 2004. - № 127. - Р.412-421.
      11. Devkota S., Wang Y., Musch M. W. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice // Nature. - 2012. - № 487(7405). - Р.104-8.
      12. Fanaro S., Chierici R., Guerrini P. et al. Intestinal microflora in early infancy: composition and development // Acta pediat. - 2003. - V. 91(441). - P. 48-55.
      13. Flint H.J., Bayer E. A., Rincon M. T. et al. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis // Nat. Rev. Microbiol. - 2008. - № 6. - Р.121-131.
      14. Floch M. H. Intestinal microecology in health and wellness // J Clin Gastroenterol. - 2011. - № 45. - Р.108-10.
      15. Frank D.N., Amand A. L. St, Feldman R. A. et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases // Proc. Natl. Acad. Sci. U.S.A. - 2007. - № 104. - P. 13780-13785.
      16. Frick J.S., Autenrieth I. B. The gut microflora and its variety of roles in health and disease // Curr Top Microbiol Immunol. - 2013. - № 358. - Р.273-89.
      17. Garrett W.S., Gallini C. A., Yatsunenko T. et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis // Cell Host Microbe. - 2010. - № 8. - Р.292-300.
      18. Geddes K., Rubino S., Streutker C., et al. Nod1 and Nod2 regulation of inflammation in the Salmonella colitis model // Infect. Immun. - 2010. - № 78. - Р.5107-5115.
      19. Hugot J.P., Chamaillard M., Zouali H. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease // Nature. - 2001. - № 411. - Р.599-603.
      20. Kang S., Denman S. E., Morrison M. et al. Dysbiosis of fecal microbiota in Crohn’s disease patients as revealed by a custom phylogenetic microarray // Inflamm. Bowel Dis. - 2010. - № 16. - P. 2034-2042.
      21. Khor B., Gardet A., Xavier R. J. Genetics and pathogenesis of inflammatory bowel disease // Nature. - 2011. - № 474. - Р. 307-17.
      22. Lopez-Siles M., Khan T. M., Duncan S. H. et al. Cultured representatives of two major phylogroups of human colonic Faecalibacterium prausnitzii can utilize pectin, uronic acids, and host-derived substrates for growth // Appl. Environ. Microbiol. - 2012. - № 78. - Р.420-428.
      23. Machiels K., Joossens M., Sabino J. et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis // Gut. - 2014. - № 63. - Р.1275-1283.
      24. Man S.M., Kaakoush N. O., Leach S. T. et al. Host attachment, invasion, and stimulation of proinflammatory cytokines by Campylobacter concisus and other non-Campylobacter jejuni Campylobacter species // J. Infect. Dis. - 2010. - № 202. - Р.1855-1865.
      25. Martinez-Medina M., Garcia-Gil L. J. Escherichia coli in chronic inflammatory bowel diseases: An update on adherent invasive Escherichia coli pathogenicity // World J Gastrointest Pathophysiol. - 2014. - № 5(3). - Р.213-27.
      26. Mazmanian S. K., Round J. L., Kasper D. L. A microbial symbiosis factor prevents intestinal inflammatory disease // Nature. - 2008. - № 453. - Р.620-625.
      27. Morgan X.C., Tickle T. L., Sokol H. et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment // Genome Biol. - 2012. - № 13. - R79.
      28. Mukhopadhya I., Hansen R., El-Omar E.M. et al. IBD-what role do Proteobacteria play? // Nat. Rev. Gastroenterol. Hepatol. - 2012. - № 9. - Р.219-230.
      29. Muyzer G., Stams A. J. The ecology and biotechnology of sulphate-reducing bacteria // Nat. Rev. Microbiol. - 2008. - № 6. - Р.441-454.
      30. Nagao-Kitamoto H., Kamada N. Host-microbial Cross-talk in Inflammatory Bowel Disease // Immune Netw. - 2017. - № 17(1). - Р.1-12.
      31. Ogura Y., Bonen D. K., Inohara N. et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease // Nature. - 2001. - № 411. - Р. 603-6.
      32. Qin J.J., Li R. Q., Raes J. et al. A human gut microbial gene catalogue established by metagenomic sequencing // Nature. - 2010. - № 464. - Р.59-65.
      33. Rehman A., Lepage P., Nolte A. et al. Transcriptional activity of the dominant gut mucosal microbiota in chronic inflammatory bowel disease patients // J. Med. Microbiol. - 2010. - № 59. - Р.1114-1122.
      34. Rowan F., Docherty N. G., Murphy M. et al. Desulfovibrio bacterial species are increased in ulcerative colitis // Dis. Colon Rectum. - 2010. - № 53. - Р.1530-1536.
      35. Ruby T., McLaughlin L., Gopinath S. et al. Salmonella’s long-term relationship with its host // FEMS Microbiol. Rev. - 2012. - № 36. - Р.600-615.
      36. Sender R., Fuchs S., Milo R. Revised estimates for the number of human and bacteria cells in the body // PLoS Biol. - 2016. - 14: e1002533.
      37. Sokol H., Pigneur B., Watterlot L. et al. O. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients // Proc. Natl. Acad. Sci. U.S.A. - 2008. - № 105. - Р.16731-16736.
      38. Sokol H., Seksik P., Rigottier-Gois L. et al. Specificities of the fecal microbiota in inflammatory bowel disease // Inflamm. Bowel Dis. - 2006. - № 12. - Р.106-111.
      39. Sonnenburg J.L., Backhed F. Diet-microbiota interactions as moderators of human metabolism // Nature. - 2016. - № 535. - Р.56-64.
      40. Theriot C.M., Koenigsknecht M. J., Carlson P. E. et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection // Nat. Commun. - 2014. - № 5. - Р.3114.
      41. Wagner J., Short K., Catto-Smith A.G. et al. Identification and characterisation of Pseudomonas 16S ribosomal DNA from ileal biopsies of children with Crohn’s disease // PLoS One. - 2008. - 3: e3578.
      42. Waidmann M., Bechtold O., Frick J. S. et al. Bacteroides vulgatus protects against Escherichia coliinduced colitis in gnotobiotic interleukin-2-deficient mice // Gastroenterology. - 2003. - № 125. - Р.162-177.
      43. Wexler H. M. Bacteroides: the good, the bad, and the nitty-gritty // Clin. Microbiol. Rev. - 2007. - № 20. - Р.593-621.
      44. Wilson J.C., Furlano R. I., Jick S. S. et al. Inflammatory bowel disease and the risk of autoimmune diseases // J Crohns Colitis. - 2016. - № 10. - Р.186-93.
      45. Wong S.H., Ng S. C. What can we learn from inflammatory bowel disease in developing countries? // Curr Gastroenterol Rep. - 2013. - № 15. - Р.313.
      46. Yatsunenko T., Rey F. E., Manary M. J. et al. Human gut microbiome viewed across age and geography // Nature. - 2012. - № 486. - Р.222-7.
     


