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

№ 03 (139) 2017

  • Уважаемые коллеги !
    Мартовский номер нашего журнала «Экспериментальная и клиническая гастроэнтерология» мы посвятили заболеваниям кишечника.
    По уровню распространенности и заболеваемости воспалительные заболевания кишечника (ВЗК) значительно уступают другим заболеваниям органов пищеварения, но по тяжести течения, частоте осложнений и летальности они во всем мире занимают одну из ведущих позиций в структуре болезней желудочно-кишечного тракта. Поэтому открывает этот выпуск статья О. В. Князева и соавт., подробно освещающая заболеваемость и распространенность ВЗК в нашей стране и мире, определяющая основные тенденции этих показателей и обосновывающая необходимость создания единого регистра больных язвенным колитом и болезнью Крона, что будет способствовать совершенствованию мер оказания медицинской помощи больным с ВЗК.
    Раздел «Клиническая гастроэнтерология» представлен рядом оригинальных работ по различным аспектам заболеваний тонкой и толстой кишки. В статье М. Д. Ардатской (Москва) изложены собственные результаты по изучению содержания короткоцепочечных жирных кислот у больных раком толстой кишки в слизистой оболочке толстой кишки и кале, и предложено использовать определение короткоцепочечных жирных кислот в кале в качестве неинвазивного теста, позволяющего косвенно судить о наличии опухолевого процесса, его локализации и степени прорастания стенки кишки опухолью.
    Быкова А. П. и Козлова И. В. из Саратовского государственного медицинского университета имени В. И. Разумовского установили, что хронический панкреатит ассоциирован с функционально-структурными изменениями толстой кишки. Определено, что баланс цитокинов и морфологическая картина в слизистой оболочке толстой кишки наиболее значимо изменены при токсико-метаболическом варианте хронического панкреатита.
    Еще одна работа поступила от сотрудников кафедры госпитальной терапии лечебного факультета Саратовского государственного медицинского университета имени В. И. Разумовского. Наличие у больных алекситимии (сниженной способности или затрудненности в вербализации эмоциональных состояний со следующими особенностями в когнитивно-афферентной сфере) может затруднить оценку тяжести атаки у больных язвенным колитом. Изучению соотношения между показателями выраженности болевого синдрома и алекситимии посвящена представленная работа.
    Профессор И. Л. Халиф и соавт. (ГНЦК им А. Н. Рыжих, Москва) продемонстрировали, что баллонная манометрия позволяет выявлять определенную корреляцию между клинической картиной СРК (преобладание запоров, поносов или неустойчивый стул) и типом моторики прямой кишки, а, следовательно, быть рекомендована как метод функционального исследования у пациентов с СРК.
    Е. А. Маевская и соавт (МГМСУ им. А. И. Евдокимова) доказали, что в сравнении с лактулозой длительная терапия функционального запора псиллиумом характеризуется более выраженным слабительным действием, лучше переносится, снижает выраженность СИБР и улучшает качество жизни больных.
    Следующие две работы посвящены изучению динамики показателей системного воспаления и локального иммунного статуса у больных с СРК при лечении различными фармпрепаратами.
    В разделе «Хирургическая гастроэнтерология» собственным опытом хирургического лечения идиопатического мегаколон и мегаректум у больных различных возрастов делятся наши коллеги из ФГБУ «Государственный научный центр колопроктологии им. А. Н. Рыжих» Минздрава России
    Колоректальный рак является одной из распространенных форм злокачественных новообразований в мире. Сегодня колоректальное стентирование является малоинвазивной альтернативой экстренному оперативному вмешательству. Ведущие сотрудники отделения эндоскопической хирургии ФГБУ «ГНЦ ЛМ ФМБА» суммируют показания и противопоказания, положительные результаты и осложнения стентирования при толстокишечной непроходимости Раздел «Обзоры» открывает статья С. М. Захаренко (Санкт-Петербург), в которой представлены данные последних лет о распространенности и видах Clostridium difficile-инфекции, о циркуляции микроорганизма в окружающей среде и человеческой популяции, об особенностях взаимодействия с нормальной микрофлорой желудочно-кишечного тракта человека.
    Продолжает раздел обзор профессора О. Н. Минушкина и соавт., посвященный проблеме синдрома избыточного бактериального роста. Даны определения синдрома, эпидемиология, клинические проявления, критерии диагностики. Приведены новые данные по изучению синдрома избыточного бактериального роста у больных воспалительными заболеваниями кишечника.
    В следующем материале профессор И. Д. Лоранская., Е. В. Степанова и В. А. Купаева (Москва) рассматривают современные представления о механизмах развития ожирения при нарушениях кишечного микробиоценоза, перспективы использования биокоррекции микрофлоры в лечении метаболических заболеваний.
    В мае 2016 мировое гастроэнтерологическое сообщество на Американской гастроэнтерологической неделе познакомилось с Римскими критериями IV. Основные положения этого консенсуса представлены в настоящей публикации, подготовленной группой авторов под руководством академика РАН И. В. Маева. В отличие от предыдущей классификации в разделе функциональных заболеваний кишечника появилась новая форма — ​опиоид-индуцированный запор. Скорректирована и дефиниция функциональной диареи.
    Нецелиакийная чувствительность к глютену (НЦЧГ) — ​это новый синдром, проявляющийся непереносимостью глютена, это не аллергическое и не аутоиммунное состояние, при котором употребление глютена может приводить к возникновению симптомов, сходных с проявлениями целиакии. Вопросы патогенеза, дифференциальной диагностики и принципов лечения этой нозологии рассматривает в своей статье профессор Е. А. Корниенко (Санкт-Петербург).
    Завершает наш номер материал авторов из ГНИЦ колопроктологии, посвященный терапии по принципу «биологической обратной связи» (БОС-терапия) — ​одному из ведущих методов лечения запора, вызванного функциональным расстройством дефекации (диссинергической дефекации). В статье представлены актуальные данные об эффективности и методах проведения БОС-терапии. Отражен действующий стандартный протокол лечения, предложенный Обществами нейрогастроэнтерологии и моторики США и Европы, представлены перспективы дальнейших научных исследований БОС-терапии в лечении хронического запора. Надеемся, что этот выпуск журнала будет интересным и полезным для очень широкого круга врачей!

    Ответственный за выпуск редактор,

      профессор кафедры гастроэнтерологии ФГБУ ДПО
    «Центральная государственная медицинская академия» УПД Президента РФ

    доктор медицинских наук, профессор 
      Мария Дмитриевна Ардатская 
       
    1. Moscow clinical scientific practical center of the Department of health of Moscow (Moscow, Russian Federation)
    2. Research Institute for health organization and medical management of the Department of health of Moscow (Moscow, Russian Federation)
    3. State budgetary educational institution of higher professional education “North-Western state medical University n. a. I. I. Mechnikov” (St. Petersburg, Russian Federation)

    Keywords:Inflammatory bowel disease,epidemiology,incidence

    Abstract:Prevalence and incidence of inflammatory bowel disease (IBD) is significantly inferior to other diseases of the digestive system, but in severity, frequency of complications and mortality in the world they occupy one of the leading positions in the structure of diseases of the gastrointestinal tract. The results of the large controlled studies show a steady increase in the incidence of this disease in the world. According to recent reports the prevalence of ulcerative colitis (UC) in North America is 249 per 100 thousand population, Crohn’s disease (CD) - 319 to 100 thousand, the prevalence of UC in Europe is 505 per 100 thousand population, CD - 322 to 100 thousand; the incidence of UC in North America is 19.2 per 100 thousand population, CD - 20.2 per 100 thousand, UC incidence in Europe is 24.3 per 100 thousand population, CD - 12.7 per 100 thousand. However, there are no clear data on the prevalence and incidence of this pathology in our country. This circumstance requires the creation of a unified register of patients with UC and CD, which will contribute to the improvement of the measures of care for patients with IBD.

      1. Cecil W. Ward and Derek T. Thomas. Crohn’s Disease with Acute Obstruction, Br Med J. 1941 Nov 1; 2(4217): 613-614.
      2. Crohn BB. Inflammatory diseases of the small intestine. J Omaha Midwest Clin Soc. 1946 Aug;7(3):77-83.
      3. Molodecky NA, Soon IS, Rabi DM, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46-54. e42; quiz e30.
      4. Kappelman MD, Rifas-Shiman SL, Kleinman K, Ollendorf D, Bousvaros A, Grand RJ, Finkelstein JA. The prevalence and geographic distribution of Crohn’s disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol. 2007; 5:1424-9.
      5. Loftus EV., Jr The burden of inflammatory bowel disease in the United States: a moving target? Clin Gastroenterol Hepatol. 2007;5:1383-4.
      6. Muller-Lissner S.A., Bollani S., Brummer R. J. et al. Epidemiological aspects of irritable bowel syndrome in Europe and North America // Digestion. -2001. - Vol. 64, № 3. - P. 200-204.
      7. Schultes H., Guminski W., Rosche U. et al. Irritable bowel syndrome costs sickness funds DM 2.8 billion per year // Value in Health. - 2001. - Vol. 4, Sep-Oct, Issue 2. - P. 123.
      8. Воробьев П. А., Воробьев П. А., Авксентьева М. В., Юрьев А. С., Сура М. В. Клинико-экономический анализ (оценка, выбор медицинских технологий и управление качеством медицинской помощи) (Под ред. Воробьева П. А.) - М.: «Ньюдиамед», 2004. - 404 с.
      9. Lesage S., Zouali H., Colombel J.-F. et al. Genetic analyses of chromosome 12 loci in Crohn’s disease. // Gut. - 2000. - 47. - p. 787-791.
      10. Satsangi J. Genetics of inflammatory bowel disease. // Falk Symp. № 85. -1995. - p.50.
      11. Е. Ю. Валуйских, И.О, Светлова, С. А. Курилович, М. Ф. Осипенко, В. Н. Максимов, М. И. Воевода. Клинико-генетические аспекты воспалительных заболеваний кишечника. Российский журнал гастроэнтерологии, гепатологии и колопроктологии. - 2008. - Т. 18. - № 6. - С. 68-74.
      12. Никулина И. В. Златкина А. Р., Белоусова Е. А., Румянцев В. Г., Юрков М. Ю. Оценка клинико-эпидемиологических показателей воспалительных заболеваний кишечника в Московской области // Российский журнал гастроэнтерологии, гепатологии, колопроктологии.-1997.-, № 2. - с. 67-71.
      13. Николаева Н. Н., Чечеткина И. Д., Николаева Л. В., Гигилева Н. Л. Эпидемиология язвенного колита и болезни Крона в Красноярском крае // Российский журнал гастроэнтерологии, гепатологии, колопроктологии. - 2004.-14. - № 5. - с. 133; 496.
      14. Irvine EJ, Farrokhyar F., Swarbrick E. T. A critical review of epidemiological studies in inflammatory bowel disease. Scand, Scand.J.Gastroenterol.2001.36[1]:2-15.
      15. Cachia E, Calleja N, Aakeroy R, Degaetano J, Vassallo M. Incidence of inflammatory bowel disease in Malta between 1993 and 2005: a retrospective study. Inflamm Bowel Dis. 2008 Apr;14(4):550-3.
      16. Mendelof A. I. The epidemiolody of chronic inflammatory bowel disease // Inflammatory Bowel Disease. Ed. by G. Jarnerot.-1992. - p.15-34.
      17. Langholz E. Epidemiology of IBD and colorectal cancer in IBD. // International meeting “Prevention of colorectal cancer with 5-ASA. - Copenhagen, May 2000.
      18. Söderlund S, Granath F, Broström O, Karlén P, Löfberg R, Ekbom A, Askling J. Inflammatory bowel disease confers a lower risk of colorectal cancer to females than to males. Gastroenterology. 2010 May;138(5):1697-703.
      19. Binder V. Epidemiology // Inflammatory Bowel Disease-from bench to bedside. Ed. Andus T. et al. Proceeding of Falk-symposiun № 96.-1996.-Kluwer Academic Publishers.
      20. Munkholm P., Langholz E., Nielsen J, H, et al. Incidence and prevalence of Crohn’s disease in the county of Copenhagen, 1962-87: a sixfold increase in incedence. Scand.J.Gastroenterol.1992, B 7):609-614.
      21. Binder V., Both H., Hansen P. K. et al. Incidence and prevalence of ulcerative colitis and Crohn’s disease in the County of Copenhagen, 1962-1978. Gastroenterology, 1982,83C):563-568.
      22. Shivanada S., Lennard-Jones J., Logan R. Incidence of inflammatory bowel disease across Europe: is there a difference between north and south? Results of the European Collaborative Study of the Inflammatory Bowel Disease EC-IBD).-Gut.-1996.-39 E).-690-697.
      23. Fonager K., Sorensen H. T., Olsen J. Change in incidence of Crohn’s disease and ulcerative colitis in Denmark. A study based on National Registry of Patients, 1981-1992. Int J. Epidemiol.-1997.;26;1003-1008.
      24. Trallori G., Palli D., Saieva С et al. A population-based study of inflammatory disease in Florence over 15 years A978-92). Scand.J, Gastroenterol.1996;31(9):892-899.
      25. Ekbom A., Helmick C, Zack M., Adami H-O. The epidemiolody of inflammatory bowel disease: a large population-based study in Sweden // Gastroenterology. - 1991. - v. 100. - p. 350-358.
      26. Zvidi I, Hazazi R, Birkenfeld S, Niv Y. The prevalence of Crohn’s disease in Israel: a 20-year survey. Dig Dis Sci. 2009 Apr;54(4):848-52.
      27. Haug A., Schrumpf E., Barsted S., Fluge. Et al. Epidemiolody of ulcerative colitis in Western Norway// Scand. J. Gastroenterol.-1988. - 23. - p.517-522.
      28. Kildebo S., Nordgard K., Aronsen O., Brecken P. et al. The incidence of ulcerative colitis in Northern Norway from 1983 to 1986. // Scand. J. Gastroenterol.-1990. - v.25. - p.890-896.
      29. Myren J., Gjone E., Hertzberg J. Epidemiolody of ulcerative colitis and regional enterocolitis (Crohn’s disease) in Norway//Scand. J. Gastroenterol.-1971. - v.6. - p.511-514.
      30. Vatn M., Mourn В., the IBSEN study group. IBD in Norway // Inflammatory Bowel Disease. Ed. Monteiro E., Taravela F. Proceedings of Falk-symposiun № 76.-1994.-Kluwer Academic Publishers.
      31. Mourn B., Vatn M. H. Ekbom A. et al. Incidence of inflammatory bowel disease in southeastern Norvay: evaluation of methods after 1 year of registration. Southeastern Norvay IBD Study Group of Gastroenterologists. Digestion 1995,56E):377-381.
      32. Bianchi P., Bodini P., Zambelli A., Curzio M., Ranzi T. Epidemiolody of IBD in Italy//Inflammatory Bowel Disease. Ed. Monteiro E., Taravela F. Proceedings of Falk-symposiun № 76.-1994.-Kluwer Academic Publishers.
      33. Sonnenberg A., McCarty D.J., Jacobsen S. J. Geographic variation of inflammatory bowel disease within the United States. Gastroenterology 1991, 100[1]: 143-149.
      34. Mayberry J. F., Mann R. Inflammatory bowel disease in rural sub-Saharan Africa: rarity of diagnosis in patients attending mission hospitals. Digestion 1989; 44C):172-176.
      35. Yang S. K., Loftus E. V. Jr, Sandbom W. J. Epidemiology of inflammatory bowel disease in Asia. Inflamm.Bowel Dis. 2001, 7C);260-270.
      36. Yoshida Y., Murata Y. Inflammatory Bowel Disease in Japan: studies of epidemiology and etiopathogenesis // Med Clin North Am.-1990.-V. 74. - p. 67-89.
      37. Burisch J., Pedersen N., Сukoviс-Сavka S., et al. East-West gradient in the incidence of inflammatory bowel disease in Europe: the ECCO-EpiCom inception cohort. Gut.2014;63(4):588-97.
      38. Braegger CP, Ballabeni P, Rogler D, Vavricka SR, Friedt M, Pittet V. Epidemiology of inflammatory bowel disease: is there a shift towards onset at a younger age? Swiss IBD Cohort Study Group.J Pediatr Gastroenterol Nutr. 2011 Aug;53(2):141-4.
      39. Bernstein C.N, Blanchard J. F., Rawsthorne P., Wajda A. Epidemiology of Crohn’s disease and ulcerative colitis in the central Canadian province: a population-based studi. Am J Epidemiol 1999,149A0):916-924.
      40. Farrokhyar F., Swarbrick E., Grace R. et al. Low mortality in ulcerative colitis and Crohn’s disease in three regional centers in England. Am J. Gastroenterol.;96B):501-507.
      41. Colombel J., Salomaz J., Cortot A. Incidence of inflammatory bowel disease in Northwestern France. Preliminary results in redion Pas-de-Calais // Scand. J. Gastroenterol.-1989. - 24.-(Suppl.170).-p.22-24.
      42. Tysk CJarnerot G. Ulcerative proktocolitis in Orebro, Sweden- a retrospektive epidemiologic study, 1963-87. Scand.J.Gastroenterol. 1992;27:945-950.
      43. Mendelof A. I. The epidemiolody of chronic inflammatory bowel disease // Inflammatory Bowel Disease. Ed. by G. Jarnerot.-1992. - p.15-34.
      44. Veluswamy H, Suryawala K, Sheth A, Wells S, Salvatierra E, Cromer W, Chaitanya GV, Painter A, Patel M, Manas K, Zwank E, Boktor M, Baig K, Datti B, Mathis MJ, Minagar A, Jordan PA, Alexander JS. African-American inflammatory bowel disease in a Southern U. S. health center. BMC Gastroenterol. 2010 Sep 9;10:104.
      45. Bernstein C. N., Kraut A., Blanchard J. F. et al. The relationship between inflammatory bowel disease and socioeconomic variables. Am J. Gastroenterol.2001;96G):2117-2125.
      46. Loftus E., Silverstein M., Sandbom W.et al. Ulcerative colitis in Olmsted County, Minnesota, 1940-1993; incidence, prevalence, and survival. Gut 2000,46C):336-343.
      47. Berner J., Kiaer Т. Ulcerative colitis and Crohn’s disease on the Faroe Islands 1964-83. A retrospective epidimiological survey. -Scand. J. Gastroenterol. - 1986. - 21.-188-192.
      48. Siddique I, Alazmi W, Al-Ali J, Al-Fadli A, Alateeqi N, Memon A, Hasan F. Clinical epidemiology of Crohn’s disease in Arabs based on the Montreal classification. Inflamm Bowel Dis. 2011 Oct 10. doi: 10.1002/ibd.21890.
      49. Malaty HM, Hou JK, Thirumurthi S. Epidemiology of inflammatory bowel disease among an indigent multi-ethnic population in the United States. Clin Exp Gastroenterol. 2010;3:165-70.
      50. Fireman Z., Grossman A., Lilos P. Epidemiology Crohn’s disease in the Jewish population of Central Israil.//Am J Gastroenterol.-1989.-v.84.-p.255-258.
      51. Odes H., Fraser D., Krawuec J. Incidence of idiopathic ulcerative colitis in Jewish population subdroup in the Beer Sheva region of Israil.// Am J Gastroenterol.-1987. - v.82.-p.854-858.
      52. Левитан М. Х., Смесова Р. Неспецифический язвенный колит и беременность // Врачебное дело, 1981. - № 3. - С. 29-32.
      53. Allan, RN et al. Inflammatory Bowel Disease. London: Churchill Livingstone, 1997.
      54. Höie O, Wolters F, Riis L, Aamodt G, Solberg C, Bernklev T, Odes S, Mouzas IA, Beltrami M, Langholz E, Stockbrügger R, Vatn M, Moum B; European Collaborative Study Group of Inflammatory Bowel Disease (EC-IBD). Ulcerative colitis: patient characteristics may predict 10-yr disease recurrence in a European-wide population-based cohort. Am J Gastroenterol. 2007 Aug;102(8):1692-701.
      55. Sonnenberg A, Walker JT. Occupational mortality associated with inflammatory bowel disease in the united states 1984-1998. Inflamm Bowel Dis. 2011 Jun 24. doi: 10.1002/ibd.21807.
      56. Kennedy NA, Clark DN, Bauer J, Crowe AM, Knight AD, Nicholls RJ, Satsangi J. Nationwide linkage analysis in Scotland to assess mortality following hospital admission for Crohn’s disease: 1998-2000.Aliment Pharmacol Ther. 2011 Nov 9. doi: 10.1111/j.1365-2036.2011.04906.x.
      57. Brostrom O., Monsen U., Nordenwall B. et al. Prognosis and mortality of ulcerative colitis in Stockholm County, 1955-1979. Scand.J.Gastroenterol. 1987;22(8):907-91.
      58. Ekbom A., Helmick C. G., Zack M. et al. Survival and causes of death in patients with inflammatory bowel disease; a population-based study. Gastroenterology, 1992;103C):954-960.
      59. Probert C. S., Jayanthi V., Wicks A. C., Mayberry J. F. Mortality in patients with ulcerative colitis in Leicestershire, 1972-1989. An epidemiological study. Dig.Dis, Sci, 1993;38{3}:538-541.
      60. Romberg-Camps M, Kuiper E, Schouten L, Kester A, Hesselink-van de Kruijs M, Limonard C, Bos R, Goedhard J, Hameeteman W, Wolters F, Russel M, Stockbrügger R, Dagnelie P. Mortality in inflammatory bowel disease in the Netherlands 1991-2002: results of a population-based study: the IBD South-Limburg cohort. Inflamm Bowel Dis. 2010 Aug;16(8):1397-410.
     