    Full text is published :
    Loranskaya I.D., Khalif I.L., Boldyreva M.N., Kupaeva V.A. CHARACTERISTIC OF MICROBIOME IN INFLAMMATORY BOWEL DISEASE (REVIEW). Experimental and Clinical Gastroenterology Journal. 2018;153(05):104-111
    Read & Download full text

    1. A. I. Yevdokimov Moscow state university of medicine and dentistry (Moscow, Russian Federation)
    2. Penza state university (Penza, Russian Federation)
    3. Infectious Clinical Hospital № 2 of the Moscow Department of Health care (Moscow, Russian Federation)

    Keywords:HIV-infection, viral hepatitis, remaxol

    Abstract:The aim of the study was to evaluate the effectiveness of remaxol long-term use in HIV-infected patients with chronic viral hepatitis. The results of observation and treatment of 46 HIV-infected patients with diffuse liver damage of a viral etiology (chronic hepatitis C and / or B) were analyzed. Patients were divided into two groups, depending on the treatment regimen. 16 patients of the first group were treated with remaxol (intravenous drip 400 ml daily for 12 days, 2-4 times a year at intervals of 3-6 months) and patients of the second group received standard symptomatic therapy. No specific treatment of HIV was carried out before remaxol prescription. The effectiveness of the therapy was evaluated by clinical, biochemical (liver enzymatic activity) and instrumental (elastography including an assessment of fibrosis levels with METAVIR scale). Inclusion of Remaxol in complex therapy of HIV-infected patients with hepatic damage of viral etiology (CHC and / or CHB) stabilized patients’ general condition. It shows hepatoprotective effect, which is manifested by severity of specific symptoms and enzymatic activity decrease, as well as fibrotic changes in liver tissue. To illustrate this, we describe a clinical case of long-term treatment of a patient using remaxol. The revealed positive effect of remaxol on condition of HIV-infected patients with diffuse liver lesions allows to recommend inclusion of the drug in therapy regimens of these patients.

      1. Zaplutanov V. A. Remaxol: A reference collection of experimental and clinical scientific papers cited in PubMed (a database of medical and biological publications) Zaplutanov V. A., Romantsov M. G., Sukhanov D. S. – SPb, 2012. – 184 s.
      2. Okovityy S. V. Hepatoprotectors/ Okovityy S. V., Bezborodkina N. N., Uleychik S. G., Shulepin S. N.. – M.: GEOTAR-Media, 2010. –112 s.
      3. Remaxol. Solution for infusions: Information about the innovative product for clinicians. – SPb, 2011. – 16 s.
      4. Romancov M.G., Sologub T. V., Goryacheva L.G, Kovalenko S. N., Suhanov D. S. SHul'dyakov A.A., Bondarenko A. N., Kovalenko A. L., Petrov A. Yu. Pathogenetically substantiated, with an assessment of the quality of life, calculation of the risk of the outcome of the disease, therapy for patients with viral hepatitis C Antibiotiki i himioterapiya. 2010; № 3–4: 45–55
      5. Romantsov M. G. Sologub T. V., Goryacheva L. G. Modern approach to adequate therapy of chronic hepatitis. Efficiency of etiotropic and pathogenetic therapy, assessment of quality of life, risk of development of disease outcomes SPb., 2010, – 64 s.
      6. Fedoskina E. A. Treatment of complications of cirrhosis: a method. Rec. For doctors./ Fedoskina E.A Maevskaya M. V., Galimova S. F.; pod red. Ivashkina V. T..; Rossiyskaya gastroenterolog. assots., Rossiyskoye o-vo po izucheniyu pecheni. – M.: 4TE Art, 2009. – 60 s
      7. Yushchuk N. D. Recommendations for the diagnosis and treatment of adult patients with hepatitis B and C. / N. D. Yushchuk [i dr.]. M.: GEOTAR-Media, 2015. 302 s.
      8. Muriel P., Rivera-Espinoza Y. Bentficial drugs for liver diseases J/Appl. Toxicol. 2008; Vol.28 (2): 93–103
      9. Mato J.M., S. C. Lu Role of S-adenosyl-L-methionine in liver health and Hepatology 2007; Vol.45(5): 1306–1312
     


    Full text is published :
    Sundukov A.V., Mel’nikov L.V., Alikeeva G.K., Vdovina E.T., Safiullina N.Kh. HEPATOPROTECTIVE THERAPY FOR DIFFUSION LIVER DISEASES IN HIV-INFECTED PATIENTS. Experimental and Clinical Gastroenterology Journal. 2018;153(05):112-116
    Read & Download full text

    1. FSBI “A. N. Ryzhykh State scientific center of coloproctology” of Ministry of health of Russian Federation
    2. FSBEI FPE RMACPE MOH Russia (Moscow, Russian Federation)
    3. Pirogov Russian National Research Medical University (RNRMU) (Moscow, Russian Federation)

    Keywords: impedance planimetric technique, anal sphincter deficiency, pelvic floor muscles, high resolution anorectal manometry, elasticity, extensibility, intraluminal imaging probe EndoFLIP

    Abstract:The article gives an overview of publications on the use of impedance planimetric method in the study of anorectal function. Anal sphincters normally respond reflexively to the stretching of the rectum by intestinal contents that fill and stretch the rectum. The idea of the method lies in the fact that is artificial stretching of the anal canal with simultaneous recording of pressure and building a virtual geometric shape directly in response to the tension, and calculates a coefficient of elasticity of the anal canal that quantitatively characterize the response of the anal sphincter in tension. For the first time in Russia, it became possible to use this method in coloproctological patients in the State Scietific Centre of Coloproctology. In the article step by step assessed the main uses of the techniques described in the literature: the description of the method and normative values, then - comparison of extensibility of anal sphincters at rest and with voluntary reduction in norm in volunteers and patients with anal incontinence, including patients with scleroderma, then compared the results of this technique with high-resolution 3D manometry. In addition, the use of the Endoflip method as a dynamic control of the treatment of anal sphincter failure by means of sacral stimulation is analyzed.

      1. Шелыгин Ю. А., редактор. Колопроктология/Клинические рекомендации. М.: ГЭОТАР-Медиа; 2015. С. 190-213.
      2. Johanson J. F., Lafferty J. Epidemiology of fecal incontinence: the silent affliction. Am J Gastroenterol. 1996; 91: 33-6.
      3. Фролов С.А., Титов А. Ю., Костарев И. В., Полетов Н. Н., Джанаев Ю. А. Тибиальная нейромодуляция в лечении больных с различными формами недостаточности анального сфинктера. Колопроктология. 2013; 2(44): 37-43.
      4. Whitehead W. E., Borrud L., Goode P. S. et al. Fecal incontinence in US adults: epidemiology and risk factors. Gastroenterology. 2009; 137: 512-7.
      5. Bharucha A. E. Pelvic floor: anatomy and function. Neurogastroenterol Motil. 2006; 18: 507-19.
      6. Sangwan Y. P., Solla J. A. Internal anal sphincter: advances and insights. Dis Colon Rectum. 1998; 41: 1297-311.
      7. Rasmussen O. O., Christiansen J. Physiology and pathophysiology of anal function. Scand J Gastroenterol. Suppl. 1996; 216: 169–74.
      8. Delancey J. O., Toglia M. R., Perucchini D. Internal and external anal sphincter anatomy as it relates to midline obstetric lacerations. Obstet Gynecol. 1997; 90: 924-7.
      9. Bharucha A. E. Anorectal manometry and imaging are necessary in patients with fecal incontinence. Am J Gastroenterol. 2006; 101: 2679-81.
      10. Azpiroz F., Enck P., Whitehead W. E. Anorectal functional testing: review of collective experience. Am J Gastroenterol. 2002; 97: 232-40.
      11. Rao S.S., Hatfield R., Soffer E., Rao S., Beaty J., Conklin J. L. Manometric tests of anorectal function in healthy adults. Am J Gastroenterol. 1999; 94: 773-83.
      12. Eckardt V. F., Kanzler G. How reliable is digital examination for the evaluation of anal sphincter tone? Int J Colorectal Dis. 1993; 8: 95-7.
      13. Harris L. D., Winans C. S., Pope C. E. 2nd. Determination of yield pressures: a method for measuring anal sphincter competence. Gastroenterology. 1966; 50: 754-60.
      14. Kwiatek M. A., Kahrilas P. J., Soper N. J. et al. Esophagogastric junction distensibility after fundoplication assessed with a novel functional luminal imaging probe. J Gastrointest Surg. 2010; 14: 268-76.
      15. Gregersen H., Drewes А. Functional oesophago-gastric junction imaging. World J Gastroenterol. 2006 May 14; 12(18): 2818-2824. www.wjgnet.com World Journal of Gastroenterology ISSN1007-9327
      16. McMahon B., Frøkjær J., Kunwald Р., Liao D., Funch-Jensen Р., Drewes А. et al. The functional lumen imaging probe (FLIP) for evaluation of the esophagogastric junction. Am J Physiol Gastrointest Liver Physiol 292: G377-G384, 2007. First published August 31, 2006; doi:10.1152/ajpgi.00311.2006.
      17. Alqudah M. M., Gregersen H., Drewes A. M., McMahon B. P. Evaluation of anal sphincter resistance and distensibility in healthy controls using EndoFLIP Neurogastroenterol Motil. 2012 Dec; 24(12): 591-9.
      18. Alqudah М., Whelan М., McNamara D., Neary Р., McMahon В. EndoFLIP® to Measure Anal Sphincters Distensibility in Healthy and Faecal Incontinence Gastroenterology. 2014; 146, Issue 5: 715
      19. Luft F., Fynne L., Gregersen H., Lundager F., Buntzen S, Lundby L. et al. Functional luminal imaging probe: a new technique for dynamic evaluation of mechanical properties of the anal canal. Tech Coloproctol. 2012 Aug 31; 16(6): 451-7.
      20. Fynne L., Luft F., Gregersen H., Buntzen S., Lundby L., Lundager F. Distensibility of the anal canal in patients with systemic sclerosis: A study with the Functional Lumen Imaging Probe. Colorectal Dis. 2012 Oct 16. doi: 10.1111/codi.12063.
      21. Gourcerol G., Granier S., Bridoux V., Menard J. F., Ducrotté P., Leroi A. M. Do endoflip assessments of anal sphincter distensibility provide more information on patients with fecal incontinence than high-resolution anal manometry? Neurogastroenterol Motil. 2015 Dec 15. doi: 10.1111/nmo.12740.
      22. Haas S., Liao D., Gregersen H., Lundby L., Laurberg S., Krogh K. Increased yield pressure in the anal canal during sacral nerve stimulation: a pilot study with the functional lumen imaging probe. Neurogastroenterol Motil. 2016 Aug 21; 15: 25-31.
      23. Sorensen G., Liao D., Lundby L., Fynne L., Buntzen S., Gregersen H. et al. Distensibility of the anal canal in patients with idiopathic fecal incontinence: a study with the Functional Lumen Imaging Probe Neurogastroenterol Motil. 2014; 26: 255-263.
      24. Liu J., Guaderrama N., Nager C. W., Pretorius D. H., Master S., Mittal R. K. Functional correlates of anal canal anatomy: puborectalismuscle and anal canal pressure. Am J Gastroenterol. 2006; 101: 1092-1097.
     


    Full text is published :
    Fomenko O.Yu., Shelygin Yu.A., Poryadin G.V., Mudrov A.A. PLANIMETRIC IMPEDANCEOMETRY IN ASSESSMENT OF THE FUNCTIONAL STATE OF ANAL SPHINCATORS. Experimental and Clinical Gastroenterology Journal. 2018;153(05):117-122
    Read & Download full text

    1. Volgograd state medical University (Volgograd, Russian Federation)

    Keywords: whipple’s disease, intestinal lipodystrophy, maladsorption syndrome, maldigestion syndrome, Tropheryma nodes whippeli lumen, polyarthralgia, abdominal pain, polyarthralgia, polyserositis, adrenal insufficiency, endocarditis, antibiotic therapy

    Abstract:This review focuses on the clinical picture, approaches to diagnosis and therapy of Whipple’s disease.