    Full text is published :
    Knyazev O.V., Shkurko T.V., Fadeeva N.A., Bakulin I.G. et. al EPIDEMIOLOGY OF CHRONIC INFLAMMATORY BOWEL DISEASE. YESTERDAY, TODAY, TOMORROW. Experimental and Clinical Gastroenterology Journal. 2017;139(03):04-12
    Read & Download full text

    1. Central state medical Academy (Moscow, Russian Federation)

    Keywords:microflora,of low molecular weight metabolites of the intestinal microbiota,short chain fatty acids (SCFA),colon cancer,butyric acid (butyrate),psyllium,butyrate+inulin

    Abstract:The purpose of the study is to examine the content of short chain fatty acids (SCFA) in patients with colon cancer (CC) in the mucosa of the colon (CM) and feces by GLC-analysis: to assess their significance. Material and methods: the Study was performed in 52 patients operated on for colon cancer. Patients ranged in age from 61 to 81 years, 32 women and 20 men. In 14 cases the tumor was localized in the blind and ascending Department of the colon, 6 - in descending, 26- in the sigmoid colon, in 6 - in rectosigmoid part. In this regard, in 14 cases, the right hemicolectomy, 14 left hemicolectomy, 16 resection of the Sigma, 8 - anterior resection of the rectum. Histological examination revealed colon adenocarcinoma varying degrees of differentiation (high difference-25, moderate - -20, low of -7). Distribution of patients according to prevalence of tumor process was as follows: -2 I, IIA -4, -36 IIIA, IIIB -10. By GLC analysis to determine the content SCFA in CM and feces. Results: the Absolute contents SCFA in CM, the group of patients with CC tends to decrease compared to the control group. The total absolute concentration SCFA when the tumor in the right departments is higher in comparison with the concentration of SCFA when left-sided localization (0,420±0,110 mg/g and of 0.350±0.001 mg/g, respectively). In the study profiles in samples of tumor and feces when left localization of the significantly elevated level of propionic and butyric acids (up to 0,159±0,001 ed and 0,105±0,002 units, respectively, at a rate of 0,134±0,001 ed and 0,087±0,004 units, respectively, in CM; to 0,241±0,001 0,221 and u of±0.002 units, respectively, at a rate 0,189±0,001 ed and 0,176±0,004 units, respectively, in the feces), when right - level of isolated oil acid (0,093±0,011 u, at a rate of up to 0,067±0,01 IU per CM; to 0.281±0,002 units, at a rate of up to 0,176±0,004 ed feces). Among CC patients with increasing tumor stage marks an increase in the level of butyric acid (0,099±0,021 ed at T 1-3, to 0,110±0,030 ed at T4 in SOTK, 0,201±0,002 ed at T 1-3, to 0,220±0,002 ed at T4 in the feces) and increasing the ratio of isocyclic to acids in CM and feces (1,929±0,010 u at T 1-3, to 2,217±0,019 ed at T4 in CM, with 2,432±0,120 ed at T 1-3, to 4,327±0,110 units at T4 in feces). By reducing the degree of tumor differentiation (highly, moderately -, poorly-differentiated) in CM marked increase in the proportion of propionic (of 0.152±0,010 u, 0,209±0,01 units, 0,224±0,010 u, respectively) and mainly oil acid (of 0.085±0,010 units, 0,120±0,010 units, 0,168±0,010 units, respectively). The ratio of isocyclic forms to acids do not differ. Conclusion: in CC patients revealed specific changes SCFA in CM, depending on the localization, differentiation grade and depth of germination by the tumor of the intestinal wall is comparable to the change in feces. Changes SCFA in the stool can be used as a non-invasive test that allows indirectly to judge the presence of tumor, its localization and extent of germination of the bowel wall by the tumor. The use of preparations containing precursors of butyric acid (psyllium) or itself butyric acid, is a promising direction of CC canceroprevention.

      1. Шапошников А. В. Колоректальный рак. Канцерогенез и онкопревенция: пособие для врачей. - Москва: Форте принт, 2015.
      2. Marta Borges-Canha, et al. Role of colonic microbiota in colorectal carcinogenesis: A systematic review. Rev Esp Enferm Dig (Madrid) Vol. 107, № 11, pp. 659-671, 2015.
      3. Scott J. Bultman. Emerging roles of the microbiome in cancer. Carcinogenesis vol.35 №.2 pp.249-255, 2014.
      4. Short Chain Fatty Acides.// Congress Short Report Falk Symposium, comp. by Scheppach W. - Strasbourg, 1993. - 50 p.
      5. Siavoshian S. Et al. Butyrate and trichostatin A effects on the proliferation/differentiation of humen intestinal epithelial cells: induction of cyclin D 3 and p21 expression.// Gut. - 2000. - V.46. - Pp. 507-514.
      6. Ардатская М. Д. Масляная кислота и инулин в клинической практике. Москва, Прима Принт, 2016.
      7. Lupton, J. R. Microbial degradation products influence colon cancer risk: the butyrate controversy. 2004, J. Nutr., 134, 479-482.
      8. Donohoe DR, et al. (2014) A Gnotobiotic Mouse Model Demonstrates That Dietary Fiber Protects against Colorectal Tumorigenesis in a Microbiota- and Butyrate-Dependent Manner. Cancer discovery 4(12):1387-1397.
      9. Sebastian C & Mostoslavsky R (2014) Untangling the fiber yarn: butyrate feeds warburg to suppress colorectal cancer. Cancer discovery 4(12):1368-1370.
      10. Scott J. Bultman. The Microbiome and Its Potential as a Cancer Preventive Intervention. Semin Oncol. 2016 Feb;43(1):97-106.
      11. Головенко О. В., Халиф И. Л., Головенко А. О. Лечение заболеваний кишечника масляной кислотой. Поликлиника № 2, 2012.
      12. Thangaraju M, et al. (2009) GPR 109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer research 69(7):2826-2832.
      13. Singh N, Gurav A, Sivaprakasam S, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014;40:128-39. DOI: 10.1016/j.immuni.2013.12.007
      14. Terzic J, Grivennikov S, Karin E, & Karin M (2010) Inflammation and colon cancer. Gastroenterology 138(6):2101-2114 e2105.
      15. Itzkowitz SH & Harpaz N (2004) Diagnosis and management of dysplasia in patients with inflammatory bowel diseases. Gastroenterology 126(6):1634-1648.
      16. Ардатская М. Д. Клиническое значение короткоцепочечных жирных кислот при патологии желудочно-кишечного тракта. Докторская диссертация, М., 299 с., 2003.
      17. Иконников Н. С., Ардатская М. Д., Дубинин А. В., и др. Патент РФ «Способ разделения смеси жирных кислот фракции С2-С7 методом газожидкостной хроматографию». N 2145511 от 9.04.1999 г.
      18. Осина В. А. Абсорбционная функция толстой кишки в норме и при патологии.// Экспериментальная и клиническая гастроэнтерология. - 2002. - № 1. - С. 81-85.
      19. Emenaker N. J., Basson M. D. Short chain fatty acids differentially modulate cellular phenotype and c-myc protein levels in primary human nonmalignant and malignant colonocytes. Dig Dis Sci. 2001 Jan;46(1):96-105.
      20. Emenaker N. J., Calaf G. M., Cox D, Basson M. D., Qureshi N. Short-chain fatty acids inhibit invasive human colon cancer by modulating uPA, TIMP-1, TIMP-2, mutant p53, Bcl-2, Bax, p21 and PCNA protein expression in an in vitro cell culture model. J Nutr. 2001 Nov;131 (11 Suppl): 3041S-6S.
      21. Федоров Н. А. Циклические пуриновые нуклеотиды 3,5-АМФ и 3,5-ГМФ как факторы регуляции пролиферации и дифференцировки гемопоэтических клеток.//Успехи современной биологии, 1975, Т. 79, вып.2, с. 225-240.
      22. De Rubertis F. R., Chayoth R, Field J. B. The content and metabolism of cyclic AMP and cyclic GMP in the adenocarcinoma of the human colon.//J.Clin.Invest.,1976, v.57, p.641-649.
      23. Блохина Н. Г., Пискунова Т. В., Киркин Б. В., Чижиков Б. А. Исследование циклических нуклеотидов при раке, полипах толстой кишки и неспецифических колитах.//Вест. АМН СССР, 1985, № 3, с. 74-77.
      24. Бышевский А. Ш., Терсенов О. А. Биохимия для врача. Екатеринбург, «Уральский рабочий», 1994.
     


    Full text is published :
    Ardatskaya M.D. THE ROLE OF LOW MOLECULAR WEIGHT METABOLITES OF THE INTESTINAL MICROBIOTA IN THE PATHOGENESIS, DIAGNOSIS AND PREVENTION OF COLORECTAL CANCER. Experimental and Clinical Gastroenterology Journal. 2017;139(03):13-21
    Read & Download full text

    1. Saratov State Medical University n. a. V. I. Razumovsky (Saratov, Russian Federation)

    Keywords:chronic pancreatitis,colon,colonoscopy,cytokines

    Abstract:Purpose of the study. To investigate the clinical and morphological characteristics of the intestine in patients with chronic pancreatitis with the assessment of the quantitative determination of proinflammatory cytokines in the mucosa of the colon. Material and methods. In a study of 113 patients with chronic pancreatitis included. The analysis of intestinal dysfunction symptoms, endoscopic, morphological changes of the colon, the quantitative content of proinflammatory cytokines in colonic biopsies: IL-2, IL-6, IL-8, a more pronounced when toxic-metabolic chronic pancreatitis. Results. It has been established that chronic pancreatitis functionally associated with structural changes of the colon. In colonic biopsies increased content of IL-2, IL-6, IL-8. Morphological picture of colon mucosa in patients with normal excretory function of the pancreas and moderate exocrine insufficiency of pancreas (EIP) as a whole was normal. Revealed lymphoplasmacytic infiltration of the mucosa - 47 (68 %) patients, a decrease in the number of glands. Proliferation of the capillaries of the mucosa was observed in 21 (30,4 %) patients, in 16 (23.2 per cent) of the observed decrease in the number and size of crypts. A number of patients - 19 (40,4 %) with moderate EIP showed a growth of connective tissue fibers in the lamina propria that were regarded as early signs of fibrosis of the colon wall. In patients with severe EIP infiltration of the mucosa and submucosa by lymphocytes and plasma cells were observed more frequently in 85 %. For this group of patients was characterized by the abundance of connective-tissue elements of the mucosa and marked reduction of crypts in the tissue of the colon.

      1. Ивашкин В. Т., Маев И. В., Охлобыстин А. В. c соавт. Рекомендации российской гастроэнтерологической ассоциации по диагностике и лечению хронического панкреатита. Российский журнал гастроэнтерологии, гепатологии, колопроктологии 2014; 4: 70-97.
      2. Hoffmeister A, Mayerle J, Beglinger C und and. S 3-Leitlinie: Chronische Pankreatitis: Definition, Ätiologie, Diagnostik und konservative, interventionell endoskopische und operative Therapie der chronischen Pankreatitis. Leitlinie der Deutschen Gesellschaft für Verdauungs- und Stoffwechselkrankheiten (DGVS). AWMF-online 2012: 115 P.
      3. Козлова И. В., Пахомова А. Л. Современный пациент гастроэнтерологического профиля: штрихи к клиническому портрету. Экспериментальная и клиническая гастроэнтерология. 2015; 6: 4-10.
      4. Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 2013; 144(6): 1252-1261.
      5. Raghuwansh PS, Rajinder KD, Ashok KS. New Insights into the Pathogenesis of Pancreatitis. Current Opinion in Gastroenterology 2013 Sep; 29(5): 523-530.
      6. Полуэктова Е. А. Диагностика и лечение внешнесекреторной недостаточности поджелудочной железы у больных с хроническим панкреатитом. Consilium Medicum 2009; 8: 77-79.
      7. Mattar R, Lima GA, Costa MZ et al. Comparison of fecal elastase 1 for exocrine pancreatic insufficiency evaluation between ex-alcoholics andchronic pancreatitis patients. Arq Gastroenterol. 2014;51(4):297-301
      8. Быкова А. П., Козлова И. В. Механизмы развития и особенности патологии органов желудочно-кишечного тракта при хроническом панкреатите. Экспериментальная и клиническая гастроэнтерология 2016;11:69-75.
      9. Васильев А. В., Бакарев М. А., Лапий Г. А. с соавт. Структурная реорганизация слизистой оболочки двенадцатиперстной кишки при хроническом панкреатите. Фундаментальные исследования 2012; 7: 278-282.
      10. Токарева Е. В., Хлынова О. В., Горовиц Э. С. Микрофлора кишечника у больных хроническим панкреатитом. Вестник Здоровье и образование в XXI веке 2011; 2: С. 84-85.
      11. Сираева А. С., Фаизова Л. П. Показатели колонизационной резистентности толстой кишки у больных хроническим панкреатитом. Экспериментальная и клиническая гастроэнтерология 2014; 5: 21-26.
      12. Мингазова С. К., Хасанова А. Р., Нигматуллина А. Э. c соавт. Показатели цитокинового воспаления при хроническом панкреатите. Медицинский вестник Башкортостана 2012; 6: 49-52.
      13. Аскаров Т. А., Сулейманов С. Ф., Махмудов Б. Ф., Узаков Ф. М. Коррекция иммунных расстройств у больных хроническим панкреатитом. Вісник проблем біології і медицини 2013; 3: 20-22.
      14. Etemad B, Whitcomb DC. Chronic pancreatitis: diagnosis, classification, and new genetic developments. Gastroenterology 2001;120(3):682-707.
      15. Vonlaufen A, Spahr L, Apte MV, Frossard JL. Alcoholic pancreatitis: A tale of spirits and bacteria. World Journal of Gastrointestinal Pathophysiology 2014; 5(2): 82-90.
      16. Кулик И. А., Бойко В. В., Шевченко А. Н. c соавт. Иммунологический статус пациентов при хроническом рецидивирующем панкреатите. Инновации в науке 2013; 23: 141-149.
      17. Lindley KJ. Chronic Pancreatitis. Indian Journal of Pediatrics 2006; 73(10): 907-912.
      18. Basuroy S, Sheth P, Mansbach CM, Rao RK. Acetaldehyde disrupts tight junctions and adherens junctions in human colonic mucosa: protection by EGF and L-glutamine. American Journal of Physiology - Gastrointestinal and Liver Physiology 2005; 289: 367-375.
     