      1. Sparsa L., Fenollar F., Gossec L., Leone J., Pennaforte J. L., Dougados M., Roux C. Whipple disease revealed by anti-TNFα therapy// Rev Med Interne. 2013. Vol. 34 (2). Р. 105-109.
      2. Moos V., Schneider T. Changing paradigms in Whipple’s disease and infection with Tropherymawhipplei // Eur J ClinMicrobiol Infect Dis. 2011. Vol. 30(10).Р.1151-1158.
      3. Lagier J. C., Lepidi H., Raoult D., Fenollar F. Systemic Tropherymawhipplei: clinical presentation of 142 patients with infections diagnosed or confirmed in a reference center // Medicine (Baltimore). 2010. Vol. 89(5).Р.337-345.
      4. Meunier M., Puechal X., Hoppé E., Soubrier M., Dieudé P., Berthelot J. M., Caramaschi P., Gottenberg J. E., Gossec L., Morel J., Maury E., Wipff J., Kahan A., Allanore Y. Rheumatic and musculoskeletal features of Whipple disease: a report of 29 cases // J Rheumatol. 2013. Vol. 40 (12). Р. 2061-2066.
      5. Feurle G. E., Moos V., Schinnerling K., Geelhaar A., Allers K., Biagi F., Bläker H., Moter A., Loddenkemper C., Jansen A., Schneider T. The immune reconstitution inflammatory syndrome in whipple disease: a cohort study // Ann Intern Med. 2010. Vol. 153 (11).Р. 710-717.
      6. Chan V., Wang B., Veinot J. P., Suh K. N., Rose G., Desjardins M., Mesana T. G. Tropherymawhipplei aortic valve endocarditis without systemic Whipple’s disease // Int J Infect Dis. 2011. Vol. 15 (11).Р. 804-806.
      7. Lagier J. C., Fenollar F., Lepidi H., Raoult D. Evidence of lifetime susceptibility to Tropherymawhipplei in patients with Whipple’s disease // J AntimicrobChemother. 2011. Vol. 66 (5). Р. 1188-1189.
      8. Keita A. K., Bassene H., Tall A. et al. Tropherymawhipplei: a common bacterium in rural Senegal. PLoSNegl Trop Dis 2011;5(12): e1403
      9. Bonhomme C. J., Renesto P., Desnues B. et al. Tropherymawhipplei glycosylation in the pathophysiologic profile of Whipple’s disease.J InfectDis 2009;199(7):1043-52.
      10. Parfenov A. I. Bolezni kishechnika: Rukovodstvo dlya vrachej [Diseases of the intestine: A guide for doctors.] - M.: Medicina, 2000. - 613 s.
      11. Maev I. V., Zhilyaev E. V. Bolezn’ Uippla [Whipple’s disease] // Klinicheskaya medicina. 2014. T.18, № 5. S. 2063-2065.
      12. Belov B. S. Bolezn’ Uippla [Whipple’s disease] // Sovremennaya Revmatologiya. 2013. № 1. 12-16 s.
      13. Grebenev A. L., Myagkova L. P. Bolezni kishechnika (sovremennye dostizheniya v diagnostike i terapii) [Bowel diseases (modern advances in diagnosis and therapy)]. - M.: Medicina, 2012. - 130-132 s.
      14. Puéchal X. Whipple’s disease // Ann Rheum Dis. 2013. Vol. 72 (6).Р. 797-803.
      15. Lange U., Teichman J. Whipple arthritis diagnosis by molecular analysis of synovial fluid - current status of diagnosis and therapy // Rheumatology. - 2015. - Vol. 42. - P. 312-313.
      16. Misbah S. A., Mapstone N. P. Whipple’s disease revisited. J. Clin. Pathol. 2014; 53: 750-755.
      17. Geissdörfer W., Moos V., Moter A. et al. High frequency of Tropherymawhipplei in culture-negative endocarditis. J Clin Microbiol 2012; 50(2):216-22.
     


    Full text is published :
    Skvortsov V. V., Tumarenko A. V., Pavlov V. K. DIAGNOSIS AND TREATMENT OF WHIPPLE’S DISEASE. Experimental and Clinical Gastroenterology Journal. 2018;153(05):123-127
    Read & Download full text

    1. North –Western State Medical University named after I. I. Mechnikov (St. Petersburg, Russian Federation)

    Keywords: colon, pathology, mucous membrane

    Abstract:The review contains articles analyzing the structural changes in the epithelium mucous membrane of various parts of the large intestine of humans under normal and some clinical pathology. Analysis of structural changes revealed main characteristics in the dystrophic and regenerative processes developing in epithelium. Changes in the epithelium capture some structures (microvilli, cellular contacts, organelles, nucleus) forming its cells and are nonspecific. They differ in the degree of severity, which depends on the severity of the disease and the individual characteristics of the organism.