    Full text is published :
    Bykova A.P., Kozlova I.V. CLINICAL-ENDOSCOPIC AND MORPHOLOGICAL FEATURES OF THE COLON IN CHRONIC PANCREATITIS. Experimental and Clinical Gastroenterology Journal. 2017;139(03):22-27
    Read & Download full text

    1. Federal State Budget Educational Institution of High Education «Saratov State Medical University named after V. I. Razumovsky» of the Ministry of Public Health of Russia (Saratov, Russian Federation)

    Keywords:ulcerative colitis,pain syndrome,alexithymia

    Abstract:Purpose of the study: to investigate the relationship between the index of alexithymia and severity of pain in patients with ulcerative colitis and comparable severity of the next attack. Materials and methods: the study included 59 patients with acute exacerbation of ulcerative colitis who were hospitalized in the gastroenterological department of the Saratov Regional Hospital. On admission to the hospital for all patients was determined the level of alexithymia using the Toronto alexithymia scale (1985), adapted in the Institute named after VM Bekhterev (2005). The severity of the next attack of ulcerative colitis was evaluated according to the criteria of Truelove-Witts (1955). Results: with comparable severity of the next attack of ulcerative colitis patients with high alexithymia significantly more often reported higher levels of pain than at low values one. Conclusion: in the choice of optimal tactics analgesic drug therapy in patients with ulcerative colitis in the period of exacerbation it is necessary to consider psychological characteristics of their personality.

      1. Под ред. Ивашкина В. Т. Рекомендации Российской гастроэнтерологической ассоциации и Ассоциации колопроктологов России по диагностике и лечению взрослых больных язвенным колитом. Росс. Журн. Гастроэнтерол, гепатол., колопроктол. - 2015. - № 1. - С. 48-65.
      2. Dignass A., Eliakim R., Magro F., Maaser C., et al. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 1: definitions and diagnosis. J Crohns Colitis. 2012; 6(10):965-90.
      3. Carter MJ, Lobo AJ, Travis SP et al. IBD Section, British Society of Gastroenterology. Guidelines for the management of inflammatory bowel disease in adults. Gut 2004;53 (Suppl. 5): V1-16.
      4. Маркова A. A., Кашкина Е. И. Особенности течения неспецифического язвенного колита в зависимости от длительности заболевания. Росс. журнал гастроэнтерологии, гепатологоо, колопроктологии. (приложение 38). - 2011. - Т. XXI. - № 5. - С. 53.
      5. Койнова И. А., Белобородова Е. В., Бурковская В. А. и др. Оcобенности течения воспалительных заболеваний кишечника в зависимости от психологического статуса пациента. Современные проблемы науки и образования. - 2016. - № 6.; URL: https://www.science-education.ru/ru/article/view?id=25615 (дата обращения: 09.01.2017).
      6. Брель Е. Ю. Проблема изучения алекситимии в психологических исследованиях. Вестник КемГУ. - 2012. - № 3(51). - С. 173-176.
      7. Ересько Д. Б. Алекситимия и методы ее определения при пограничных психосоматических расстройствах. Пособие для психологов и врачей. Санкт-Петербург 2005. -25с.
      8. S. Nahon, P. Lahmek, C. Saas et al. “Socioeconomic and psychological factors associated with nonadherence to treatment in inflammatory bowel disease patients: results of the ISSEO survey,” Inflammatory Bowel Diseases, vol. 17, no. 6, pp. 1270-1276, 2011.
      9. Truelove SC, Witts LJ. Cortisone in ulcerative colitis: final report on a therapeutic trial. Br Med J 1955; ii:1041-8.
      10. Харченко Ю. А. Адекватная оценка боли - залог ее успешного лечения //. Universum: Медицина и фармакология: электрон. научн. журн. 2014. № 4 (5).
     


    Full text is published :
    Fedosova M.A., Kashkina E.I., Kunitsuna M.A. CORRELATION BETWEEN SEVERETY OF PAIN SYNDROME AND ALEXITHYMIA IN PATIENTS WITH ULCERATIVE COLITIS. Experimental and Clinical Gastroenterology Journal. 2017;139(03):28-31
    Read & Download full text

    1. State scientific center of coloproctology (Moscow, Russian Federation)

    Keywords:rectal motility,gastrocolic reflex,irritable bowel syndrome (IBS),constipation,diarrhea,rectal manometry

    Abstract:Background and aims: the article presents the results of the study rectal motility in patients with IBS. Our aim was to investigate the possibility of using balloon manometry to assess the locomotor activity of the rectum in patients with IBS. Material and methods: The investigation was conducted on total 168 patients in the clinical pathophysiology laboratory for the period June 2014 - September 2015. Male - 41 (24.4 %), mean age was 40,1 ± 17,2 years, female - 127 (75.6 %), mean age - 44,1 ± 15,3 years. The average age of patients older than 60 years was 69,4 ± 7,7 years. All patients fulfilling the Rome III criteria for IBS, and the prevalence of constipation was observed in 109 (64.9 %), diarrhea - in 29 (17.3 %), unstable stool (alternating diarrhea and constipation) - 30 (17.8 %).Rectal motility was evaluated by using air-filling catheters with manometry system Solar GI (MMS/Laborie, The Netherlands). We studied the background activity of the rectum and meal stimulated motility in both phases of gastrocolic reflex in patients with different clinical manifestations of IBS. Results: We diagnosed “irritable colon” type of motor activity in 56 (33.3 %) of 168 patients by analyzing the background motility. Irritable colon was found in 25 (22.9 %) of 109 patients with constipation, in 12 (41.4 %) of 29 patients with diarrhea and in 19 (63.3 %) of 30 patients with unstable stool. Remaining patients had hypokinetic background motility of the rectum, regardless of clinical manifestations (even in patients with diarrhea). Irritable bowel syndrome after eating was diagnosed in 111 (66.1 %) of 168 patients. They are 63 (57.8 %) patients with constipation, 20 (68.9 %) patients with diarrhea and 28 (96.6 %) patients with an unstable chair. The response to stimulation was not expressed in the remaining patients. They preserved hypokinetic motility type. Conclusions: Balloon manometry reveals a certain correlation between the clinical picture of IBS (the prevalence of constipation, diarrhea, or an unstable chair) and rectal motility type. This allows recommending balloon manometry as method of functional studies in patients with IBS.

      1. Соколов Л. К., Минушкин О. Н., Саврасов В. М., Терновой С. К. Клинико-инструментальная диагностика болезней органов гепатопанкреатодуоденальной зоны. - М.: Медицина. - 1987. - 279 с.
      2. Ступин В. А. Функциональная гастроэнтерология. Инструментальные методы исследования. Пособие для врачей // М.:2009. -27с.
      3. Турскова И. И. Гастроинтестинальная моторика и ее связь с некоторыми показателями вегетативного баланса при язвенной болезни/И.И. Турскова//Клин. Медицина,2002, № 8, - С. 38-41.
      4. Хендерсон Дж. М. Патофизиология органов пищеварения // М., Бином, С-Пб., Невский диалект. - 1997. - С. 65-82.
      5. Чернякевич С. А. Моторная функция желудка и двенадцатиперстной кишки при дуоденальной язве и ее осложнениях//Росс, журнал гастроэнтерологии, гепатологии, колопроктологии. 1995, Т.V, № 4. - С. 55-60.
      6. Чурин Б. В. Фронт двигательной активности в процессе периодической деятельности пищеварительного тракта у здоровых и больных язвенной болезнью//Росс, журнал гастроэнтерологии, гепатологии, колопроктологии., 2001., Т.ХI., № 2. - С. 38-43.
      7. Chen J., Richards R. D., McCallum R. W. The cutaneous electrogastrogram reveals important information about gastric motility// Gastroenterology. 1990. Vol. 99. A1208.
      8. Cucchiara S. A normal gastrointestinal motility excludes chronic intestinal pseudoobstruction in children/S. Cucchiara, O. Borrelli, G. Sulvia et al.//Dig.Dis.Sci. - 2000. - Vol.45,№ 2. - P. 258-264
      9. Karantanos T, Markoutsaki T, Gazouli M, Anagnou NP, Karamanolis DG Current insights in to the pathophysiology of Irritable Bowel Syndrome // Gut Pathog. 2010 May 13;2(1):3.
      10. Levy J. Use of electrogastrography in children. Curr Gastroenterol Rep. 2002 Jun; 4(3):259-65.
      11. Бачев И. И. Современные методы исследования моторной деятельности пищеварительного тракта // Хирургия. - 1978. - № 8. - С. 125-128.
      12. Буянов В. М. Ультразвуковое исследование в диагностике механической кишечной непроходимости/В.М. Буянов В. Д. Ишутинов, И. А. Доралев// Медиц, радиология, 1993. № 4. - С. 11-13.
      13. Саблин О. А., Гриневич В. Б., Успенский Ю. П., Ратников В. А. Функциональная диагностика в гастроэнтерологии. Учебно-методическое пособие// Сп.-Б., - 2002., 38с.
      14. Смирнова Г. О., Силуянов С. В. Периферическая электрогастроэнтерография в клинической практике. Пособие для врачей // М.:2009. -19с
      15. Калинин А. В. Физиологические и клинические аспекты нарушений моторики толстой кишки. Возможности фармакологической коррекции//Клинические перспективы гастроэнтерологии, гепатологии.2001, № 4. - С. 25-32.
      16. Ревин Г. О. Моторная функция толстой кишки после стволовой ваготомии с пилоропластикой: Клинико-экспериментальное исследование // Автореф. дис. … канд.мед.наук., Сп-Б.,2003.-21с.
      17. Celebi Kobak A, Bor S. The acute effect of oral ethanol intake on gastric myoelectrical activity in healthy volunteers // Turk J Gastroenterol. 2007 Dec;18(4):221-4.
      18. Chen J; McCallum RW. Electrogastrographic parameters and their clinical significance. In: Chen JZ, McCallum RW, eds. Electrogastrography, Principles and Applications. New York: Raven Press Ltd, 1994: 45-74.
      19. Cucchiara S. A normal gastrointestinal motility excludes chronic intestinal pseudoobstruction in children/S. Cucchiara, O. Borrelli, G. Sulvia et al.//Dig.Dis.Sci. - 2000. - Vol.45,№ 2. - P. 258-264.
      20. Miller T. G. Small intestinal intubation: experiences with a doublelumened tube/T.G. Miller, W. O. Abbolt//Ann.intern.Med. - 1934. - Vol. 8. - P. 85-92.
      21. Simonian HP; Panganamamula K; Parkman HP; Xu X; Chen JZ; Lindberg G; Xu H; Shao C; Ke MY; Lykke M; Hansen P; Barner B; Buhl H. Multichannel electrogastrography (EGG) in normal subjects: a multicenter study. Dig Dis Sci 2004 Apr; 49(4):594-601
      22. Wiesław T, Adam K, Artur B, Lech B, Krzysztof B. Nissen fundoplication improves gastric myoelectrical activity characteristics and symptoms in gastroesophageal reflux patients: evaluation in transcutaneous electrogastrography // Surg Endosc. 2008 Jan;22(1):134-40. Epub 2007 May 12.
      23. Bassotti G, Gaburri M, Imbimbo BP, Rossi L, Farroni F, Pelli MA, Morelli A. Colonic mass movements in idiopathic chronic constipation. Gut.1988; 29:1173-1179.
      24. Bassotti G, Betti C, Pelli MA, Morelli A. Extensive investigation on colonic motility with pharmacological testing is useful for selecting surgical options in patients with inertia colica. Am J Gastroenterol. 1992;87:143-147.
      25. Rao SS, Sadeghi P, Beaty J, Kavlock R. Ambulatory 24-hour colonic manometry in slow-transit constipation. Am J Gastroenterol. 2004;99:2405-2416.
      26. Camilleri M, Bharucha AE, di Lorenzo C, Hasler WL, Prather CM, Rao SS, Wald A. American Neurogastroenterology and Motility Society consensus statement on intraluminal measurement of gastrointestinal and colonic motility in clinical practice. Neurogastroenterol Motil. 2008;20:1269-1282.
      27. Philip G Dinning, Marc A Benninga, Bridget R Southwell, S Mark Scott. Paediatric and adult colonic manometry: A tool to help unravel the pathophysiology of constipation. World J Gastroenterol. 2010 November 7; 16(41): 5162-5172
      28. Лебедев Н. Н. Пищеварительная и внепищеварительная моторика ЖКТ в функциональнной диагностики. // Материалы пленума правления ВНОГ. - Рига, - 1986. - с. 232-234.
      29. Салмин И. П. Рефлекторный механизм перистальтики. / Тезисы докладов на заседании 9-го съезда Всероссийского общества физиологов, биохимиков. Фармакологов. - Минск. - 1953
      30. Kreulen DL, Szurszewski JH. Reflex pathways in the abdominal prevertebral ganglia: evidence for a colo-colonic inhibitory reflex.//J Physiol. -1979. - 295. - 21-32.
      31. Bazzochi G, Ellis J, Villlanueva-Meyr et al. Postprandial colonic transit and motor activity in chronic constipation. // Gastroenterol. - 1990. - 98. - 686-93.
      32. Kerlin P, Zinsmeister A, Phillips S. Motor responses to food of the ileum, proximal colon and distal colon of healthy humans. // Gastrotnterology. - 1983. - 84. - 4. - 762-770.
      33. Orkin BA, Hanson RB, Kelly KA, et al. Human anal motility while fasting, and during sleep. //Gastroenterology. - 1991. - 100. - 4. - 1016-23.
      34. Wald A. Colonic and anorectal motility testing in clinical practice. //Am J Gastroenterol. - 1994. - 89. - 12. - 2108.
      35. Большой толковый медицинский словарь. 2001.
      36. Подмаренкова Л. Ф. Автореф.дисс…док.мед.наук. М. - 2000.
      37. Яковенко Э. П., Агафонова Н. А. Механизмы развития запоров и методы их лечения. Клинический перспективы гастроэнтерологии, гепатологии, 2003, 3, с. 25-32.
      38. Drossman D. A. The functional gastrointestinal disorders and the Rome III Process. Gastroenterology 2006; 130(5):1377-90.
      39. Bixquert Jiménez M. Treatment of irritable bowel syndrome with probiotics. An etiopathogenic approach at last? Rev Esp Enferm Dig 2009; 101: 553-564.
      40. Brian E. Lacy, Fermin Mearin, Lin Chang, William D. Chey et al. Bowel disorders. Gastroenterology. 2016. 150. 1393-1399.
      41. Шелыгин Ю. А., Титов А. Ю., Джанаев Ю. А., Бирюков О. М., Мудров А. А., Краснопольская И. В. Особенности клинической картины и характер нейро-функциональных нарушений у больных ректоцеле// Колопроктология. 2012, № 4(42). - С. 27-33
     


    Full text is published :
    Fomenko O.Yu., Halif I.L., Aleshin D.V., Belousova S.V. RECTAL NONPERFUSION MANOMETRY AS A WAY OF RECTAL MOTILITY ASSESSMENT IN PATIENTS WITH IRRITABLE BOWEL SYNDROME. Experimental and Clinical Gastroenterology Journal. 2017;139(03):32-37
    Read & Download full text

    1. Moscow state medical dental University named A. I. Evdokimov Ministry of health of the Russian Federation (Moscow, Russian Federation)

    Keywords:comorbid pathology,functional constipation,non-alcoholic steatohepatitis,treatment,psyllium,lactulose

    Abstract:The aim of the study was to Evaluate the effectiveness of long-term therapy dietary fiber (DF) or lactulose on the course of functional constipation when combined with nonalcoholic steatohepatitis (NASH) Material and methods. Was conducted an open prospective comparative study in parallel groups with the inclusion of 102 patients with NASH, among whom were allocated to patients with symptoms of functional constipation. The main group consisted of 70 people (NASH and functional constipation), a comparison group of 32 people with just NASH. The main group patients were randomized into two equal subgroups of 30 people IA and IB, differentiated by the treatment of functional constipation: patients of subgroup IA in addition to modification of lifestyle was prescribed the drug lactulose, patients IB - preparation of dietary fiber (psyllium). All patients 1 time per 3 months was carried out dynamic assessment of specific markers of functional constipation, frequency of defecation, stool type, indicators of quality of life, check bacterial overgrowth syndrome. The observation period of the patients was 6 months. Results. For the period 6-month observation the most significant regression of markers of functional constipation (natureline, frequency of defecation), better tolerance of therapy, including less pronounced side effects significantly greater reduction of bacterial overgrowth syndrome (p < 0.05) was achieved only in the subgroup of dietary fiber, which is reflected in a statistically more significant improvement in quality of life than in the subgroup of lactulose. Conclusion: long-term treatment of functional constipation with psyllium is characterized by a more pronounced laxative effect, better tolerated, reduces the severity bacterial overgrowth syndrome that regularly impact on improving the quality of life compared with lactulose.