      1. Avdeeva TG, Ryabukhin YuV, Parmenova LP, eds. Children's gastroenterology: guide. Moscow, GEOTAR-MEDIA, 2009. 192 p.
      2. Aruin LI, Kapuller LA, Isakov VA Morphological diagnosis of diseases of the stomach and intestines. Moscow, Triada-X, 1998. 496 p.
      3. The report on a condition of health care in Europe in 2009. 2010. 205 p
      4. Ivanova VF, Puzyriov AA, Kostiukevitch SV, Drai RV. Structural changes in rat intestinal wall during starvation. Morfologiia (Saint-Petersburg, Russia), 2009; vol. 136, no. 6, pp. 62–68.
      5. Kvetnoy IM, Yuzhakov VV. Diffusive endocrine system: Guide to histology. SPb, Speclit, 2011. vol. 1, pp 728–750.
      6. Kozlova IV, Osadchuk MA, Kvetnoy IM, eds. Melatonin and serotonin at inflammatory colon diseases and a colorectal cancer. Clinical medicine, 2000; vol. 78, no. 6, pp 32–35.
      7. Komarov FI, Raykhlin NT, Rapoport SI, eds. Syndrome inflammatory bowel diseases. Cliniko-morfologichesky aspects at treatment by Melaksen. Clinical medicine, 2006; vol. 84, no. 11, pp 30–35.
      8. Kostyukevich SV. Differentiation of endocrine epithelial cells of the colon mucosa in humans and some vertebrates. Cytology, 2004; vol. 46, no. 6, pp 506–513.
      9. Kostyukevich SV, Anichkov NM, Ivanova VF, eds. Endocrine cells of rectal epithelium in health, in nonspecific ulcerative colitis and irritable colon syndrome in the treatment with prednisolone and salofalk and in the absence of treatment. Archive of pathology, 2004; vol. 4, pp 23–27.
      10. Kostyukevich SV, Ivanova VF. Colon. Rectum: Guide to histology. SPb, Speclit, 2011. vol. 2, pp 145–152.
      11. Lychkova AE. Serotoninergic regulation of colonic motor function. Therapeutic archive, 2013; vol. 2, pp 89–92.
      12. Makarova M, Rybakova A, Gushchin YA, eds. Anatomical and physiological characteristics of digestive tract in humans and laboratory animals. International bulletin of veterinary science, 2016; vol. 1, pp 82–104.
      13. Markova AA, Kashkina EI. Modern methods of diagnostics and evaluation of the severity of ulcerative colitis. Bulletin of the TGU, 2012; vol. 17, no. 3, pp 915–919.
      14. Osadchuk AM, Osadchuk AM. Morphological and functional upgrading of epithelial cells of the colon and APUD-cells in the pathogenesis and prognosis of ulcerative colitis. Clinical medicine, 2006; vol. 84, no. 12, pp 35–39.
      15. Osadchuk AM, Osadchuk MA, Balashov AV, eds. The role of diffuse endocrine system and cellular renewal of colonocytes in formation of clinical variants of irritable bowel syndrome at persons of young age. Clinical medicine, 2008; vol. 86, no. 3, pp 33–37.
      16. Potekhin PP, Obryadov VP, Lukoyanova GM, eds. The role of morphological research in the diagnosis of ulcerative colitis in children. Clinical medicine, 2010; vol. 2, pp 45–49.
      17. Ham A., Cormac D. Colon: Histology. Moscow, Mir, 1983. vol. 4, pp. 152–158.
      18. Shubnikova EA. Epithelial tissue system. The epithelium of the intestine: Guide to histology. SPb, Speclit, 2011. vol. 1, pp 178–186.
      19. Akiho H., Ihara E., Nakamura K. Low-grade inflammation plays a pivotal role in gastrointestinal dysfunction in irritable bowel syndrome // World Journal Gastrointestinal Pathophysiology. 2010. Vol. 1, № 3. P. 97-105.
      20. Balázs M., Kovács A. Ulcerative colitis: electron microscopic studies with special reference to development of crypt abscesses // Dis. Colon Rectum. 1989. Vol.32, № 4. P. 327-334.
      21. Barrett P., Hobbs R. G., Coats P. J. et al. Endocrine cells of the human gastrointestinal tract have no proliferative capacity // Histochem. J. 1995. Vol. 27. P. 482-486.
      22. Baumgart D.C., Sandborn W. J. Crohn’s disease // The Lancet. 2012. Vol. 380. P. 1590-1605.
      23. Bertrand P.P., Bertrand R. L. Serotonin release and uptake in the gastrointestinal tract //Autonomic Neuroscience: Basic and Clinical. 2010. Vol. 153, № 1-2. P 47-57.
      24. Cheng P., Yao J., Wang C. et al. Molecular and cellular mechanisms of tight junction dysfunction in the irritable bowel syndrome // Molecular Medicine Reports. 2015. Vol. 12, № 3. P. 3257-3264.
      25. Dobbins W.O. 3rd Colonic epithelial cells and polymorphonuclear leukocytes in ulcerative colitis. An electron-microscopic study // Am J. Dig. Dis. 1975. Vol. 20, № 3. P. 236-252.
      26. Dorofeyev A.E., Kiriyan E. A., Vasilenko I. V. et al. Clinical, endoscopical and morphological efficacy of mesalazine in patients with irritable bowel syndrome // Clinical and Experimental Gastroenterology. 2011. Vol. 4. P 141-153.
      27. Dunlop S.P., Jenkins D., Neal K. R., Spiller R. C. Relative importance of enterochromaffin cell hyperplasia, anxiety, and depression in postinfectious IBS // Gastroenterology. 2003. Vol. 125, № 6. P. 1651-1659.
      28. Dvorak A.M., Silen W. Differentiation between Crohn’s disease and other inflammatory conditions by electron microscopy // Ann. Surg. 1985. Vol. 201. P. 53-63.
      29. El-Salhy M., Danielsson A., Stenling R., Grimelius L. Colonic endocrine cells in inflammatory bowel disease // Journal of Internal Medicine. 1997. Vol. 242, № 5. P. 413-419.
      30. Fratila O.C., Craciun C. Ultrastructural evidence of mucosal healing after infliximab in patients with ulcerative colitis // J. Gastrointestin. Liver Dis. 2010. Vol. 19, № 2. P. 147-153
      31. Hsieh S-Y., Shih T. C., Yeh C-Y. et al. Comparative proteomic studies on the pathogenesis of human ulcerative colitis // Proteomics. 2006. Vol. 6, № 19. P. 5322-5331.
      32. Kong W-M., Gong J., Dong L., Chen M-X. Changes in tight junction of intestinal mucosa in patients with irritable bowel syndrome: a study with tracing electron microscope // Nan Fang Yi Ke Da XueXueBao. 2007. Vol. 27, № 8. P. 1167-1170.
      33. Qiao X. T., Ziel J. W., McKimpson W. et al. Prospective identification of a multi-lineage progenitor in murine stomach epithelium // Gastroenterology. 2007. Vol. 133, № 6. P. 1989-1998.
      34. Skinner J. M., Whitehead R., Piris J. Argentaffin cells in ulcerative colitis // Gut. 1971. Vol. 12. P. 636-638.
      35. Solcia E., Greutzfeldt W., Falkmer S. et al. Human gastroenteropancreatic endocrine-paracrine cells: Santa Monica 1980 classification // In: Cellular Basis of Chemical Messengers in the Digestive System, USA, New -York: Academic Press, 1981. Р. 159-165.
      36. Watson A. J., Roy A. D. Paneth cells in the large intestine in ulcerative colitis // J. Path. Bact. 1960. Vol. 80. P. 309-316.
     


    Full text is published :
    Churkova M.L., Kostyukevich S.V. THE EPITHELIUM MUCOSAL OF COLON IN NORMAL AND IN FUNCTIONAL AND INFLAMMATORY BOWEL DISEASES. Experimental and Clinical Gastroenterology Journal. 2018;153(05):128-132
    Read & Download full text

    1. Federal State Budget Educational Institution of Higher Education “Samara State Medical University” of the Ministry of Health of the Russian Federation (Samara, Russian Federation)

    Keywords: Microbiota, non-alcoholic fatty liver disease, non-alcoholic fatty disease of the pancreas, inflammatory bowel disease, irritable bowel syndrome

    Abstract:Aim of the review. Present at the modern level of the development of medicine information on the role of the microbiota of the gastrointestinal tract in the formation of pathology of internal organs and the prospects for correcting the disturbed microbial balance. It is shown that a change in the gastrointestinal microbiota can be associated with various diseases of the internal organs. The most complex issue that needs to be addressed is to determine the coefficient of participation of the bacterial overgrowth factor and the role of individual microbiota in the development of non-alcoholic fatty liver disease, non-alcoholic pancreatic fatty disease, metabolic syndrome, type 2 diabetes mellitus, which is due to the current insufficiency evidence base. It is necessary to conduct large randomized controlled trials on the role of normal gastrointestinal microbiota in the prevention and treatment of these diseases. It seems promising to improve and introduce new methods for restoring the microbiota of the gastrointestinal tract by creating preparations containing bacteriocines aimed at eradication of the target microorganism and vaccines capable of preventing the infection with pathogenic microorganisms.

      1. Qin J., Li R., Raes J., Arumugam M. et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010; 464: 59–65.
      2. Guinane С.M., Cotter P. D. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol. 2013; 6(4): 295–308.
      3. Gumayunova N. G. Detection of excessive growth bacteria syndrome in small intestine with psoriasis disease. Aspirantskij vestnik Povolzh'ja. 2009; № 3–4: 162–4.
      4. Osadchuk A.M., Davydkin I. L., Gricenko T. A., et al. Esophagitis in persons receiving cytostatic and antitumor therapy. The current state of the problem. Izvestiya of the Samara Scientific Center of the Russian Academy of Sciences. 2015. 17 (2–3): 603–610.
      5. Yang I., Woltemate S., Piazuelo M. B. et al. Different gastric microbiota compositions in two human populations with high and low gastric cancer risk in Colombia. Sci Rep. 2016; 6: 18594.
      6. Laniro G., Molina-Infante J., Gasbarrini A. Gastric microbiota. Helicobacter. 2015; 20 (Suppl 1): 68–71.
      7. Dicksved J., Linberg M., Rosenquist M., et al. Molecular characterization of the stomach microbiota in patients with gastric cancer and in controls. J. Med. Microbiol. 2009; 58 (Pt 4): 509–16.
      8. Aviles-Jimenes F., Varquez-Jimenes F., Medrano-Guzman R. et al. Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci Rep. 2014; 4: 4202.
      9. Xiao M., Gao Y., Wang Y. Helicobacter species infection may be associated with cholangiocarcinoma. Int J Clin Pract. 2014; 68: 262–70.
      10. Malfertheiner P., Megraud F., O. Morain C.A. et al. Management of Helicobacter pylori infection – the Maastricht V/Florence Consensus Report. Gut. 2016; 0: 1–25.
      11. Corte C.D., Ferrari F., Villani A., Nobili V. Epidemiology and natural history of NAFLD. J. Med. Biochem. 2015; 34: 13–7.
      12. Romero-Gomez M., Zelber-Sagi S., Trenell M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017; 67(4): 829–846.
      13. Leung C., Rivera L., Furness J. B., Angus PW. The role of the gut microbiota in NAFLD. Nat Rev Gastroenterol Hepatol. 2016; 13(7): 412–25.
      14. Hoy-Schulz Y.E., Jannat K., Roberts T. et al. Safety and acceptability of Lactobacillus reuteri DSM 17938 and Bifidobacterium longum subspecies infantis 35624 in Bangladeshi infants: a phase I randomized clinical trial. BMC Complement Altern Med. 2016; 16:44.
      15. Yilmaz Y., Eren F. Nonalcoholic steatohepatitis and gut microbiota: Future perspectives on probiotics in metabolic liver diseases. Turk J. Gastroenterol. 2017; 28: 327–8.
      16. de Medeiros I. C., de Lima J. G. Nonalcoholic fatty pancreas disease as an endogenous alcoholic fatty pancreas disease. Gastroenterol Hepatol Endosk. 2016; 1(2): 32–39.
      17. Golubeva T.I., Troshina I. A., Medvedeva I. V. Non-alcoholic fatty pancreatic disease in patients with obesity and metabolic syndrome. Universitetskaja medicina Urala. 2017; 2 (9): 50–52.
      18. Simerzin V. V. Fatenkov O. V. Gagloeva I. V., et al. Innovations in the diagnosis and treatment of patients with hypertriglyceridemia. Nauka i innovacii v medicine. 2017; 1(5): 43–51.
      19. Osadchuk A.M., Osadchuk M. A., Kvetnoj I. M. Irritable Bowel Syndrome. 2007; 85(3): 46–50.
      20. Osadchuk M.A., Osadchuk M. M. Irritable Bowel Syndrome and Microbiota: Ways to Optimize Therapy. Vrach. 2015; 56 47–51.
      21. Ghoshal U.C., Shukla R., Ghoshal U. et al. The gut microbiota and irritable bowel syndrome: friend or foe? Int J Inflam. 2012; 2012:151085.
      22. Stojanovic-Stojanovic M., Biagi E., Heilig H. et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011; 141: 1792–1801.
      23. Saulnier D., Riehle K., Mistretta T. et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology. 2011. 141: 1782–1791.
      24. Komarov F.I., Osadchuk A. M., Osadchuk M. A., Kvetnoj I. M. Nonspecific ulcerative colitis. – М.: МИА. – 2008. – 256 с.
      25. Davydova O.E., Katorkin S. E., Ljamin A. V., Andreev P. S. Improving the results of treatment of patients with ulcerative colitis using individual schemes of eradication therapy of opportunistic microflora based on microbiological monitoring. Vrach-aspirant. 2016; 77(4); 49–55.
      26. Li Q., Wang C., Tang C., Li N., Li J. Molecular-phylogenetic characterization of the microbiota in ulcerated and non-ulcerated regions in the patients with Crohn’s disease. PLoS One. 2012; 7: e34939.
      27. Barbut F. Managing antibiotic associated diarrhea. BMJ. 2002; 324 (7350): 1345–1346.
      28. McCoy A., Araujo-Perez F., Azcarate-Peril A. et al. Fusobacterium is associated with colorectal adenomas. PLoS One. 2013; 8(1): e53653.
      29. Arthur J., Perez-Chanona E., Muhlbauer M. et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012; 338: 120–123.
      30. Turnbaugh P., Ridaura V., Faith J. et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009; Nov 11; 1(6): 6ra14.
      31. Qin J., Li Y., Cai Z. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012; 490: 55–60.
      32. Andreasen A., Larsen N., Pedersen-Skovsgaard T. et al. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br J Nutr. 2010; Dec; 104(12):1831–8.
      33. Murphy E., Cotter P., Hogan A. et al. Divergent metabolic outcomes arising from targeted manipulation of the gut microbiota in diet-induced obesity. Gut. 2013; Feb; 62(2):220–6.
      34. Kadooka Y., Sato M., Imaizumi K. et al. Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized controlled trial. Eur J Clin Nutr. 2010; 64: 636–643.
      35. Skvorcov V. V. Dysbiosis of the intestine and antibiotic-associated diarrhea, diagnosis and treatment. Lechashhij vrach. 2008; 2: 43–47.
      36. Kinnear C.L., Strugnell R. A. Vaccination Method Affects Immune Response and Bacterial Growth but Not Protection in the Salmonella Typhimurium Animal Model of Typhoid. PLoS One. 2015; 10(10): e0141356.
      37. Zeng M., Mao X. H., Li J. X. et al. Efficacy, safety, and immunogenicity of an oral recombinant Helicobacter pylori vaccine in children in China: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015 Oct 10; 386(10002): 1457–64.
     