      1. Overweight and gastrointestinal symptoms among adults of working age in Iaşi City, Romania / I. Chirila, VL. Drug, FD. Petrariu [et al.] // Rev Med Chir Soc Med Nat Iasi. - 2012. - Vol.116, № 1. - P. 268-73.
      2. Talley, NJ. Functional gastrointestinal disorders as a public health problem / N. J. Talley // Neurogastroenterol Motil. - 2008. - Vol. 20(Suppl 1). - P. 121-9.
      3. Клинические рекомендации Российской гастроэнтерологической ассоциации по диагностике и лечению взрослых пациентов с хроническим запором / В. Т. Ивашкин, С. Р. Абдулхаков, Е. К. Баранская [и др.] // Росс. журн. гастроэнтерол., гепатол., колопроктол. - 2014. - № 5. - С. 69-75.
      4. Pare, P. The approach to diagnosis and treatment of chronic constipation: suggestions for a general practitioner / P. Pare // Can. J. Gastroenterol. - 2011. - 25 (Suppl B). - P. 36B-40B.
      5. Locke, G.III. American Gastroenterological Association Medical Position Statement: guidelaines on constipation / G.III. Locke, J. Pemberton, S. Philips // Gastroenterology. - 2000. - Vol. 119, № 6. - P. 1761-1766.
      6. Опыт применения препарата Мукофальк в комплексной терапии у пациентов с ожирением и метаболическим синдромом. Отчет НИИ диетологии и диетотерапии [Электронный ресурс] / И. В. Губарева, Т. Е. Чембарова, М. М. Гинзбург [и др.] // Самара, 2010. - Режим доступа: www.mucofalk.ru/download/ginsburg.pdf
      7. Чиркин, В. И. Долгосрочные эффекты препарата пищевых волокон псиллума (Мукофальк) у пациентов с метаболическим синдромом / В. И. Чиркин, И. А. Лазарев, М. Д. Ардатская // Клин. перспективы гастроэнтерол., гепатол. - 2012. - № 1. - С. 34-42.
      8. Pal, S. Effects of psyllium on metabolic syndrome risk factors / S. Pal, S. Radavelli-Bagatini // Obes Rev. - 2012. - Vol. 13, № 11. - P. 1034-47.
      9. Полевая, Е. В. Энтеросорбционные свойства псиллиума (Мукофалька) и возможные механизмы его действия при кишечных инфекциях / Е. В. Полевая, Т. Я. Вахитов, С. И. Ситкин // Клин. перспективы гастроэнтерол., гепатол. - 2011. - № 2. - С. 35-39.
      10. Особенности течения неалкогольного стеатогепатита, ассоциированного с хроническим запором / Маевская Е. А., Маев И. В., Кучерявый Ю. А. [и др.] // Экспериментальная и клиническая гастроэнтерология. - 2015. - № 9. - С. 36-44.
      11. Оценка влияния лактулозы или пищевых волокон на динамику показателей липидного профиля у пациентов с функциональным запором и неалкогольным стеатогепатитом / Маевская Е. А., Маев И. В., Кучерявый Ю. А. [и др.] // Лечащий врач. - 2016. - № 4. - С. 1-6.
      12. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association / N. Chalasani, Z. Younossi, JE. Lavine [et al.] // Hepatology. - 2012. - Vol. 55, № 6. - P. 2005-23.
      13. Drossman DA. The Functional Gastrointestinal Disorders and the Rome III Process // Gastroenterology. - 2006. - Vol. 130. - P. 1377-1390.
      14. Маевская М. В., Шульпекова О. Ю., Буеверов О. А. и соавт. Диагностика и лечение неалкогольной жировой болезни печени / Под ред.Ивашкина В. Т. Методические рекомендации для врачей. М., 2015. 32 с.
      15. World Gastroenterology Organisation global guidelines: Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis / LaBrecque D. R., Abbas Z., Anania F. [et al]. // J Clin Gastroenterol. - 2014. - Vol.48, № 6. - P. 467-473.
      16. World Gastroenterology Organisation Global Guidelines// Constipation: a global perspective. November 2010. www.worldgastroenterology.org/constipation. Html.
      17. Ware JE, Kosinski M, Keller SD. SF-36 Physical and Mental Health Summary Scales: A User’s Manual. Boston, MA: Th e Health Institute; 1994.
      18. Development and validation of the Patient Assessment of Constipation Quality of Life questionnaire / P. Marquis, C. De La Loge, D. Dubois [et al.] // Scand J Gastroenterol. - 2005. - Vol. 40, № 5. - P. 540-51.
      19. Черемушкин, С. В. Доказательная медицина и лечение хронического запора / С. В. Черемушкин, Ю. А. Кучерявый // Врач. - 2012. - № 10. - С. 42-47.
      20. Consensus statement AIGO/SICCR diagnosis and treatment of chronic constipation and obstructed defecation (part II: treatment) / A. Bove, M. Bellini, E. Battaglia [et al.] // World J Gastroenterol. - 2012. - Vol. 18, № 36. - P. 4994-5013.
      21. Liu, L. W. Chronic constipation: current treatment options / L. W. Liu // Can. J. Gastroenterol. - 2011. - Vol. 25 (Suppl B). - P. 22-28.
      22. Primary Care Management of Chronic Constipation in Asia: The ANMA Chronic Constipation Tool / KA. Gwee, UC. Ghoshal, S. Gonlachanvit [et al.] // J Neurogastroenterol Motil. - 2013. - Vol. 19, № 2. - Р. 149-60.
      23. Pare, P. Systematic review of stimulant and nonstimulant laxatives for the treatment of functional constipation / P. Pare, RN. Fedorak // Can J Gastroenterol Hepatol. - 2014. - Vol. 28, № 10. - P. 549-57.
      24. Mueller-Lissner, SA. Constipation in adults [Электронный ресурс] / SA. Mueller-Lissner, A. Wald // BMJ Clin Evid. - 2010. - Vol. 2010. - P. 0413. - Режим доступа: www.ncbi.nlm.nih.gov/pmc/articles/PMC 3217654/
      25. Randomised, controlled comparison of low-dose polyethylene glycol 3350 plus electrolytes with Ispaghula Husk in the treatment of adults with chronic functional constipation / H. J. Wang, X. M. Liang, Z. L. Yu [et al.] // Clin Drug Investig. - 2004. - Vol. 24, № 10. - P. 569-76.
      26. The treatment of chronic constipation in adults. A systematic review / S. M. Tramonte, M. B. Brand, C. D. Mulrow [et al.] // J. Gen. Intern. Med. - 1997. - Vol. 12. - P. 15-24.
      27. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation / A. Attaluri, R. Danahoe, J. Valestin [et al.] // Aliment Pharmacol Ther. - 2011. - Vol. 33, № 7. - P. 822-8.
      28. Effect of dietary fiber on constipation: a meta analysis / J. Yang, H. P. Wang, L. Zhou [et al.] // World J Gastroenterol. - 2012. - Vol. 18, № 48. - P. 7378-83.
      29. Ruiz-López, MC. Quality of life in patients with different constipation subtypes based on the Rome III criteria / MC. Ruiz-López, E. Coss-Adame // Rev Gastroenterol Mex. - 2015. - Vol. 80, № 1. - P. 13-20.
      30. Sanchez, MI. Epidemiology and burden of chronic constipation / MI, Sanchez, P. Bercik // Can J Gastroenterol. - 2011. - Vol. 25(Suppl B). - P. 11B-15B.
      31. Неалкогольный стеатогепатит и кишечная микрофлора: есть ли потенциал пребиотических препаратов в лечении?/ Ю. А. Кучерявый, Е. А. Маевская, М. Л. Ахтаева [и др.] // Мед. совет. - 2013. - № 3 (2). - С. 46-51.
      32. Маевская, Е. А. Кишечная микрофлора и неалкогольный стеатогепатит: от механизмов патогенеза к терапии / Е. А. Маевская, Ю. А. Кучерявый, И. В. Маев // Леч. Врач. - 2014. - № 8. - С. 45-51.
      33. Буеверов, А. О. Цитокиновый взрыв и прогрессирование неалкогольного стеатогепатита / А. О. Буеверов, П. О. Богомолов // Клин. перспективы гастроэнтерол., гепатол. - 2012. - № 5. - С. 12-18.
      34. Obesity and functional constipation; a community-based study in Iran / MA. Pourhoseingholi, SA. Kaboli, A. Pourhoseingholi [et al.] // J Gastrointestin Liver Dis. - 2009. - Vol.18, № 2. - P. 151-5.
      35. Mozaffari, S. The safety of novel drugs used to treat irritable bowel syndrome / S. Mozaffari, S. Nikfar, M. Abdollahi // Expert Opin Drug Saf. - 2014. - Vol. 13, № 5. - P. 625-38.
      36. The efficacy and safety of rifaximin for the irritable bowel syndrome: a systematic review and meta-analysis / SB. Menees, M. Maneerattannaporn, HM. Kim [et al.] // Am J Gastroenterol. - 2012. - Vol. 107, № 1. - P. 28-35.
      37. Small intestinal bacterial overgrowth syndrome / J. Bures, J. Cyrany, D. Kohoutova [et al.] // World J. Gastroenterol. - 2010. - Vol. 16, № 24 - Р. 2978-2990.
      38. Abnormal breath testing in IBS: a meta-analysis / Shah ED, Basseri RJ, Chong K [et al.] // Dig Dis Sci. - 2010. - Vol.55, № 9. - P. 441-9.
      39. Psyllium improves fecal consistency and prevents enhanced secretory responses in jejunal tissues of piglets infected with ETEC / U.L Hayden., S.M McGuirk., S. E. West [et al.] // Dig. Dis. Sci. - 1998. - Vol. 43, № 11. - P. 2536-2541.
      40. Salminen, S. Lactulose, lactic acid bacteria, intestinal microecology and mucosal protection / S. Salminen, E. Salminen // Scand. J. Gastroenterol. - 1997. - Vol. 222. - Р. 45-48.
      41. Panesar, P. S. Lactulose: production, purification and potential applications / P.S Panesar, S. Kumari // Biotechnol. Adv. - 2011. - Vol. 29, № 6. - Р. 940-948.
     


    Full text is published :
    Maevskaya E.A., Cucheryavy Y.A., Cheremushkin S.V., Shestakov V.A. et. al. THE EFFECTIVENESS OF PSYLLIUM AND LACTULOSE IN TREATMENT OF FUNCTIONAL CONSTIPATION WHEN COMBINED WITH NONALCOHOLIC STEATOHEPATITIS. Experimental and Clinical Gastroenterology Journal. 2017;139(03):38-47
    Read & Download full text

    1. Volgograd state medical University (Volgograd, Russian Federation)
    2. First Moscow state medical University. I. M. Sechenov, the Ministry of health of the Russian Federation (Moscow, Russian Federation)

    Keywords:irritable bowel syndrome,systemic inflammation,cytokines

    Abstract:In the presented article the data gained from recent publications and our original investigations conducted in term to determine the role of low grade systemic inflammation in the pathogenesis and clinical manifestation of irritable bowel syndrome (IBS) were analyzed. It was shown that in patients with IBS levels of circulating key pro-inflammatory cytokines (TNFα, IL-1, IL-6, IL-8) were increased while the levels of anti-inflammatory IL-10 tends to decrease. Revealed cytokine imbalance correlated with severity of the intestinal symptoms such as abdominal pain and changes in bowel habits. The raised levels of pro-inflammatory cytokines were detected in 30-40 % pts with IBS and in 60-85 % pts with ulcerative colitis (UC) versus 2-6 % in controls. In pts with active UC pro-inflammatory cytokines raised much more prominent than in IBS, nevertheless in IBS group levels of these cytokines were significantly higher than in controls. The results of randomized placebo controlled study had demonstrated the efficacy and safety of combination of affinity purified release-active antibodies (RAAT) to histamine, TNFα and brain-specific S-100 protein with anti-cytokine property in the treatment of IBS. In pts treated with combination of affinity purified RAAT to histamine, TNF-α and brain-specific S-100 protein the significant improvement in intestinal and non-intestinal systemic symptoms was registered. These data support the concept of relationship between cytokine imbalance and functional GI disorders and suggest the recommendation to include combination of affinity purified RAAT to histamine, TNF-α and brain-specific S-100 protein as disease modifying drug in IBS treatment.

      1. Авалуева Е. Б., Адашева Т. В., Бабаева А. Р. Эффективность и безопасность применения Колофорта при синдроме раздраженного кишечника: итоги многоцентрового двойного плацебо-контролируемого рандомизированного клинического исследования//Consilium medicum. Гастроэнтерология. - 2014. - № 1: С. 2-7
      2. Бабаева А.Р, Солоденкова К. С., Сергеева С. А. Доказательная база антицитокиновой терапии ревматоидного артрита.// Вестник ВолГМУ.-2006.-№ 4.-С.15-22
      3. Елизаветина Г. А. Эволюция взглядов на этиологию, патогенез и лечение синдрома раздраженного кишечника.// Медицинский совет. - 2015. - № 13. - С. 86-89.
      4. Осадчук А. М., Осадчук М. А. Артрофоон-альтернативный препарат в лечении неосложненных форм специфического язвенного колита.// Бюл. экспер. биол.-2009.-Т.148.-№ 8.-Приложение. - С. 121-124.
      5. Осадчук М. А. Колофорт новый препарат в терапии функциональных расстройств кишечника.//Медицинский альманах,-2015,-№ 1(36),-С. 57-59.
      6. Осадчук М. А., Будрина В. О. Новые патогенетические подходы к терапии синдрома раздраженного кишечника, основанные на морфофункциональных особенностях данной патологии. // Практическая медицина, - 2014, - № 1(77). - С.12-20.
      7. Родионова О.Н, Трубина Н. В., Реутова Э. Ю., Видикер Р. В., Бабаева А. Р. Особенности нарушений нейрогуморальной регуляции, цитокинового и тиреоидного статуса у больных с функциональными расстройствами желудочно-кишечного тракта // Вестник Санкт-Петербургского университета. - Выпуск 1 (11). - 2009. - С. 51-57.
      8. Родионова О.Н, Трубина Н.В, Реутова Э.Ю, Видикер Р. В., Бабаева А.Р, Гальченко О. Е. Клиническое значение оценки нейромедиаторов и цитокинового статуса у больных функциональными заболеваниями желудочно-кишечного тракта//Вестник Волгоградского государственного медицинского университета, -2008 - № 3 (27) - С. 44-47.
      9. Шептулин А. А. СРК: Что мы видим “внутри хрустального шара”?.//РМЖ, Гастроэнтерология, - 2014 № 20, - С.1437-1441.
      10. An S, Zong G, Wang Z, Shi J, Du H, Hu J. 2016 Mar 4. Expression of inducible nitric oxide synthase in mast cells contributes to the regulation of inflammatory cytokines in irritable bowel syndrome with diarrhea. //Neurogastroenterol Motil. 2016 Jul;28(7):1083-93. doi: 10.1111/nmo.12811. Epub
      11. Barbaro MR, Di Sabatino A, Cremon C, Giuffrida P, Fiorentino M, Altimari A, Bellacosa L, Stanghellini V, Barbara G. Interferon-γ is increased in the gut of patients with irritable bowel syndromeand modulates serotonin metabolism. //Am J Physiol Gastrointest Liver Physiol. 2016 Mar 15;310(6): G439-47. doi:10.1152/ajpgi.00368.2015. Epub 2016 Jan 7.
      12. Bennet SM, Polster A, Törnblom H, Isaksson S, Capronnier S, Tessier A, Le Nevé B, Simrén M, Öhman L. Global Cytokine Profiles and Association With Clinical Characteristics in Patients With Irritable Bowel Syndrome. //Am J Gastroenterol. 2016 Aug;111(8):1165-76. doi: 10.1038/ajg.2016.223. Epub 2016 Jun 7.
      13. Brzozowski B, Mazur-Bialy A, Pajdo R, Kwiecien S, Bilski J, Zwolinska-WcisloM, Mach T, Brzozowski T. Mechanisms by which Stress Affects the Experimental and Clinical InflammatoryBowel Disease (IBD). Role of Brain-Gut Axis. //Curr Neuropharmacol. 2016 Apr 4. [Epub ahead of print]
      14. Ciszek BP, Khan AA, Dang H, Slade GD, Smith S, Bair E, Maixner W, Zolnoun D, Nackley AG. MicroRNA expression profiles differentiate chronic pain condition subtypes. //Transl Res. 2015 Dec;166(6):706-720.e11. doi: 10.1016/j.trsl.2015.06.008. Epub2015 Jun 24.
      15. Cheung CK, Wu JC. Genetic polymorphism in pathogenesis of irritable bowel syndrome.// World J Gastroenterol. 2014 Dec 21;20(47):17693-8.
      16. Hustoft TN, Hausken T, Ystad SO, Valeur J, Brokstad K, Hatlebakk JG, Lied GA. Effects of varying dietary content of fermentable short-chain carbohydrates onsymptoms, fecal microenvironment, and cytokine profiles in patients withirritable bowel syndrome.// Neurogastroenterol Motil. 2016 Oct 16. doi: 10.1111/nmo.12969. [Epub ahead of print
      17. Jonefjäll B, Öhman L, Simrén M, Strid H. IBS-like Symptoms in Patients with Ulcerative Colitis in Deep Remission Are Associated with Increased Levels of Serum Cytokines and Poor Psychological Well-beingЮ// Inflamm Bowel Dis. 2016 Nov;22(11):2630-2640.
      18. Keightley PC, Koloski NA, Talley NJ. Pathways in gut-brain communication: evidence for distinct gut-to-brain and brain-to-gut syndromes. //Aust N Z J Psychiatry. 2015 Mar;49(3):207-14. doi: 10.1177/0004867415569801.
      19. Khanijow V, Prakash P, Emsellem HA, Borum ML, Doman DB. Sleep Dysfunction and Gastrointestinal Diseases.// Gastroenterol Hepatol (N Y). 2015 Dec;11(12):817-25.
      20. Martin-Viñas JJ, Quigley EM. Immune response in irritable bowel syndrome: A systematic review of systemic and mucosal inflammatory mediators.// J Dig Dis. 2016 Sep;17(9):572-581. doi: 10.1111/1751-2980.12379.
      21. O’Malley D. Immunomodulation of enteric neural function in irritable bowel syndrome.// World J Gastroenterol. 2015 Jun 28;21(24):7362-6. doi: 10.3748/wjg.v21.i24.7362.
      22. Pike BL, Paden KA, Alcala AN, Jaep KM, Gormley RP, Maue AC, Christmann BS, Elson CO, Riddle MS, Porter CK. Immunological Biomarkers in Postinfectious Irritable Bowel Syndrome //J Travel Med. 2015 Jul-Aug;22(4):242-50. doi: 10.1111/jtm.12218. Epub 2015 Jun 8.
      23. Popa SL, Dumitrascu DL. Anxiety and IBS revisited: ten years later. //Clujul Med. 2015;88(3):253-7. doi: 10.15386/cjmed-495. Epub 2015 Jul 1.
      24. Staud R. Cytokine and immune system abnormalities in fibromyalgia and other central sensitivity syndromes.// Curr Rheumatol Rev. 2015;11(2):109-15.
      25. Sundin J, Rangel I, Repsilber D, Brummer RJ. Cytokine. Response after Stimulation with Key Commensal Bacteria Differ in Post-Infectious Irritable Bowel Syndrome (PI-IBS) Patients Compared to Healthy//Controls. PLoS One. 2015 Sep 14;10(9): e0134836.doi:10.1371/journal.pone.0134836.Collection 2015.
      26. Zhang G, Yu L, Chen ZY, Zhu JS, Hua R, Qin X, Cao JL, Zhang YM. Activation of corticotropin-releasing factor neurons and microglia inparaventricular nucleus precipitates visceral hypersensitivity induced bycolorectal distension in rats. //Brain Behav Immun. 2016 Jul;55:93-104. doi: 10.1016/j.bbi.2015.12.022. Epub 2015 Dec 29.
      27. Zhen Y, Chu C, Zhou S, Qi M, Shu R. Imbalance of tumor necrosis factor-α, interleukin-8 and interleukin-10 productionevokes barrier dysfunction, severe abdominal symptoms and psychological disorders in patients with irritable bowel syndrome-associated diarrhea.// Mol Med Rep. 2015 Oct;12(4):5239-45. doi: 10.3892/mmr.2015.4079. Epub 2015 Jul 5
     