    Full text is published :
    Osadchuk A. M., Davydkin I. L., Gricenko T. A., Lebedeva E. A., Petrushin A. E. THE ROLE OF THE MICROBIOTA OF THE GASTROINTESTINAL TRACT IN THE DEVELOPMENT OF DISEASES OF INTERNAL ORGANS. Experimental and Clinical Gastroenterology Journal. 2018;153(05):133-139
    Read & Download full text

    1. Military Medical Academy named after S. M. Kirov (St. Petersburg, Russian Federation)

    Keywords: inflammatory bowel diseases (IBD), Crohn’s disease (CD), ulcerative colitis (UC), pathogenesis of IBD, imun, genome, exposome, NOD2/CARD15 gene, IL-23R gene, autophagy, T-cell, microbiome, intestinal microbiota, rebiosis

    Abstract:The relevance of inflammatory bowel disease (IBD) is caused by the growth of primary morbidity, as well as the lack of identity of the mechanisms of their pathogenesis. The purpose of this review - to consider the basic and most fundamental laws of genome changes, the immune system, the role and participation of intestinal microflora in the pathogenesis of these diseases. The report is a synthesis of the accumulated information about the relationship of genetic, immunological and exposomes concept, their role and influence on the development of IBD. Results. It is concluded that the pathogenetic cascade of inflammation in IBD begins with changes in microbiota and the appearance of unknown antigens in the intestine, which, in certain genetically determined defects of the immune system, initiate a cascade of inflammatory reactions that form a clinical picture of Crohn’s disease (CD) or ulcerative colitis (UC). The pathogenesis of IBD is unambiguously linked to changes in environmental factors. Conclusion. The most promising targets for therapeutic intervention in the near future to reduce inflammatory reactions will be intestinal microflora and diet.

      1. Belousova EA. Ulcerative colitis and Crohn's disease. – Tver: Publishing House "Triada", 2002. – p.128.
      2. Jonkers D., D. Renders, Masks, A., Pierik M. Probiotics in the treatment of inflammatory bowel disease (a systematic review of intervention studies among adult patients). Gastroenterology and Hepatology, 2013, No. 2, pp. 28–48.
      3. Maev I. V., Andreev D. N., Dichev, D. T., E. V. Giants Crohn's Disease: etiopathogenesis, diagnosis and conservative treatment.//Manual for doctors. – Moscow, 2016. – p.67.
      4. Kirsner JB, Spencer JA. Family occurrences of ulcerative colitis, regional enteritis, and ileocolitis. // Ann. Intern. Med. – 1963. – vol. 59. – pp.133–144.
      5. Hugot J-P, Laurent-Puig P, Gower-Rousseau C et al. Mapping of a susceptibility locus for Crohn’s disease on chromosome 16. Nature, 1996, vol. 379, pp. 821–823.
      6. Cooney R., Baker J., Brain O et al. Nod2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presention. Nat. Med, 2010, vol.16, pp.90–97.
      7. Hooper K., Barlou P., Stevens C., Henderson P. Inflammatory bowel disease drugs: a focus on autophagy. Jornal of Crohn’s and Colitis, 2017, vol.11, № 1, pp. 118–127.
      8. Ogura Y., Bonen D. K., Inohara N. et al. A frameshift mutation in Nod2 associated with susceptibility to Crohn’s desease. Nature, 2001, vol.411, pp. 603–606.
      9. Hugot J-P, Chamaiilard M., Zouali H. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature, 2001, vol.411, pp. 599–603.
      10. Сhassaing B., Vijay-Kumar M., Gewitz A. T. How diet can impact gut microbiota to promote or endanger health. Curr. Opin.Gastroenterolog, 2017, vol.33, № 6, pp. 417–421.
      11. Duerr R. H., Taylor K. D., Brant S. R. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science, 2006, vol. 314, pp. 1461–1463.
      12. Kitahora T, Utsunomiya T, Yokota A. Epidemiological study of ulcerative colitis in Japan: incidence and familial occurrence. The Epidemiology Group of the Research Committee of Inflammatory Bowel Disease in Japan. J. Gastroentero, 1995, vol. 30 (Suppl. 8), pp. 5–8.
      13. Fiocchi С. Inflammatory bowel disease pathogenesis: Where are we? J. Gastroenter. Hepatol, 2015, vol. 30 (Suppl. 1), pp. 12–18.
      14. Brown A., Rampertab C., Mullin G. E. Existing Dietary Guidelines for Crohn’s Disease and Ulcerative Colitis. Expert Rev. Gastroenterol. Hepatol, 2011, vol.5, № 3, pp. 411–425.
      15. Wu G. D., Chen J., Hoffmann C. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science, 2011, vol. 334, pp. 105–108.
      16. Desai M.S, Seekatz A.M, Koropatkin N. M., et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell, 2016, vol, 167, pp.1339–1353.
      17. Hamer H. M., Jonkers D., Venema K. et al. Review article: the role of butyrate on colonic function. Aliment. Pharmacol.Ther, 2008, vol.27, pp. 833–839.
      18. Park J, Kim M, Kang SG, et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol, 2015, vol.8, pp.80–93.
      19. Rambaud Y.C, Buts Y. P., Corthier G., Flourie B. Gut microflora. Digestive physiology and pathology. – Paris, 2006.
      20. Maev I. V., Andreev D. N., Dichev, D. T., E. V. Giants Crohn's Disease: etiopathogenesis, diagnosis and conservative treatment.//Manual for doctors. – Moscow, 2016. – p.67.
      21. Salazar N., Dewulf E. M., Neyrinck A. M., et al. Inulin-type fructans modulate intestinal bifidobacterium species populations and decrease fecal short-chain fatty acids in obese women. ClinNutr, 2015, vol.34, pp. 501–507.
      22. Swidsinski A., Ung V., Sydora BC, et al. Bacterial overgrowth and inflammation of small intestine after carboxymethylcellulose ingestion in genetically susceptible mice. Inflamm. Bowel Dis, 2009, vol.15, pp.359–364.
      23. Chassaing B., Van de Wiele T., De Bodt J., et al. Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut, 2017, vol. 66, pp. 1414–1427.
      24. Roberts C. L., Keita A. V., Duncan S. H., et al. Translocation of Crohn›s disease Escherichia coliacross M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut, 2010, vol. 59, pp.1331–1339.
      25. Chassaing B., Koren O., Goodrich J. K., et al. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature, 2015, vol. 519, pp.92–96.
      26. Yan X., Huang Y., Wang H. et al. Maternal obesity induces sustained inflammation in both fetal and offspring large intestine of sheep. Inflamm. Bowel Dis, 2011, vol. 17, pp. 1513–22.
      27. Gonzalez-Correa C.A., Mulett-Vásquez E., Miranda D. A., et al. The colon revisited or the key to wellness, health and disease. Medical Hypotheses, 2017, Vol. 108, p.133.
      28. Chassaing B., Koren O., Carvalho F. A., et al. AIEC pathobiont instigates chronic colitis in susceptible hosts by altering microbiota composition. Gut, 2013, vol. 63, pp.1069–1080.
      29. Chassaing B., Raja S. M., Lewis J. D., et al. Colonic microbiota encroachment correlates with dysglycemia in humans. Cell MolGastroenterolHepatol, 2017, vol. 4, pp. 205–221.
      30. Faith J. J., Guruge J. L., Charbonneau M., et al. The long-term stability of the human gut microbiota. Science, 2013, vol. 341, p.1237439.
      31. Human Microbiome Project Collaboration Structure, function and diversity of the healthy human microbiome. Nature, 2012, vol. 486, pp.207–214.
      32. Kostic A. D., Gevers D., Siljander H., et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe, 2015, vol.17, pp.260–273.
      33. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med, 2016, vol. 8, p.51.
      34. Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established bymetagenomic sequencing. Nature, 2010, vol. 464, pp.59–65.
      35. Butto LF, Haller D. Dysbiosis in intestinal inflammation: cause or consequence. Int. J. Med. Microbiol, 2016, vol. 306, pp. 302–309.
      36. Wu GD, Compher C, Chen EZ, et al. Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production. Gut, 2014, vol. 65, pp.63–72.
      37. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology, 2009, vol.137, pp.1716–1724.
      38. Filippo C, Cavalieri D, Di Paola M et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci, 2010, vol.107, pp. 14691–14696.
      39. Devkota S, Wang Y, Musch MW et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il 10- mice. Nature, 2012, vol. 487, pp. 104–108.
      40. Chen Y., Blaser M. J. Helicobacter pylori colonization is inversely assosiated with childhood asthma. J. Infect. Dis, 2008, vol. 198, pp.553–560.
      41. Luther J., Dave M., Higgins R. et al. Association between Helicobacter pillory and inflammatory bowel disease: a meta- analysis and systematic review of the literature. Inflam. Bow.Dis, 2010, vol.16, pp.1077–1084.
      42. Luther J., Stephanie Y., Owyang Y. et al. Helicobacter pylori DNA decreases pro-inflammotory cytokine production by dendritic cells and attenuates dextran sulphate-induced cjlitis. Gut, 2011, vol.60, № 11, pp.1479–1486.
      43. Jonkers D., D. Renders, Masks, A., Pierik M. Probiotics in the treatment of inflammatory bowel disease (a systematic review of intervention studies among adult patients). Gastroenterology and Hepatology, 2013, № 2, pp. 28–48.
      44. Borchers A. T., SelmiC., Meyers F. J. et al. Probiotics and immunity. Gastroenterol, 2009, vol. 44, № 1, pp. 26–46.
      45. Pilipenko V. I. Probiotics as signal molecules: Saccharomyces boulardii. Clinical gastroenterology and Hepatology, 2008, vol. 1, № 6, pp. 456–462
      46. Oeschlaeger T. A. Mechanisms of probiotic actions: a review. Int. J. Med. Microbiol, 2010, vol.300, № 1, pp. 57–62.
      47. Karczewski J., Troost F. J., Konings I. et al. Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier. Am. J. Physiol. Gastrointest, 2010, vol.298, № 6, pp. G851-G859.
      48. Mattar A. F., Teitelbaum D. H., Drongowski R. A. et al. Probiotics up-regulate MUC-2 mucin gene expression in a Caco-2 cell-culture model. Pediatr. Surg. Int, 2002, vol.18, № 7, pp. 586–590.
      49. Claes I. J., Keersmaecker S. C., Vanderleyden J. et al. Lessons from probiotic-host interaction stadies in murine models of experimental colitis. Mol. Nutr.Food Res, 2011, vol.55, № 10, pp. 1441–1453.
      50. Guslandi M., Mezzi G., Testoni P. A. Saccaromycesboulardimaitenanse treatment of Crohnsdesease. Dig. Dis. Sci, 2000, vol.11, pp.1462–1464.
      51. Khalili H., Granath F., Smedby K. E. et al. Association Between Long-term Oral Contraceptive Use and Risk of Crohn’s Disease Complications in a Nationwide Study. Gastroenterology, 2016, vol.150, № 7, pp. 1561–1567.
      52. Moayyedi P. Fecal Transplantation: Any Real Hope for Inflammatory Bowel Disease? Curr. Opin.Gastroenterol, 2016, vol. 32, № 4. – pp. 282–286.
     


    Full text is published :
    Pershko A. M., Grinevich V. B., Solovyov I. A., Shotik A. V., Kurilo D. P. PRIVATE THE PATHOGENESIS OF INFLAMMATORY BOWEL DISEASES. Experimental and Clinical Gastroenterology Journal. 2018;153(05):140-149
    Read & Download full text