    Full text is published :
    Babaeva A.P., Osadchuk M.A., Vidiker R.V., Kalinina E.V. et. al. MARKERS OF SYSTEMIC INFLAMMATION IN PATHOGENESIS AND OPTIMIZATION OF PHARMACOTHERAPY OF IRRITABLE BOWEL SYNDROME. Experimental and Clinical Gastroenterology Journal. 2017;139(03):48-55
    Read & Download full text

    1. State scientific centre of coloproctology (Moscow, Russian Federation)

    Keywords:idiopathic megacolon,idiopathic megarectum,longitudinal proctoplasty

    Abstract:The study focuses on surgical treatment of idiopathic megacolon and megarectum. Methods: 36 patients (18-78 years, 20 male) were operated in 2004-2014. 25 (69.4 %) patients had idiopathic megacolon. 11 (30.6 %) patients had idiopathic megarectum with or without megacolon. The choice of surgical procedure depended on extent and localization of megacolon and function of nondilated colon. Left colectomy was performed in 12 (33.3 %) cases, subtotal colectomy and ascendorectal anastomosis - in 13 (36.1 %) cases. All patients with idiopathic megarectum were undergone longitudinal proctoplasty combined with left colectomy or subtotal colectomy. Results: After surgery 1 (2.8 %) patient died and 5 (13.9 %) had complications that required reoperation. In follow up 1 patient have got megacolon recurrence. 9 of idiopathic megarectum patients whom stoma was closed were followed up. There were not megacolon or megarectum recurrence. In 8 (88.9 %) patients were detected significant improvement and satisfaction with surgery. There also was found significant reduction of constipation rate and rectal function improvement. No one patient had sexual or urological impairment. Conclusion: In cases of idiopathic megacolon only dilated colon resection may be justified in selected patients with good function of nondilated colon. Longitudinal proctoplasty leads to significant improvement of rectal function and may be used in idiopathic megarectum surgical treatment.%

      1. Ачкасов С. И., Алешин Д. В. Способ хирургического лечения идиопатического мегаректум. Патент № 2516118 Приоритет изобретения 20.12.2012.
      2. Воробьев Г.И., Жученко А. П., Ачкасов С. И., Капуллер Л. Л., Бирюков О. М. Возможности модификации биопсии стенки прямой кишки по Свенсону в диагностике пороков развития интрамуральной нервной системы у взрослых. Хирургия, 2005, № 10, с. 4-7.
      3. Воробьев Г. И. Мегаколон у взрослых (клиника, диагностика и хирургическое лечение). Диссертация докт. мед наук. Москва, 1982. 402 с.
      4. Ding W1, Jiang J, Feng X, Yao A, Liu J, Li N, Li J. Outcomes after surgery for refractory constipation patients complicated with megacolon. Zhonghua Wei Chang Wai Ke Za Zhi. 2014 May;17(5):453-6.
      5. Gattuso J.M., Kamm M. A., Talbot I. C. Pathology of idiopathic megarectum and megacolon. Gut 1997; 41: 252-257.
      6. Gattuso JM, Kamm MA. Clinical features of idiopathic megarectum and idiopathic megacolon. Gut. 1997;41:93
      7. Gladman MA, Scott SM, Lunniss PJ, Williams NS. Systematic review of surgical options for idiopathic megarectum and megacolon. Ann Surg 2005; 241: 562-74
      8. Gladman MA, Lunniss PJ, Williams NS, Scott SM. Rectal hyposensitivity: pathophysiological mechanisms. Neurogastroenterol Motil 2005; 17(Suppl. 2): 18.
      9. Huang ZC 1, Liu Q, Li SG, Li D, Su J, Yan DY, Xiao ZG, Dong HY, Zhou K. Diagnosis and treatment of slow transit constipation complicated with adult megacolon. Zhonghua Wei Chang Wai Ke Za Zhi. 2011 Dec;14(12):941-3.
      10. Waldron D, Bowes KL, Kingma YJ, Cote KR. Colonic and anorectal motility in young women with severe idiopathic constipation. Gastroenterology 1988; 95: 1388-1394.
      11. Williams NS, Fajobi OA, Lunniss PJ, Scott SM, Eccersley AJP, Ogunbiyi OA. Vertical reduction rectoplasty: a new treatment for idiopathic megarectum. Br J Surg 2000; 87: 1203-1208.
      12. Varma JS, Smith AN. Neurophysiological dysfunction in young women with intractable constipation. Gut 1988; 29: 963-968.
     


    Full text is published :
    Aleshin O.V., Achkasov S.I., Fomenko O.Yu. SURGICAL TREATMENT OF IDIOPATHIC MEGACOLON AND MEGARECTUM. Experimental and Clinical Gastroenterology Journal. 2017;139(03):56-59
    Read & Download full text

    1. RUDN University (Moscow, Russian Federation)

    Keywords:colorectal cancer,colonic obstruction,stenting

    Abstract:Colorectal cancer is one of the common forms of malignancies in the world. Despite advances in the diagnosis of early stage of the disease in a large enough number of patients develop clinical bowel obstruction. Colorectal stenting is a minimally invasive alternative to emergency surgery, associated with high morbidity and mortality. Since its first application in 1991, has accumulated a lot of experience not only as palliative care for inoperable patients, but also as a «bridge to surgery» for radical treatment. In this review, the literature data relating to various techniques, indications and contraindications, results and complications of stenting in colonic obstruction.

      1. Alcantara M., Serra-Aracil X., Falco J. et al. Prospective, controlled, randomized study of intraoperative colonic lavage versus stent placement in obstructive left-sided colonic cancer.World J Surg, 2011, Vol.35, pp.1904-10.
      2. Baron T. Technique of colonic stenting. Techniques in Gastrointestinal Endoscopy, 2014, Vol.16, рр.108-111.
      3. Cennamo V., Luigiano C., Coccolini F., et al. Meta-analysis of randomized trials comparing endoscopic stenting and surgical decompression for colorectal cancer obstruction. Int J Colorectal Dis, 2013, Vol.28, pp.855-863.
      4. Chang IS., Park SW., Hwang D-Y. The Eficacy of the Coaxial Technique Using a 6-Fr Introducer Sheath in Stent Placement for Treating the Obstructions Proximal to the Descending Colon. Korean J Radiol, 2011, Vol.12, рр.107-112.
      5. Cheung DY., Lee YK., Yang CH. Status and Literature Review of Self-Expandable Metallic Stents for Malignant Colorectal Obstruction. Clinical Endoscopy,2014, Vol. 47, рр. 65-73.
      6. Cirocchi R., Farinella E., Trastulli S., et al. Safety and efficacy of endoscopic colonic stenting as a bridge to surgery in the management of intestinal obstruction due to left colon and rectal cancer: a systematic review and meta-analysis. Surg Oncol, 2013, Vol.22, pp. 14-21.
      7. De Ceglie A., Filiberti R., Baron TH., et al. A metaanalysis of endoscopic stenting as bridge to surgery versus emergency surgery for left-sided colorectal cancer obstruction. Crit Rev Oncol Hematol, 2013, Vol.88, pp.387-403.
      8. Dohmoto M., Rupp KD., Hohlbach G. Endoscopically-implanted prosthesis in rectal carcinoma. Dtsch Med Wochenschr 1990, Vol.115, рр.915.
      9. Fiori E., Lamazza A., Schillaci A., et al. Palliative management for patients with subacute obstruction and stage IV unresect-able rectosigmoid cancer: colostomy versus endoscopic stenting: final results of a prospective randomized trial. Am J Surg, 2012, Vol.204, pp.321-326.
      10. Govindarajan А., Naimark D., Coburn NG. Use of Colonic Stents in Emergent Malignant Left Colonic Obstruction: A Markov Chain Monte Carlo Decision Analysis. Dis Colon Rectum, 2007, Vol. 50, N. 11, pp.1811-24.
      11. Ho KS., Quah HM., Lim JF., et al. Endoscopic stenting and elective surgery versus emergency surgery for left-sided malignant colonic obstruction: a prospective randomized trial. Int J Colorectal Dis, 2012, Vol.27, pp.355-362.
      12. Huang X., Lv B., Zhang S., Meng L. Preoperative colonic stents versus emergency surgery for acute left-sided malignant colonic obstruction: a meta-analysis. J Gastrointest Surg, 2014, Vol.18, pp.584-591.
      13. Jeong JB., Lee KL., Kwon SB., et al. Palliative self-expanding metal stents in the treatment of malignant colorectal obstruction. Intest Res, 2010, Vol.8, рр.135-141.
      14. Kobayashi Y., Komazawa Y., Kusunoki M. Novel stenting method for malignant right colonic stenosis using ultra-thin endoscopy: Repor t of four cases. Digestive Endoscopy, 2015 Vol.27, рр.704-707
      15. Lee HJ., Park SJ., Cheon JH. What is the necessity of endoscopist for successful endoscopic stenting in patients with malignant colorectal obstruction? Int J Colorectal Dis. 2015, Vol.30, pp.119-125.
      16. Lee J., Byeon J. Colorectal Stents: Current Status. Clinical Endoscopy,2015, Vol. 48, рр.194-200.
      17. Lee KJ., Kim SW., KimTI., et al. Evidence-Based Recommendations on Colorectal Stenting: A Report from the Stent Study Group of the Korean Society of Gastrointestinal Endoscopy. Clinical Endoscopy, 2013, Vol.46, pp.355-367.
      18. Liang TW., Sun Y., Wei YC., et al. Palliative treatment of malignant colorectal obstruction caused by advanced malignancy: a self-expanding metallic stent or surgery? A system review and meta-analysis. Surg Today, 2014, Vol.44, pp. 22-33.
      19. Meisner S. Stent for palliation of advanced colorectal cancer. Techniques in Gastrointestinal Endoscopy, 2014, Vol.16, рр. 125-128.
      20. Park S., Cheon JH., Park JJ., Moon CM., Hong SP., Lee SK. Comparison of efficacies between stents for malignant colorectal obstruction: a randomized, prospective study. Gastrointest Endosc, 2010, Vol.72, рр. 304-10.
      21. Sagar J. Colorectal stents for the management of malignant colonic obstructions. Cochrane Database Syst Rev, 2011, Vol.11, CD 007378
      22. Sagar J. Role of colonic stents in the management of colorectal cancers. World J Gastrointest Endosc, 2016, Vol.8, N.4, pp.198-204.
      23. Sebastian S., Johnston S., Geoghegan T., et al. Pooled analysis of the efficacy and safety of self-expanding metal stenting in malignant colorectal obstruction. Am J Gastroenterol, 2004, Vol.99, pp.2051-2057.
      24. Shimura T., Joh T. Evidence-based Clinical Management of Acute Malignant Colorectal Obstruction. J Clin Gastroenterol Volume 50, Number 4, April 2016.
      25. Sloothaak D., van den Berg MM., Dijkgraaf M. Oncological outcome of malignant colonic obstruction in the Dutch Stent-In 2 trial. Br J Surg, 2014, Vol.101, pp. 1751-7.
      26. Song HY., Kim JH., Shin JH., et al. A dual-design expandable colorectal stent for malignant colorectal obstruction: results of a multicenter study. Endoscopy 2007, Vol.39, рр. 448-54.
      27. Song H-Y., Nam DH., Lee H. Usefulness of a Guiding Sheath for Fluoroscopic Colorectal Stent Placement. Korean J Radiol, 2012, Vol.13, Supp.1, S 83-88.
      28. Srinivasan N., Kozarek R. Stents for colonic strictures: Materials, designs, and more. Techniques in Gastrointestinal Endoscopy, 2014, Vol.16, рр.100-107.
      29. Tack J., Gevers AM., Rutgeerts P. Self-expandable metallic stents in the palliation of rectosigmoidal carcinoma: a follow-up study. Gastrointestinal Endoscopy 1998, Vol.48, рр.267-71.
      30. Tan CJ., Dasari BV., Gardiner K. Systematic review and metaanalysis of randomized clinical trials of self-expanding metallic stents as a bridge to surgery versus emergency surgery for malignant left-sided large bowel obstruction. Br J Surg, 2012, Vol.99, pp.469-476.
      31. Tejero E., Mainar A., Fernández L., et al. New procedure for the treatment of colorectal neoplastic obstructions. Dis Colon Rectum 1994, Vol.37, рр.1158-9.
      32. The role of endoscopy in the management of patients with known and suspected colonic obstruction and pseudo-obstruction. Gastrointestinal Endoscopy, 2010, Vol. 71, N. 4, pp.669-79.
      33. Tung KL., Cheung HY., Ng LW., et al. Endolaparoscopic approach versus conventional open surgery in the treatment of obstructing left-sided colon cancer: long-term follow up of a randomized trial. Asian J Endosc Surg, 2013, Vol.6, pp.78-81.
      34. van Halsema EE., van Hooft JE. Outcome and complications of stenting for malignant obstruction Techniques in Gastrointestinal Endoscopy, 2014, Vol.16, pp.129-134.
      35. van Hooft JE, Bemelman WA, Oldenburg B, et al. Colonic stenting versus emergency surgery for acute left-sided malignant colonic obstruction: a multicentre randomized trial. Lancet Oncol, 2011, Vol.12, рр.344-352.
      36. van Hooft JE et al. Self-expandable metal stents for obstructing colonic and extracolonic cancer: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline. Endoscopy, 2014, Vol.46, рр.990-1002.
      37. van Hooft JE., Fockens P., Marinelli AW., et al. Early closure of a multicenter randomized clinical trial of endoscopic stenting versus surgery for stage IV left-sided colorectal cancer. Endoscopy, 2008, Vol.40, pp.184-191.
      38. van den Berg E., Bergmann J. F., Ledeboer M. Radiological Position and Clinical Outcome of Preoperative Self-Expanding Metal Stents for Obstructing Colonic Cancer: A Single-Centre Cohort Study. Dig Surg, 2015, Vol.32, рр.262-268.
      39. World Cancer Report 2014/ Edited by Sterwart B. W. and Wild C. P. Lyon, 2014. p.630
      40. Xinopoulos D., Dimitroulopoulos D., Theodosopoulos T., et al. Stenting or stoma creation for patients with inoperable malignant colonic obstructions? Results of a study and cost-effectiveness analysis. Surg Endosc. 2004, Vol.18, pp.421-426.
      41. Ye GY., Cui Z., Chen L., Zhong M. Colonic stenting vs emergent surgery for acute left-sided malignant colonic obstruction: a systematic review and meta-analysis. World J Gastroenterol, 2012, Vol.18, pp.5608-5615
      42. Yoon J., Kwon SH., Lee C-K. Radiologic Placement of Uncovered Stents for the Treatment of Malignant Colonic Obstruction Proximal to the Descending Colon. Cardiovasc Intervent Radiol
      43. Yoon JY., Jung YS., Hong SP., et al. Outcomes of secondary stent-in-stent self-expandable metal stent insertion for malignant colorectal obstruction. Gastrointest Endosc, 2011, Vol.74, рр.625-633.
      44. Yoon JY., Park SJ., Hong SP., et al. Outcomes of secondary self-expandable metal stents versus surgery after delayed ini-tial palliative stent failure in malignant colorectal obstruction. Digestion, 2013, Vol.88, рр.46-55.
      45. Young CJ., De-loyde KJ., Young JM., et al. Improving Quality of Life for People with Incurable Large-Bowel Obstruction: Randomized Control Trial of Colonic Stent Insertion. Dis Colon Rectum, 2015, Vol.58, pp.838-849.
      46. Zhang Y., Shi J., Shi B., et al. Self-expanding metallic stent as a bridge to surgery versus emergency surgery for obstructive colorectal cancer: a meta-analysis. Surg Endosc, 2012, Vol.26, pp.110-119.
      47. Zhao XD., Cai BB., Cao RS., et al. Palliative treatment for incurable malignant colorectal obstructions: a meta-analysis. World J Gastroenterol, 2013, Vol.19, pp. 5565-5574.
     


    Full text is published :
    Vodoleev A.S., Duvanskiy V.A. COLORECTAL STENTING FOR THE TREATMENT OF MALIGNANT COLONIC OBSTRUCTION. Experimental and Clinical Gastroenterology Journal. 2017;139(03):60-66
    Read & Download full text

    1. Federal state budget military educational institution of higher professional education “Military medical Academy named after S. M. Kirov” Ministry of defense of the Russian Federation (St. Petersburg, Russian Federation)

    Keywords:Clostridium difficile infection,epidemiology,treatment,prevention

    Abstract:The review presents recent data on the prevalence and types of Clostridium difficile infection, about the circulation of the microorganism in the environment and the human population, the peculiarities of interaction with the normal microflora of the gastrointestinal tract that will allow for a fresh look at the epidemiology of diseases caused by this microorganism, to enhance the effectiveness of preventive interventions, to improve treatment outcomes.

      1. Magill S.S., Edwards J. R., Bamberg W., Beldavs Z. G., Dumyati G., Kainer MA. et al. Multistate point-prevalence survey of health care-associated infections. N Engl J Med 2014; 370: 1198-208.
      2. Rupnik M., Wilcox M. H., Gerding D. N. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nat Rev 2009; 7: 526-36.
      3. Gerding D.N., Lessa F. C. The epidemiology of Clostridium difficile infection inside and outside health care institutions. Infect Dis Clin N Am 2015; 29: 37-50.
      4. O’Brien J.A., Lahue B. J., Caro J. J., Davidson D. M. The emerging infectious challenge of Clostridium difficile-associated disease in Massachusetts hospitals: clinical and economic consequences. Infect Control Hosp Epidemiol 2007; 28: 1219-1227.
      5. Kuijper E.J., Coignard B., Tull P. Emergence of Clostridium difficile-associated disease in North America and Europe. Clin Microbiol Infect 2006; 12(Suppl. 6): 2-18.
      6. Bouza E. Consequences of Clostridium difficile infection: understanding the healthcare burden. Clin Microbiol Infect 2012; 18(Suppl. 6): 5-12.
      7. Desai, K., Gupta, S.B., Dubberke, E.R., Prabhu, V.S., Browne, C., Mast, T.C. Epidemiological and economic burden of Clostridium difficile in the United States: estimates from a modeling approach. BMC Infect. Dis. 2016; 16: 303.
      8. See I., Mu Y., Cohen J. et al. NAP1 strain type predicts outcomes from Clostridium difficile infection. Clin Infect Dis 2014; 58: 1394-1400.
      9. Elliott B., Androga G. O., Knight D. R., Riley T. V. Clostridium difficile infection: Evolution, phylogeny and molecular epidemiology. Infect Genet Evol. 2016 Dec 22; 49: 1-11.
      10. McDonald L.C., Coignard B., Dubberke E., Song X., Horan T., Kutty PK. Recommendations for surveillance of Clostridium difficile-associated disease. Infect Control Hosp Epidemiol 2007; 28: 140-145.
      11. van Beurden Y. H., Bomers M. K., van der Werff S. D., Pompe E. A.P.M., Spiering S., Vandenbroucke-Grauls C.M.J.E., Mulde C. J.J. Cost analysis of an outbreak of Clostridium difficile infection ribotype 027 in a Dutch tertiary care centre. Journal of Hospital Infection xxx (2017) 1-5. http://dx.doi.org/10.1016/j.jhin.2016.12.019.
      12. Cohen S.H., Gerding D. N., Johnson S., Kelly C. P., Loo V. G., McDonald L.C., et al. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America (IDSA). Infect Control Hosp Epidemiol 2010; 31: 431-55.
      13. Butler M., Bliss D., Drekonja D., Filice G., Rector T., MacDonald R., et al. Effectiveness of early diagnosis, prevention, and treatment of Clostridium difficile infection. Rockville (MD): Agency for Healthcare Research and Quality, US Department of Health and Human Services; 2011.
      14. Centers for Disease Control and Prevention (CDC). Vital signs: preventing Clostridium difficile infections. MMWR Morb Mortal Wkly Rep 2012; 61: 157-62.
      15. Domeniconi G., Serafino S., De Amicis M. M., Formica S., Lanzoni M., Maraschini A., Sisto F., Consonni D., Cappellini M. D., Fabio G., Castaldi S. Clostridium difficile infection epidemiology and management: Comparison of results of a prospective study with a retrospective one in a reference teaching and research hospital in Northern Italy. Am. J. Infect. Control. 2016; 44: 1214-8.
      16. Khanna S., Pardi D. S., Aronson S. L., Kammer P. P., Orenstein R., Sauver J. L. St., Harmsen W. S., Zinsmeister A. R. The Epidemiology of Community-acquired Clostridium difficile infection: A population-based study. Am J Gastroenterol. 2012 Jan; 107(1): 89-95.
      17. McDonald L.C., Owings M., Jernigan, D. B. Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996-2003. Emerg. Infect. Dis. 2006; 12: 409-415.
      18. Worth L.J., Spelman T., Bull A. L., Brett J. A., Richards M. J. Epidemiology of Clostridium difficile infections in Australia: enhanced surveillance to evaluate time trends and severity of illness in Victoria, 2010-2014. J Hosp Infect. 2016 Jul;93(3):280-5.
      19. Furuya-Kanamori L., McKenzie S.J., Yakob L., Clark J., Paterson D. L., Riley T. V., Clements A. C. Clostridium difficile infection seasonality: patterns across hemispheres and continents - a systematic review. PLoS One 2015; 10: e0120730.
      20. Songer J.G., Uzal F. A. Clostridial enteric infections in pigs. J. Vet. Diagn. Investig. 2005; 17; 528-536.
      21. Båverud V., Gustafsson A., Franklin A., Aspan A., Gunnarsson A. Clostridium difficile: prevalence in horses and environment, and antimicrobial susceptibility. Equine Vet. J. 2003; 35; 465-471.
      22. Weese J. S. Clostridium difficile in food - innocent bystander or serious threat? Clin. Microbiol. Infect. 2010; 16; 3-10.
      23. Rodriguez C., Avesani V., Van Broeck J., Taminiau B., Delmée M., Daube G. Presence of Clostridium difficile in pigs and cattle intestinal contents and carcass contamination at the slaughterhouse in Belgium. Int J Food Microbiol. 2013 Sep 2; 166(2): 256-62.
      24. Rodriguez C., Taminiau B., Avesani V., Van Broeck J., Delmée M., Daube G. Multilocus sequence typing analysis and antibiotic resistance of Clostridium difficile strains isolated from retail meat and humans in Belgium. Food Microbiology. 2014; 42; 166-171.
      25. Rodriguez-Palacios A., Reid-Smith R.J., Staempfli H. R., Daignault D., Janecko N., Avery B. P., Martin H., Thomspon A. D., McDonald L.C., Limbago B., Weese J. S. Possible seasonality of Clostridium difficile in retail meat, Canada. Emerg Infect Dis. 2009 May; 15(5): 802-5.
      26. Weese J.S., Avery B. P., Rousseau J., Reid-Smith R. J. Detection and enumeration of Clostridium difficile spores in retail beef and pork. Appl Environ Microbiol. 2009 Aug; 75(15): 5009-11.
      27. Bauer M.P., Notermans D. W., van Benthem, B.H., Brazier, J.S., Wilcox, M.H., Rupnik, M., Monnet, D.L., van Dissel, J.T., Kuijper, E. J. Clostridium difficile infection in Europe: a hospital-based survey. Lancet 2011; 377; 63-73.
      28. Rodriguez-Palacios A., Ilic S., LeJeune J. T. Clostridium difficile with Moxifloxacin/Clindamycin Resistance in Vegetables in Ohio, USA, and Prevalence Meta-Analysis. Journal of Pathogens. 2014, Article ID 158601, 7 p.
      29. Sjörberg M., Eriksson M., Andersson J., Noren T. Transmission of Clostridium difficile spores in isolation room environments and through hospital beds. APMIS. 2014 Sep;122(9):800-3.
      30. Alqumber M. A. Clostridium difficile in retail baskets, trolleys, conveyor belts, and plastic bags in Saudi Arabia, Saudi Med. J. 2014; 35: 1274-1277.
      31. del Mar Gamboa M., Rodriguez E., Vargas P. Diversity of mesophilic clostridia in Costa Rica soils, Anaerobe. 2005; 11: 322-326.
      32. Zidaric V., Beigot S., Lapajne S., Rupnik M. The occurrence and high diversity of Clostridium difficile genotypes in rivers, Anaerobe. 2010; 16: 371-375.
      33. Rodriguez C., Van Broeck J., Taminiau B., Delmée M., Daube G. Clostridium difficile infection: Early history, diagnosis and molecular strain typing methods. Microb Pathog. 2016 Aug; 97: 59-78. doi: 10.1016/j.micpath.2016.05.018.
      34. Yutin N., Galperin M. Y. A genomic update on clostridial phylogeny: Gramnegative spore formers and other misplaced clostridia. Environ. Microbiol. 2013; 15; 2631-2641.
     


    Full text is published :
    Zakharenko S.M. EPIDEMIOLOGICAL ASPECTS OF C. DIFFICILE INFECTION. Experimental and Clinical Gastroenterology Journal. 2017;139(03):67-70
    Read & Download full text

    1. Central state medical Academy (Moscow, Russian Federation)

    Keywords:bacterial overgrowth syndrome,inflammatory bowel disease

    Abstract:Review article devoted to the problem of small intestinal bacterial overgrowth syndrome (SIBO). This definition of the syndrome, epidemiology, clinical manifestations, diagnosis. Discussed the pathogenesis and the relationship SIBO with diseases of the gastrointestinal tract. The new studies SIBO in patients with inflammatory bowel disease.

      1. Ардатская М. Д. Синдром избыточного бактериального роста и нарушение процессов пищеварения и всасывания: патогенетическая нутриционная терапия//. Экспериментальная и клиническая гастроэнтерология. 2009; 6; С. 86-96.
      2. Ардатская М. Д., Минушкин О. Н. Синдром избыточного бактериального роста: определение, современные подходы к диагностике и лечебной коррекции.// Гастроэнтерология. 2012; 2: С. 45-49
      3. Ардатская М. Д. Синдром избыточного бактериального роста в тонкой кишке, современные методы диагностики и подходы к лечебной коррекции. //Медицинский совет 2016, 14: 88-95.
      4. Белоусова Е. А. Синдром избыточного бактериального роста тонкой кишки в свете общей концепции о дисбактериозе кишечника. Взгляд на проблему. //Фарматека, 2009, № 2.
      5. Логинов В.А. кандид. Дисс. Синдром избыточного бактериального роста у больных со сниженной кислотопродуцирующей функции желудка (клиническое значение, диагностика и лечение). М. 2015.
      6. Almeida J. A., Kim R., Stoita A., McIver C.J., Kurtovic J., Riordan S. M. Lactose malabsorption in the elderly: role of small intestinal bacterial overgrowth. Scand. J. Gastroenterol. 2008;43(2): 146-54.
      7. Bures J, Cyrany J, Kohoutova D, Fo¨ rstl M, Rejchrt S, Kvetina J, et al. Small intestinal bacterial overgrowth syndrome. World J Gastroenterol WJG 2010;16:2978-90.
      8. Carrara M., Desideri S., Azzurro M., Bulighin G. M. et al. Small intestine bacterial overgrowth in patients with irritablebowel syndrome. Eur. Rev. Med. Pharmacol. Sci. 2008 May-Jun;12(3):197-202.
      9. Castiglione F, Del Vecchio Blanco G, Rispo A, et al. Orocecal transit time and bacterial overgrowth in patients with Crohn’s disease. J Clin Gastroenterol. 2000;31:63-66.
      10. Chandra S, Dutta U, Noor MT, Taneja N, Kochhar R, Sharma M, et al. Endoscopic jejunal biopsy culture: a simple and effective method to study jejunal microflora. Indian J Gastroenterol 2010;29:226-30.
      11. Chang MS, Green PHR. A review of rifaximin and bacterial overgrowth in poorly responsive celiac disease. Ther Adv Gastroenterol 2012;5:31-6.
      12. Chang MS, Minaya MT, Cheng J, Connor BA, Lewis SK, Green PHR. Double-blind randomized controlled trial of rifaximin for persistent symptoms in patients with celiac disease. Dig Dis Sci 2011;56:2939-46.
      13. Choung RS, Ruff KC, Malhotra A, Herrick L, Locke GR, Harmsen WS, et al. Clinical predictors of small intestinal bacterial overgrowth by duodenal aspirate culture. Aliment Pharmacol Ther 2011;33:1059-67.
      14. Collins BS, Lin HC. Chronic abdominal pain in children is associated with high prevalence of abnormal microbial fermentation. Dig Dis Sci 2010;55: 124-30.
      15. Collins BS, Lin HC. Double-blind, placebo-controlled antibiotic treatment study of small intestinal bacterial overgrowth in children with chronic abdominal pain. J Pediatr Gastroenterol Nutr 2011;52:382-6.
      16. Compare D, Pica L, Rocco A, De Giorgi F, Cuomo R, Sarnelli G, et al. Effects of long-term PPI treatment on producing bowel symptoms and SIBO. Eur J Clin Invest 2011;41:380-6.)
      17. Daveson AJM, Anderson RP. Small bowel endoscopy and coeliac disease. Best Pract Res Clin Gastroenterol 2012;26:315-23.
      18. Dominguez-Munoz J. E. Pancreatic enzyme therapy for pancreatic exocrine insufficiency. Curr. Gastroenterol Rep. 2007
      19. Fairris GM, Ashworth J, Cotterill JA. A dermatosis associated with bacterial overgrowth in jejunal diverticula. Br J Dermatol 1985;112:709-13.
      20. Fundaro` C, Fantacci C, Ansuini V, Giorgio V, Filoni S, Barbaro F, et al. Fecal calprotectin concentration in children affected by SIBO. Eur Rev Med Pharmacol Sci 2011;15:1328-35.
      21. George N.S,. Sankineni A., Parkman H.P. Small Intestinal Bacterial Overgrowth in Gastroparesis Dig Dis Sci. 2012. Oct 5.
      22. Ghoshal UC. How to interpret hydrogen breath tests. J Neurogastroenterol Motil 2011;17:312-7.
      23. Ghoshal UC, Kumar S, Mehrotra M, Lakshmi C, Misra A. Frequency of small intestinal bacterial overgrowth in patients with irritable bowel syndrome and chronic non-specific diarrhea. J Neurogastroenterol Motil 2010;16:40-6.
      24. Gupta A, Dhiman RK, Kumari S, Rana S, Agarwal R, Duseja A, et al. Role of small intestinal bacterial overgrowth and delayed gastrointestinal transit time in cirrhotic patients with minimal hepatic encephalopathy. J Hepatol 2010;53:849-55.
      25. Hutyra T, Iwan´ czak B. Bakteryjny przerost flory jelita cienkiego u dzieci Small bowel bacterial overgrowth in children Pediatr Wspo´ łczesna Gastroenterol Hepatol Z˙yw Dziecka 2010;12:130-4.
      26. Jouet P, Coffin B, Sabate J-M. Small intestinal bacterial overgrowth in patients with morbid obesity. Dig Dis Sci 2011;56:615. author reply 615-616.
      27. Jun DW, Kim KT, Lee OY, Chae JD, Son BK, Kim SH, et al. Association between small intestinal bacterial overgrowth and peripheral bacterial DNA in cirrhotic patients. Dig Dis Sci 2010;55:1465-71.
      28. Kim K-M, Kim B-T, Lee D-J, Park S-B, Joo N-S, Kim Y-S, et al. Erosive esophagitis may be related to small intestinal bacterial overgrowth. Scand J Gastroenterol 2012;47:493-8.
      29. Lappinga PJ, Abraham SC, Murray JA, Vetter EA, Patel R, Wu T-T. Small intestinal bacterial overgrowth: histopathologic features and clinical correlates in an underrecognized entity. Arch Pathol Lab Med 2010; 134: 264-70.
      30. Lakshmi CP, Ghoshal UC, Kumar S, Goel A, Misra A, Mohindra S, et al. Frequency and factors associated with small intestinal bacterial overgrowth in patients with cirrhosis of the liver and extra hepatic portal venous obstruction. Dig Dis Sci 2010;55:1142-8.
      31. Leiby A, Mehta D, Gopalareddy V, Jackson-Walker S, Horvath K. Bacterial overgrowth and methane production in children with encopresis. J Pediatr 2010;156:766-70. 770.e1.
      32. Leung Ki EL, Roduit J, Delarive J, Guyot J, Michetti P, Dorta G. Small intestine bacterial overgrowth. Rev Me´ dicale Suisse 2010;6:186-8. 190-1.
      33. Łokiec´ K, Klupin´ska G, Walecka-Kapica E, Błon´ ska A. Estimation of small intestinal bacterial overgrowth in patients with constipation and diarrhea irritable bowel syndrome. Pol Merkur Lek Organ Pol Tow Lek 2014;36: 307-10.
      34. Lin H. C. Small intestinal bacterial overgrowth: aframework for understanding irritable bowel syndrome.JAMA. 2004. Aug 18; 292(7): 852-8.
      35. Lisowska A, Pogorzelski A, Oracz G, Skorupa W, Cofta S, Socha J, et al. Small intestine bacterial overgrowth and fat digestion and absorption in cystic fibrosis patients. Acta Sci Pol Technol Aliment 2010;9:477-83.
      36. Lisowska A, Wo´ jtowicz J, Walkowiak J. Small intestine bacterial overgrowth is frequent in cystic fibrosis: combined hydrogen and methane measurements are required for its detection. Acta Biochim Pol 2009;56:631-4.
      37. Lisowska A, Pogorzelski A, Oracz G, Siuda K, Skorupa W, Rachel M, et al. Oral antibiotic therapy improves fat absorption in cystic fibrosis patients with small intestine bacterial overgrowth. J Cyst Fibros 2011;10:418-21.
      38. Lombardo L., Foti M., Ruggia O., Chiecchio A. Increased incidence of small intestinal bacterial overgrowth during proton pump inhibitor therapy Clin. Gastroenterol. Hepatol. 2010.Jun;8(6): 504-8.
      39. Madrid AM, Poniachik J, Quera R, Defilippi C. Small intestinal clustered contractions and bacterial overgrowth: a frequent finding in obese patients. Dig Dis Sci 2011;56:155-60.
      40. Miazga A. et al. / Advances in Medical Sciences 60 (2015) 118-124
      41. Mitchell DM, Henao MP, Finkelstein JS, Burnett-Bowie S-AM. Prevalence and predictors of vitamin D deficiency in healthy adults. Endocr Pract 2012;18: 914-23.
      42. Ojetti V, Pitocco D, Scarpellini E, Zaccardi F, Scaldaferri F, Gigante G, et al. Small bowel bacterial overgrowth and type 1 diabetes. Eur Rev Med Pharmacol Sci 2009;13:419-23.
      43. Pande C., Kumar A., Sarin S. K. Small-intestinal bacterial overgrowth in cirrhosis is related to the severity of liver diseaseAliment Pharmacol Ther. 2009. Jun 15; 29(12): 1273-81.
      44. Park JS, Yu JH, Lim HC, Kim JH, Yoon YH, Park HJ, et al. Usefulness of lactulose breath test for the prediction of small intestinal bacterial overgrowth in irritable bowel syndrome. Korean J Gastroenterol Taehan Sohwagi Hakhoe Chi 2010;56:242-8.
      45. Parodi A, Sessarego M, Greco A, Bazzica M, Filaci G, Setti M, et al. Small intestinal bacterial overgrowth in patients suffering from scleroderma: clinical effectiveness of its eradication. Am J Gastroenterol 2008;103:1257-62.
      46. Pimentel M. Evaluating a bacterial hypothesis in IBS using a modification of Koch’s postulates: part 1. Am J Gastroenterol 2010;105:718-21.
      47. Pimentel M, Mayer AG, Park S, Chow EJ, Hasan A, Kong Y. Methane production during lactulose breath test is associated with gastrointestinal disease presentation. Dig Dis Sci. 2003;48:86-92.
      48. Pyleris E, Giamarellos-Bourboulis EJ, Tzivras D, Koussoulas V, Barbatzas C, Pimentel M. The prevalence of overgrowth by aerobic bacteria in the small intestine by small bowel culture: relationship with irritable bowel syndrome. Dig Dis Sci 2012;57:1321-9.
      49. Rana SV, Sharma S, Kaur J, Sinha SK, Singh K. Comparison of lactulose and glucose breath test for diagnosis of small intestinal bacterial overgrowth in patients with irritable bowel syndrome. Digestion 2012;85:243-7.
      50. Rana S, Bhansali A, Bhadada S, Sharma S, Kaur J, Singh K. Orocecal transit time and small intestinal bacterial overgrowth in type 2 diabetes patients from North India. Diabetes Technol Ther 2011;13:1115-20.
      51. Rana SV, et al. Small Intestinal Bacterial Overgrowth and Orocecal Transit Time in Patients of Inflammatory Bowel DiseaseDig Dis Sci (2013) 58:2594-2598).
      52. Rana SV, et al. Relationship of cytokines, oxidative stress and GI motility with bacterial overgrowth in ulcerative colitis patients, J Crohns Colitis (2014).
      53. Ratuapli SK, Ellington TG, O’Neill M-T, Umar SB, Harris LA, Foxx-Orenstein AE, et al. Proton pump inhibitor therapy use does not predispose to small intestinal bacterial overgrowth. Am J Gastroenterol 2012;107:730-5.
      54. Romond M-B, Colavizza M, Romond PC, Bezirtzoglou E. Selective decontamination by antimicrobials during long term treatment: perspectives for saving host indigenous microbiota. Anaerobe 2011;17:468-73.
      55. Sachdeva S, Rawat AK, Reddy RS, Puri AS. Small intestinal bacterial overgrowth (SIBO) in irritable bowel syndrome: frequency and predictors. J Gastroenterol Hepatol 2011;26(Suppl. 3):135-8.
      56. Sachdev AH, Pimentel M. Gastrointestinal bacterial overgrowth: pathogenesis and clinical significance. Ther Adv Chronic Dis 2013;4:223-31.
      57. Schommer K, Reljic D, Ba¨rtsch P, Sauer P. Gastrointestinal complaints in runners are not due to small intestinal bacterial overgrowth. J Negat Results Biomed 2011;10:8.
      58. Singh, V. V., Toskes, P. P. Small Bowel Bacterial Overgrowth: Presentation, Diagnosis, and Treatment // Curr Treat Options Gastroenterol. 2004; 7 (1): 19-28.
      59. Shah ED, Basseri RJ, Chong K, Pimentel M. Abnormal breath testing in IBS: a meta-analysis. Dig Dis Sci 2010;55:2441-9.
      60. Spiegel BMR. Questioning the bacterial overgrowth hypothesis of irritable bowel syndrome: an epidemiologic and evolutionary perspective. Clin Gastro-enterol Hepatol 2011;9:461-9. quiz e59.
      61. Tarnopolsky MA, Pearce E, Matteliano A, James C, Armstrong D. Bacterial overgrowth syndrome in myotonic muscular dystrophy is potentially treatable. Muscle Nerve 2010;42:853-5.
      62. Shanab AA, Scully P, Crosbie O, Buckley M, O’Mahony L, Shanahan F, et al. Small intestinal bacterial overgrowth in nonalcoholic steatohepatitis: association with toll-like receptor 4 expression and plasma levels of interleukin 8. Dig Dis Sci 2011;56:1524-34.
      63. Weinstock LB, Walters AS. Restless legs syndrome is associated with irritable bowel syndrome and small intestinal bacterial overgrowth. Sleep Med 2011;12:610-3.
      64. Wilder-Smith CH, Materna A, Wermelinger C, Schuler J. Fructose and lactose intolerance and malabsorption testing: the relationship with symptoms in functional gastrointestinal disorders. Aliment Pharmacol Ther2013;37:1074-83.
      65. Yakoob J, Abbas Z, Khan R, Hamid S, Awan S, Jafri W. Small intestinal bacterial overgrowth and lactose intolerance contribute to irritable bowel syndrome symptomatology in Pakistan. Saudi J Gastroenterol 2011;17:371-5.
      66. Zhao J, Fox M, Cong Y, Chu H, Shang Y, Fried M, et al. Lactose intolerance in patients with chronic functional diarrhoea: the role of small intestinal bacterial overgrowth. Aliment Pharmacol Ther 2010;31:892-900.
      67. Zoetendal EG, Raes J, van den Bogert B, Arumugam M, Booijink CCGM, Troost FJ, et al. The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates. ISME J 2012;6:1415-26.
     


    Full text is published :
    Topchy T.V., Minushkin O.N., Skibina Y.S,, Evsikov A.E. THE BACTERIAL OVERGROWTH SYNDROME IN CLINICAL PRACTICE. Experimental and Clinical Gastroenterology Journal. 2017;139(03):71-78
    Read & Download full text

    1. Russian Medical Academy of Continuous Professional Education (Moscow, Russian Federation)

    Keywords:gut microbiota,obesity,metabolics diseases

    Abstract:The review deals modern ideas about mechanisms of obesity development due to disorders of gut microbiota, prospects of using probiotics and prebiotics in treatment of metabolics diseases.

      1. Андрианова О. Л., Мирсаева Г. Х., Ибрагимова Л. А. Эффективная стратегия ожирения// Эндокринология.-2015.-№ 3.-С.51-56.
      2. Гарднер Д., Шобек Д. Базисная и клиническая эндокринология. - 2011.-Кн.1, М.: Бином.
      3. Григорьев А. В. Желудочно-кишечный тракт как среда обитания микробов. Раздел 1. Морфология желудочно-кишечного бактериального биотопа. - 2004.-М.: СИЛМА. - 123 с.
      4. Лоранская И. Д., Болдырева М. Н., Лаврентьева О. А., Мулухова Э. В. Пристеночная микрофлора кишечника. М.: Прима Принт.-2015.-100 с.
      5. Нормы физиологических потребностей в энергии и пищевых веществах для различных групп населения Российской Федерации. Методические рекомендации МР 2.3.1.2432-08. М.: Федеральная служба по надзору в сфере защиты прав потребителя и благополучия человека.-2008.
      6. Шварц В. Жировая ткань как эндокринный орган.// Проблемы эндокринологии. - 2009. - Т. 55, № 1. С. 38-44.
      7. Aguirre M., Venema K. The use of fecal samples for studying human obesity.//Europ. J. of Epidemiology.-2015. - 30.-P.1067-1069.
      8. Al Mushref M. and Srinivasan S. Effect of high fat-diet and obesity on gastrointestinal motility.//Annals of Translational Medicine.2013,1, 14.
      9. Backhed F., Manchester J. K., Semenkovich C. F., Gordon J. I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. // Proc. Natl. Acad. Sci. USA. - 2007. - V. 104. - P. 979-984.
      10. Blaser M. J. Missing Microbes. New York: Henry Holt and Company, LLC. 2014. 274 pp.
      11. Caesar R., Fak F., Backhed F. Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism.//J Intern Med 2010. - 268 (4).-P.320-328.
      12. Canfora E. E., Jocken J. W., Black E. E. Short-chain fatty acids in control of body weight and insulin sensitivity.//Nat Rev Endocrinol.-2015,11. - P. 297-313.
      13. Chambers E. S. et. al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults.//Gut,2015, 64. - P. 1744-1754.
      14. Collado M. C., Isolauri E., Laitinen K. and Salminen S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women.//Am J Clin Nutrition.-2008,88. - P. 894-899.
      15. Cox L. M., Blaser M. J. Pathways in microbe-induced obesity.//Cell Metabolism.-2013. - 17(6). - P. 883-894.
      16. David L. A., Maurice C. F., Carmody R. N. et. al. Diet rapidly and reproducibly alters the human gut microbiome.//Nature.-2014.-505. - P. 559-563.
      17. De Filippo C., Cavalieri D., Di Paola M. et. al. Impact of diet shaping gut microbiota revealed by а comparative study in children from Europe and rural Africa.//Proc Natl Acad Sci.-2010.-107.-P.14691-14696.
      18. de Wit N., Derrien M., Bosch-Vermeulen H. et. al. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine.// Am J.of Phys Gastrointestinal Liver Phy.2012. - 303(5). - G589-G599.
      19. Degen D. L. and Phillips S. F. Variability of gastrointestinal transit in healthy women and men.//Gut.1996.-39. - P. 299-305.
      20. DeWeerdt S. Microbiome: A complicated relation-ship status.//Nature.-2014.-508. - S 61-63.
      21. Dowsey M. M., Choong P. F. M. Obese diabetic patients are at substantial risk for deep infection after primary TKA. // Clin Orthop Relat Res. - 2009. - V. 467(6). - P. 1577-1581.
      22. Fried M., Hainer V., Basdevant A. et. al. Interdisciplinary European guidelines on surgery of severe obesity.//Obes. Facts.-2008. - Vol. 1. - P. 52-59.
      23. Furet J. P., Kong L. C., Tap J. et. al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers.//Diabetes. - 2010. - 59. - P. 3049-3057.
      24. Gaci N., Borrel G., Tottey W. et. al. Archea and the human gut: new beginning of an old story.//World J. of Gastroenterology.2014.-20.-P.16062-16078.
      25. Gasbarrini A. The gut microbiota. The 4 organ of the digestive system.Composition, functions and related diseases. Verduci Editore.-2013. - 74 p.
      26. Karlsson С. L., Onnerfalt J., Xu J. et. al. The microbiota of the gut in pre-school children with normal and excessive body weight.//Obesity.2012, 20. - P. 2257-2261.
      27. Keenan M. J. et. al. Role of resistant starch in improving gut health, adiposity and insulin resistance.//Adv Nutr 2015. - 6.-P.198-205.
      28. Khan M. Y. et al. A review on obesity and its management.// Int. Journal of Scientific & Engineering Research- 2012. - Volume 3, Issue 11.
      29. Krznarić Z, Vranešić Bender D, Kunović A, Kekez D, Stimac D. Gut Microbiota and Obesity.// Dig Dis. - 2012. - 30(2). - Р.196-200.
      30. Le Chatelier E., Nielsen T., Qin J. et. al. Richness of human gut microbiome correlates with metabolic markers.//Nature. - 2014. - 500. - P. 541-546.
      31. Ley R. E., Backhed F., Turnbaugh P., Lozupone C. A., Knight R. D. and Gordon J. I. Obesity alters gut microbial ecology.// Proc. Natl. Acad. Sci.USA.-2005.-V. 102. - P. 11070-11075.
      32. Ley R. E., Turnbaugh P. J., Klein S. and Gordon J. I. Microbial ecology: human gut microbes associated with obesity.//Nature.-2006.-444. - P. 1022-1023.
      33. Lin H. V. et. al. Butirate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor3 - independent mechanisms. //PLoS One 7.-2012, e35240.
      34. Lu Y., Fan C., Li P. et. al. Short chain fatty acids prevent high-fat-diet-induced obesity in mice by regulating G protein-coupled receptors and gut microbiota.//Nature, 2016.-6. - P. 1-13.
      35. Marteau P. The gut microbiota in 20 questions. John Libbey Eurotext.-2014. - 42p.
      36. Nam Y.-D., Jung M.-J., Roh S. W. et. al. Comparative analysis of Korean human gut microbiota by barcoded pyrosequencing. 2011. - PloS One 6. - e22109.
      37. Roberts K. T., Allen-Vercoe E., Williams S. A. et. al. Comparative study of the in vitro fermentative characteristics of fenugreek gum, white bread and bread with fenugreek gum using human faecal microbes.// Bioactive Carbohydrates and Dietary Fibre. 2015,5. - P. 116-124.
      38. Scarpellini E., Tack J. Obesity and metabolic syndrome: an inflammatory condition.//Dig Dis 2012. - 30 (2). - P. 148-153.
      39. Tan J. et. al. The role of short-chain fatty acids in health and disease.//Adv Immunol.-2014.-121, P. 91-119.
      40. Torralladona D., Harris C. I. and Fuller M. F. Pigs gastrointestinal microflora provide them with essential amino acids.//J. of Nutrition. 2003.-133.-P.1127-1131.
      41. Tremalori V., Backhed F. Functional interactions between the gut microbiota and host metabolism.//Nature.-2012. - 489(7415). - P. 242-249.
      42. Walker A. W. et. al. pH and peptide supply can radically alter bacterial population and short-chain fatty acids ratios within microbial communities from the human colon.//Appl Environ Microbiol.-2005,71. - P. 3692-3700.
      43. Zhang M., Yang X.-J. Effects of a high fat diet on intestinal microbiota and gastrointestinal diseases.// World J. of Gastroenterology. 2016. - 22(40). - P. 8905-8909.
     


    Full text is published :
    Loranskaya I.D., Stepanova E.V., Kupaeva V.A. GUT MICROBIOTA AND OBESITY. Experimental and Clinical Gastroenterology Journal. 2017;139(03):79-83
    Read & Download full text

    1. “Moscow state medical dental University named A. I. Evdokimov” Ministry of health of the Russian Federation (Moscow, Russian Federation)
    2. “Central clinical hospital № 2 of them. N.. Semashko” JSC “Russian Railways” (Moscow, Russian Federation)

    Keywords:functional diarrhea,Rome criteria IV,diagnosis

    Abstract:The achievements of basic and clinical science over the past 10 years in the study of epidemiology, etiology, pathophysiology, diagnosis and therapy of functional disorders of the intestine necessitated the revision existed since 2006, the Rome III criteria. In may 2016 the global gastroenterology community on the American gastroenterological week met with Roman criteria IV, the main provisions of the consensus are presented in this publication. Functional bowel disease continues to include clinical disease as irritable bowel syndrome, functional constipation, functional diarrhea, functional abdominal bloating, and nonspecific functional bowel disorders. Unlike the previous classification in the section of functional bowel diseases has a new form - opioid-induced constipation, designed to draw attention of clinicians on the sharply increased frequency of use of opiates and associated with a large number of side effects in their application. Roman urges the Committee not to consider this form as a separate disease, and to classify the developed clinical picture as opioid-induced adverse effects. Functional diarrhea (FD) is characterized by the persistence loose or watery stools, an important distinguishing criterion for diagnosis is not a criteria of IBS, i. e. in this disorder of stool abdominal pain or bloating certainly can be present but are never dominant symptoms. In addition to these changes, according to the study, Whitehead W. E. et al. for the diagnosis of functional diarrhea is the criterion of the frequency of episodes of liquid stools or watery stools was reduced from 75 % to 25 %

      1. Маев И. В., Черемушкин С. В. Синдром раздраженного кишечника. Римские критерии III. Гастроэнтерология. Приложение к журналу Consilium Medicum. 2007. № 1. С. 29-33.
      2. Brian E. Lacy, Fermín Mearin, Lin Chang, William D. Chey, Anthony J. Lembo, Magnus Simren, and Robin Spiller. Bowel Disorders. Gastroenterology 2016;150:1393-1407.
      3. Маев И. В., Черемушкин С. В., Кучерявый Ю. А., Черемушкина Н. В. Синдром раздраженного кишечника. Римские критерии IV. Consilium medicum, № 8, vol. 18, С. 79-85, 2016.
      4. Whitehead WE, Palsson OS. Report on Rome Normative GI Symptom Survey. 2014.
      5. Porter CK, Gormley R, Tribble DR, et al. The Incidence and gastrointestinal infectious risk of functional gastrointestinal disorders in a healthy US adult population. Am J Gastroenterol 2011;106:130-138.
      6. Zhao YF, Guo XJ, Zhang ZS, et al. Epidemiology of functional diarrhea and comparison with diarrhea predominant irritable bowel syndrome: a populationbased survey in China. PLoS One 2012;7: e43749.
      7. Sorouri M, Pourhoseingholi MA, Vahedi M, et al. Functional bowel disorders in Iranian population using Rome III criteria. Saud J Gastroenterol 2010;16:154-160.
      8. Chang F-Y, Chen P-H, Wu T-C, et al. Prevalence of functional gastrointestinal disorders in Taiwan: questionnaire-based survey for adults based on the Rome III criteria. Asia Pac J Clin Nutr 2012;21:594-600.
      9. Schiller LR, Pardi DS, Sellin JH. Clin Gastroenterol Hepatol. 2017 Feb;15(2):182-193.e3. doi: 10.1016/j.cgh.2016.07.028. Review.
      10. Tack J. Functional diarrhea. Gastroenterol Clin North Am 2012;41:629-637.
      11. Choi MG, Camilleri M, O’Brien MD, et al. A pilot study of motility and tone of the left colon in patients with diarrhea due to functional disorders and dysautonomia. Am J Gastroenterol 1997;92:297-302.
      12. Zanini B, Ricci C, Bandera F, et al. Incidence of postinfectious irritable bowel syndrome and functional intestinal disorders following a water-borne viral gastroenteritis outbreak. Am J Gastroenterol 2012;107:891-899.).
      13. Parry SD, Stansfield R, Jelley D, et al. Does bacterial gastroenteritis predispose people to functional gastrointestinal disorders? A prospective, community-based, casecontrol study. Am J Gastroenterol 2003;98:1970-1975.
      14. Hobbis IC, Turpin G, Read NW. Abnormal illness behaviour and locus of control in patients with functional bowel disorders. Br J Health Psychol 2003;8:393-408.
      15. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol 1997; 32:920-924.
      16. Маев И. В., Черемушкин С. В., Кучерявый Ю. А. Синдром раздраженного кишечника. Римские критерии IV. О роли висцеральной гиперчувствительности и способах ее коррекции. Методическое пособие. М., 2016.
      17. Longstreth GF, Thompson WG, Chey WD, et al. Functional bowel disorders. Gastroenterology 2006; 130:1480-1491.
      18. Vanner SJ, Depew WT, Paterson WG, et al. Predictive value of the Rome criteria for diagnosing the irritable bowel syndrome. Am J Gastroenterol 1999;94:2912-2917.
      19. Маев И. В., Кучерявый Ю. А., Андреев Д. Н., Черёмушкин С. В. Эволюция представлений о микроскопическом колите. Терапевтический архив. 2015. Т. 87. № 4. С. 69-76.
      20. Efskind PS, Bernklev T, Vatn MH. A double-blind placebo-controlled trial with loperamide in irritable bowel syndrome. Scand J Gastroenterol 1996;31:463-468.
      21. Fernández-Bañares F, Rosinach M, Piqueras M, et al. Randomised clinical trial: colestyramine vs. hydroxypropyl cellulose in patients with functional chronic watery diarrhoea. Aliment Pharmacol Ther 2015;41:1132-1140.
     


    Full text is published :
    Cheremushkin S.V., Kucheryavy Y.A., Cheremushkina N.V., Maev I.V. FUNCTIONAL DIARRHEA. ROME CRITERIA IV. Experimental and Clinical Gastroenterology Journal. 2017;139(03):84-88
    Read & Download full text

    1. Saint-Petersburg state pediatric medical University (St. Petersburg, Russian Federation)

    Keywords:Nonceliac sensitivity to gluten,allergies to wheat,celiac disease

    Abstract:Nonceliac sensitivity to gluten (NCSG) is a new syndrome manifested by a gluten intolerance is not allergic and non-autoimmune condition in which eating gluten can cause symptoms similar to the manifestations of celiac disease. NCSG the diagnosis requires the exclusion of coeliac disease and allergies to wheat. NCSG occurs mainly by intestinal symptoms that appear soon after eating gluten-containing foods and are rapidly after discontinuation of gluten. NCSG is not accompanied by retardation in physical and sexual development, weight loss and autoimmune or allergic comorbidity. The basis for the development of this syndrome probably is the effect of microbiota and metabolites of gluten on the neuroreceptors, as well as a mild inflammation of the intestinal mucosa without atrophy. NCSG more characteristic of adults than children and often manifests at a young age. The article presents the differential diagnostic signs of NCSG, celiac disease and allergies to wheat.

      1. Shewry P. R. Wheat - J. Exp.Botanics -2009, v.60, p.1537-1553
      2. Rubio-Tapia A., Ludvigsson J. F., Brantner T. L., et al. The prevalence of celiac disease in the United States - Am. J. Gastroenterol., 2012, v.107, p.1538-1544.
      3. Czaja-Bulsa G. Non coeliac gluten sensitivity - a new disease with gluten intolerance - Clinical Nutr., 2014, http://dx.doi.org/10.1016/j.clnu.2014.08.012
      4. Catassi c., Bai J. C., Bonaz B., et al. Non-celiac gluten sensitivity^ the new frontier of gluten related disorders. - Nutrients, 2013; v.5, p.3839-3853.
      5. Sapone A., Bai J. C., Ciacci C., Dolincek J., et al. Spectrum of gluten-related disorders consensus on new nomenclature and classification - BMC Med., 2012, v.10, p.13.
      6. Ludvigsson J. F., Leffler D. A., Bai J. C., et al. The Oslo definitions for coeliac disease and related terms - Gut, 2013, v.62, p.43-52.
      7. Wahnschaffe U., Ulrich R., Riecken E. O., Schulzke J. D. Celiac disease-like abnormalities in a subgroup of patients with irritable bowel syndrome - Gastroenterology, 2001, v.121, p.1329-1338.
      8. Inomata N. Wheat allergy - Curr.Opin.Allergy Clin Immunol., 2009, v.9, p.238-243.
      9. Fasano A. Intestinal permeability and its regulation by zonulin: diagnostic and therapeutic implications - Clin. Gastroenterol.Hepatol, 2012, v.10, p.1096-1100.
      10. Mazzarella G., Stefanile R., Camarca A., et al. Gliadin activates HLA class 1-restricted CD 8+ T-cells in celiac disease intestinal mucosa and induces the enterocyte apoptosis - Gastroenterology, 2008, v.134, p.1017-1027.
      11. Tursi A. Gastrointestinal motility disturbances in celiac disease - J. Clin.Gastroenterol, 2004, v.38, p.642-645.
      12. Barrett J. S., Gibson P. R. Fermentable oligosaccharides, disaccharides, monosaccharides and poliols (FODMAPs) and nonallergic food intolerance: FODMAPs or food chemicals& - Therap.Adv.Gastroenterol., 2012, v.5, p.261-268.
      13. Leon F., Sanches L., Camarero C., Roy G. Cytocine production by intestinal intraepithelial lymphocyte subsets in celiac disease - Dig.Dis.Sci, 2005, v.50, p.593-600.
      14. Monteleone I., Serra M., Del Vecchio Blanco G., et al. Characterization of IL-17A-proucing cells in celiac disease mucosa - J. Immunol, 2010, v.184, p.2211-2218.
      15. Mansueto P., Seidita A, D’Alcamo A., et al. Non-celiac gluten sensitivity: literature review - J. Amer. College Nutr., 2014, http://www.tandfonline.com/loi/uacn20
      16. Hmida N. B., Ben Ahmed M., Moussa A., et al. Impaired control of effector T cells by regulatory T-cells: a clue to loss of oral tolerance and autoimmunity in celiac disease? - Am. J. Gastroenterol., 2012, v.107, p.604-611.
      17. Josefowicz S. Z., Lu L. F., Rudensky A. Y. Regulatory T cells mechanisms of differentiation and function - Ann.Rev.Immunol., 2012, v.30, p.531-564.
      18. Kalliomaki M., Satokari R., Lahteenoja H., et al. Expression of microbiota, toll-like receptors, and their regulators in the small intestinal mucosa in celiac disease. - J. Pediatric Gastroenterol., 2012, v.54, p.727-732.
      19. Sapone A., Lammers K. M., Casolaro V., et al. Divergence of gut permeability and mucosal immune gene expression in two gluten-associated conditions: celiac disease and gluten sensitivity - BMC Med., 2-11, v.9, p.23.
      20. Oboki K., Ohno T., Salto H., Nakae S. Th17 and allergy - Allergol.Int., 2008, v.57, p.121-134.
      21. Volta U., Tovoli F., Cicola R., ett al. Serological tests in gluten sensitivity (nonceliac gluten intolerance) - J. Clin.Gastroenterol., 2012, v.46, p.680-685.
      22. Jackson J. R., Eaton W. W., Cascella N. G., et al. Neurologic and psychiatric manifestations of celiac disease and gluten sensitivity - Psychiatr Q, v.83, p.91-102.
      23. Camilleri M., Madsen K., Spiller R., et al. Intestinal barrier function in health and gastrointestinal disease. - Neurogastroenterol. Motil., 2012, v.24, 503-512.
      24. Makela M. J., Eriksson C., Kotaniemi-Syrjanen A., et al. Wheat allergy in children - new tools for diagnostics - Clin.Experim.Allergy, 2014, v.44, p.1420-1430.
      25. Marcason W. What is the current status of research concerning use of a gluten-free, casein-free diet for children diagnosed with autism? - J. Am. Diet.Assoc., 2009, v.109(3), p.572.
      26. Lachance L. R., McKenzie K. Biomarkers of gluten sensitivity in patients with non-affective psychosis: a meta-analysis - Schisophr.Res., 2013, http:/dx.doi.org/10.1016/j.schres.2013.12.001
      27. Cascella N. G., Santora D., Gregory P., et al. Increased prevalence of transglutaminase 6 antibodies in sera from schizophrenia patients - Schizophr.Bull., 2013, v.39, p.867-871.
     


    Full text is published :
    Kornienko E.A. NONCELIAC SENSITIVITY TO GLUTEN. Experimental and Clinical Gastroenterology Journal. 2017;139(03):89-98
    Read & Download full text

    1. A. N. Ryzhikh State scientific center of coloproctology (Moscow, Russian Federation)

    Keywords:Biofeedback therapy (BFB-therapy),constipation

    Abstract:Biofeedback therapy (BFB-therapy) is one of the main treatment modalities for constipation caused by functional defecation disorder. This review contains and update on effectiveness and methods of BFB-therapy. Described are inclusion criteria for BFB-therapy based on Rome IV consensus as well as standard treatment protocol developed by US and European Societies of neurogastroenterology and motility. Perspectives for future research in BFB-therapy for chronic constipation are proposed

      1. Sanchez MIP, Bercik P. Epidemiology and burden of chronic constipation. Can J Gastroenterol. 2011;25 Suppl B(Suppl B):11B-15B.
      2. Rao SSC, Seaton K, Miller MJ, et al. Psychological profiles and quality of life differ between patients with dyssynergia and those with slow transit constipation. J Psychosom Res. 2007;63(4):441-449.
      3. Hamid SS, Malfertheiner P, et al. World Gastroenterology Organisation Global Guideline: Constipation. J Clin Gastroenterol. 2011;45(6):483-487.
      4. Rao SSC, Bharucha AE, Chiarioni G, et al. Anorectal Disorders. Gastroenterology. 2016;150(6):1430-1442.e4.
      5. Nullens S, Nelsen T, Camilleri M, et al. Regional colon transit in patients with dyssynergic defaecation or slow transit in patients with constipation. Gut. 2012;61(8):1132-1139.
      6. Rao SS, Patel RS. How useful are manometric tests of anorectal function in the management of defecation disorders? Am J Gastroenterol. 1997;92(3):469-475.
      7. Rao SSC, Mudipalli RS, Stessman M, Zimmerman B. Investigation of the utility of colorectal function tests and Rome II criteria in dyssynergic defecation (Anismus). Neurogastroenterol Motil. 2004;16(5):589-596.
      8. Lee HJ, Jung KW, Myung S-J. Technique of functional and motility test: how to perform biofeedback for constipation and fecal incontinence. 2013;19(4).
      9. Фоменко ОЮ, Титов АЮ, Бирюков ОМ, Мудров АА, Белоусова СВ. Характер функциональных нарушений мышц тазового дна у пациентов с обструктивной дефекацией. Колопроктология. 2016;(S 1):46-47.
      10. Титов АЮ, Бирюков ОМ, Фоменко ОЮ, Войнов МА. Метод биологической обратной связи в лечении проктогенных запоров у взрослых пациентов. Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2014;24(5):78-81.
      11. Фоменко ОЮ, Титов АЮ, Бирюков ОМ, Мудров АА, Белоусова СВ, Егорова ДВ. Диагностика и консервативное лечение функциональных расстройств дефекации. Колопроктология. 2016;(3 (57)):47-53.
      12. Rao SSC. Dyssynergic defecation and biofeedback therapy. Gastroenterol Clin North Am. 2008;37(3):569-86.
      13. Rao SSC, Welcher KD, Pelsang RE. Effects of biofeedback therapy on anorectal function in obstructive defecation. 1997;42(11).
      14. Lacy BE, Mearin F, Chang L, et al. Bowel Disorders. Gastroenterology. 2016;150(6):1393-1407.e5.
      15. Rao SSC, Benninga MA, Bharucha AE, Chiarioni G, Di Lorenzo C, Whitehead WE. ANMS-ESNM position paper and consensus guidelines on biofeedback therapy for anorectal disorders. Neurogastroenterol Motil. 2015;27(5):594-609.
      16. Tantiphlachiva K, Rao P, Attaluri A, Rao SSC. Digital Rectal Examination Is a Useful Tool for Identifying Patients With Dyssynergia. Clin Gastroenterol Hepatol. 2010;8(11):955-960.
      17. Minguez M, Herreros B, Sanchiz V, et al. Predictive value of the balloon expulsion test for excluding the diagnosis of pelvic floor dyssynergia in constipation. Gastroenterology. 2004;126(1):57-62.
      18. Ratuapli S, Bharucha AE, Harvey D, Zinsmeister AR. Comparison of rectal balloon expulsion test in seated and left lateral positions. Neurogastroenterol Motil. 2013;25(12): e813-20.
      19. Rao SS. Dyssynergic defecation. Gastroenterol Clin North Am. 2001;30(1):97-114.
      20. Rao SSC, Kavlock R, Rao S. Influence of Body Position and Stool Characteristics on Defecation in Humans. Am J Gastroenterol. 2006;101(12):2790-2796.
      21. Chiarioni G, Kim SM, Vantini I, Whitehead WE. Validation of the Balloon Evacuation Test: Reproducibility and Agreement With Findings From Anorectal Manometry and Electromyography. Clin Gastroenterol Hepatol. 2014;12(12):2049-2054.
      22. Ratuapli SK, Bharucha AE, Noelting J, et al. Phenotypic Identification and Classification of Functional Defecatory Disorders Using High-Resolution Anorectal Manometry. Gastroenterology. 2013;144(2):314-322.e2.
      23. Grossi U, Carrington E V, Bharucha AE, Horrocks EJ, Scott SM, Knowles CH. Diagnostic accuracy study of anorectal manometry for diagnosis of dyssynergic defecation. Gut. 2016;65(3):447-455.
      24. Diamant NE, Kamm MA, Wald A, et al. AGA technical review on anorectal testing techniques. Gastroenterology. 1999;116(3):735-760.
      25. Penninckx F, Debruyne C, Lestar B, Kerremans R. Observer variation in the radiological measurement of the anorectal angle. Int J Colorectal Dis. 1990;5(2):94-97.
      26. Ferrante SL, Perry RE, Schreiman JS, Cheng SC, Frick MP. The reproducibility of measuring the anorectal angle in defecography. Dis Colon Rectum. 1991;34(1):51-55.
      27. Shorvon PJ, McHugh S, Diamant NE, Somers S, Stevenson GW. Defecography in normal volunteers: results and implications. Gut. 1989;30(12):1737-1749.
      28. Rao SSC. Biofeedback therapy for constipation in adults. Best Pract Res Clin Gastroenterol. 2011;25(1):159-166.
      29. Pelsang RE, Rao SSC, Welcher K. FECOM: a new artificial stool for evaluating defecation. Am J Gastroenterol. 1999;94(1):183-186.
      30. Rao SSC, Valestin J, Brown CK, Zimmerman B, Schulze K. Long-term efficacy of biofeedback therapy for dyssynergic defecation: randomized controlled trial. Am J Gastroenterol. 2010;105(4):890-896.
      31. Rao SSC, Seaton K, Miller M, et al. Randomized Controlled Trial of Biofeedback, Sham Feedback, and Standard Therapy for Dyssynergic Defecation. Clin Gastroenterol Hepatol. 2007;5(3):331-338.
      32. Chiarioni G, Salandini L, Whitehead WE. Biofeedback benefits only patients with outlet dysfunction, not patients with isolated slow transit constipation. Gastroenterology. 2005;129(1):86-97.
      33. Heymen S, Scarlett Y, Jones K, Ringel Y, Drossman D, Whitehead WE. Randomized, Controlled Trial Shows Biofeedback to be Superior to Alternative Treatments for Patients with Pelvic Floor Dyssynergia-Type Constipation. Dis Colon Rectum. 2007;50(4):428-441.
      34. Chiarioni G, Whitehead WE, Pezza V, Morelli A, Bassotti G. Biofeedback Is Superior to Laxatives for Normal Transit Constipation Due to Pelvic Floor Dyssynergia. Gastroenterology. 2006;130(3):657-664.
      35. Koh CE, Young CJ, Young JM, Solomon MJ. Systematic review of randomized controlled trials of the effectiveness of biofeedback for pelvic floor dysfunction. Br J Surg. 2008;95(9):1079-1087.
      36. Shim LSE, Jones M, Prott GM, Morris LI, Kellow JE, Malcolm A. Predictors of outcome of anorectal biofeedback therapy in patients with constipation. Aliment Pharmacol Ther. 2011;33(11):1245-1251.
      37. Go J, Valestin J, Brown CK, et al. Is Biofeedback Therapy Effective in Improving Quality of Life in Dyssynergic Defecation? A Randomized Clinical Trial. Gastroenterology. 2011;140(5): S-52.
      38. Bharucha AE, Dorn SD, Lembo A, Pressman A, Pressman A. American Gastroenterological Association Medical Position Statement on Constipation. Gastroenterology. 2013;144(1):211-217.
      39. Ивашкин ВТ, Абдулхаков СР, Баранская ЕК, с соавт. Клинические рекомендации российской гастроэнтерологической ассоциации по диагностике и лечению взрослых пациентов с хроническим запором. Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2014;24(6):69-76.
      40. Kawimbe BM, Papachrysostomou M, Binnie NR, Clare N, Smith AN. Outlet obstruction constipation (anismus) managed by biofeedback. Gut. 1991;32(10):1175-1179.
      41. Patankar SK, Ferrara A, Levy JR, Larach SW, Williamson PR, Perozo SE. Biofeedback in colorectal practice: a multicenter, statewide, three-year experience. Dis Colon Rectum. 1997;40(7):827-831.
      42. Rao SS, Valestin J, Brown CK, et al. Home or Office Biofeedback Therapy for Dyssynergic Defecation - Randomized Controlled Trial. Gastroenterology. 2011;140(5): S-160.
     


    Full text is published :
    Golovenko A.O., Fomenko O.Yu., Egorova D.V., Belous S.S. BIOFEEDBACK THERAPY FOR TREATMENT OF CHRONIC CONSTIPATION. Experimental and Clinical Gastroenterology Journal. 2017;139(03):99-105
    Read & Download full text